Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
BMC Cancer ; 14: 290, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24766732

RESUMO

BACKGROUND: The efficacy and safety of axitinib, a potent and selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, and 3 in combination with pemetrexed and cisplatin was evaluated in patients with advanced non-squamous non-small-cell lung cancer (NSCLC). METHODS: Overall, 170 patients were randomly assigned to receive axitinib at a starting dose of 5-mg twice daily continuously plus pemetrexed 500 mg/m(2) and cisplatin 75 mg/m(2) on day 1 of up to six 21-day cycles (arm I); axitinib on days 2 through 19 of each cycle plus pemetrexed/cisplatin (arm II); or pemetrexed/cisplatin alone (arm III). The primary endpoint was progression-free survival (PFS). RESULTS: Median PFS was 8.0, 7.9, and 7.1 months in arms I, II, and III, respectively (hazard ratio: arms I vs. III, 0.89 [P = 0.36] and arms II vs. III, 1.02 [P = 0.54]). Median overall survival was 17.0 months (arm I), 14.7 months (arm II), and 15.9 months (arm III). Objective response rates (ORRs) for axitinib-containing arms were 45.5% (arm I) and 39.7% (arm II) compared with 26.3% for pemetrexed/cisplatin alone (arm III). Gastrointestinal disorders and fatigue were frequently reported across all treatment arms. The most common all-causality grade ≥3 adverse events were hypertension in axitinib-containing arms (20% and 17%, arms I and II, respectively) and fatigue with pemetrexed/cisplatin alone (16%). CONCLUSION: Axitinib in combination with pemetrexed/cisplatin was generally well tolerated. Axitinib combinations resulted in non-significant differences in PFS and numerically higher ORR compared with chemotherapy alone in advanced NSCLC. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00768755 (October 7, 2008).


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/administração & dosagem , Glutamatos/administração & dosagem , Guanina/análogos & derivados , Imidazóis/administração & dosagem , Indazóis/administração & dosagem , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica , Axitinibe , Carcinoma Pulmonar de Células não Pequenas/patologia , Intervalo Livre de Doença , Feminino , Guanina/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Pemetrexede , Fator A de Crescimento do Endotélio Vascular/genética
2.
BMC Cancer ; 12: 361, 2012 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-22906006

RESUMO

BACKGROUND: The investigational oral DNA vaccine VXM01 targets the vascular endothelial growth factor receptor 2 (VEGFR-2) and uses Salmonella typhi Ty21a as a vector. The immune reaction elicited by VXM01 is expected to disrupt the tumor neovasculature and, consequently, inhibit tumor growth. VXM01 potentially combines the advantages of anti-angiogenic therapy and active immunotherapy. METHODS/DESIGN: This phase I trial examines the safety, tolerability, and immunological and clinical responses to VXM01. The randomized, placebo-controlled, double blind dose-escalation study includes up to 45 patients with locally advanced and stage IV pancreatic cancer. The patients will receive four doses of VXM01 or placebo in addition to gemcitabine as standard of care. Doses from 106 cfu up to 1010 cfu of VXM01 will be evaluated in the study. An independent data safety monitoring board (DSMB) will be involved in the dose-escalation decisions. In addition to safety as primary endpoint, the VXM01-specific immune reaction, as well as clinical response parameters will be evaluated. DISCUSSION: The results of this study shall provide the first data regarding the safety and immunogenicity of the oral anti-VEGFR-2 vaccine VXM01 in cancer patients. They will also define the recommended dose for phase II and provide the basis for further clinical evaluation, which may also include additional cancer indications. TRIAL REGISTRATION: EudraCT No.: 2011-000222-29, NCT01486329, ISRCTN68809279.


Assuntos
Vacinas Anticâncer/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/imunologia , Vacinas de DNA/administração & dosagem , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Administração Oral , Adulto , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Ensaios Clínicos Fase I como Assunto/métodos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Método Duplo-Cego , Humanos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/imunologia , Neovascularização Patológica/prevenção & controle , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/prevenção & controle , Placebos , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Salmonella typhi/genética , Vacinas de DNA/efeitos adversos , Vacinas de DNA/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Gencitabina
3.
Immunol Rev ; 222: 117-28, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18363997

RESUMO

Four novel oral DNA vaccines provide protection against melanoma, colon, breast, and lung carcinoma in mouse models. Vaccines are delivered by attenuated Salmonella typhimurium to secondary lymphoid organs and respectively target vascular endothelial growth factor receptor-2, transcription factor Fos-related antigen-1, anti-apoptosis protein survivin and Legumain, an asparaginyl endopeptidase specifically overexpressed on tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). These vaccines are all capable of inducing potent cell-mediated protective immunity against self-antigens, resulting in marked suppression of tumor growth and dissemination. Key mechanisms induced by these DNA vaccines include efficient suppression of angiogenesis in the tumor vasculature and marked activation of cytotoxic T cells, natural killer cells, and antigen-presenting dendritic cells. The vaccine targeting Legumain establishes the new paradigm whereby a reduction in the density of TAMs in the TME decreases the release of factors potentiating tumor growth and angiogenesis. This, in turn, remodels the TME and decreases its immunosuppressive milieu and thereby potentiates the DNA vaccine's ability to effectively suppress tumor cell proliferation, vascularization, and metastasis. It is anticipated that such research efforts will lead to novel DNA-based vaccines that will be effective for the treatment of cancer.


Assuntos
Imunoterapia , Neoplasias Experimentais/terapia , Neovascularização Patológica/prevenção & controle , Vacinas de DNA/administração & dosagem , Administração Oral , Animais , Linfócitos T CD8-Positivos/imunologia , Quimiocina CCL21/imunologia , Técnicas de Transferência de Genes , Memória Imunológica , Proteínas Inibidoras de Apoptose , Interleucina-18/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Camundongos , Proteínas Associadas aos Microtúbulos/imunologia , Invasividade Neoplásica , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/imunologia , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Proteínas Proto-Oncogênicas c-fos/imunologia , Proteínas Repressoras , Salmonella typhimurium , Tolerância a Antígenos Próprios/imunologia , Survivina , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
4.
Clin Pharmacol Drug Dev ; 11(8): 997-1006, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35509219

RESUMO

Preclinical studies in animal models of obesity and inflammation have shown that oral administration of ARD-101, a potential TAS2R agonist, reduced food intake and body weight and downregulated inflammatory cytokines. We present results from a first-in-human phase 1 randomized, placebo-controlled trial that evaluated safety, pharmacokinetics, and pharmacodynamics of single or multiple ascending doses of oral ARD-101 (40, 100, and 240 mg) in healthy adults. A total of 43 subjects were randomly assigned and dosed to ARD-101 or placebo with 42 subjects completing the study treatment. ARD-101 was found to be >99% restricted to the gut with minimal systemic exposure, demonstrated a favorable safety profile, and was well tolerated at all dose levels. Blood samples taken 1 hour after administration showed that subjects dosed with 240 mg of ARD-101 had elevated circulating levels of several gut peptide hormones. It is postulated that ARD-101 activates enteroendocrine cells to achieve its effects regulating metabolism and inflammation. The phase 1 clinical results demonstrated safety of ARD-101 and indicated activation of gut peptide hormone release in healthy adults. Further clinical trials will evaluate ARD-101 in patients with metabolic and inflammatory disorders.


Assuntos
Acetatos , Paladar , Adulto , Animais , Relação Dose-Resposta a Droga , Humanos , Inflamação , Compostos de Amônio Quaternário
5.
Nat Med ; 8(12): 1369-75, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12415261

RESUMO

Tumor cells are elusive targets for immunotherapy due to their heterogeneity and genetic instability. Here we describe a novel, oral DNA vaccine that targets stable, proliferating endothelial cells in the tumor vasculature rather than tumor cells. Targeting occurs through upregulated vascular-endothelial growth factor receptor 2 (FLK-1) of proliferating endothelial cells in the tumor vasculature. This vaccine effectively protected mice from lethal challenges with melanoma, colon carcinoma and lung carcinoma cells and reduced growth of established metastases in a therapeutic setting. CTL-mediated killing of endothelial cells indicated breaking of peripheral immune tolerance against this self antigen, resulting in markedly reduced dissemination of spontaneous and experimental pulmonary metastases. Angiogenesis in the tumor vasculature was suppressed without impairment of fertility, neuromuscular performance or hematopoiesis, albeit with a slight delay in wound healing. Our strategy circumvents problems in targeting of genetically unstable tumor cells. This approach may provide a new strategy for the rational design of cancer therapies.


Assuntos
Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Vacinas de DNA/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Citotoxicidade Imunológica , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos C57BL , Vacinação , Cicatrização
6.
J Clin Invest ; 116(7): 1955-62, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16794736

RESUMO

Tumor-associated fibroblasts are key regulators of tumorigenesis. In contrast to tumor cells, which are genetically unstable and mutate frequently, the presence of genetically more stable fibroblasts in the tumor-stromal compartment makes them an optimal target for cancer immunotherapy. These cells are also the primary source of collagen type I, which contributes to decreased chemotherapeutic drug uptake in tumors and plays a significant role in regulating tumor sensitivity to a variety of chemotherapies. To specifically kill tumor-associated fibroblasts, we constructed an oral DNA vaccine targeting fibroblast activation protein (FAP), which is specifically overexpressed by fibroblasts in the tumor stroma. Through CD8+ T cell-mediated killing of tumor-associated fibroblasts, our vaccine successfully suppressed primary tumor cell growth and metastasis of multidrug-resistant murine colon and breast carcinoma. Furthermore, tumor tissue of FAP-vaccinated mice revealed markedly decreased collagen type I expression and up to 70% greater uptake of chemotherapeutic drugs. Most importantly, pFap-vaccinated mice treated with chemotherapy showed a 3-fold prolongation in lifespan and marked suppression of tumor growth, with 50% of the animals completely rejecting a tumor cell challenge. This strategy opens a new venue for the combination of immuno- and chemotherapies.


Assuntos
Vacinas Anticâncer , Fibroblastos/metabolismo , Gelatinases/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias , Serina Endopeptidases/metabolismo , Vacinas de DNA , Animais , Linfócitos T CD8-Positivos/imunologia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Endopeptidases , Feminino , Gelatinases/genética , Gelatinases/uso terapêutico , Proteínas de Membrana/genética , Proteínas de Membrana/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Serina Endopeptidases/genética , Serina Endopeptidases/uso terapêutico , Taxa de Sobrevida
7.
Arterioscler Thromb Vasc Biol ; 27(9): 2050-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17600223

RESUMO

OBJECTIVE: Vascular endothelial growth factor receptor 2 (VEGFR2)-overexpressing cells may form an interesting target for the treatment of atherosclerosis because of their involvement in processes that contribute to this disease, such as angiogenesis. METHODS AND RESULTS: We vaccinated mice against VEGFR2 by an orally administered DNA vaccine, comprising a plasmid, encoding murine VEGFR2, carried by live attenuated Salmonella typhimurium. This vaccine induces cellular immunity against cells that overexpress VEGFR2. Vaccination of hypercholesterolemic mice against VEGFR2 resulted in a marked induction of CD8+ cytotoxic T cells specific for VEGFR2 and led to an inhibition of angiogenesis in a hindlimb ischemia model. Interestingly, VEGFR2 vaccination attenuated the progression of preexisting advanced atherosclerotic lesions in the brachiocephalic artery of apoE-/- mice. Furthermore, VEGFR2 vaccination strongly reduced the initiation of collar-induced atherosclerosis in the carotid arteries of LDLr-/- mice. In addition, denudation of the carotid artery, as a model for postinterventional lesion formation, resulted in delayed endothelial replacement and significantly increased neointima formation on VEGFR2 vaccination. CONCLUSIONS: These data indicate the prominent role of VEGFR2+ cells in cardiovascular diseases and show that induction of cellular immunity against atherosclerosis-associated cells by means of DNA vaccination may contribute to the development of novel therapies against atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Hipercolesterolemia/terapia , Imunoterapia Ativa/métodos , Neovascularização Patológica/prevenção & controle , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Administração Oral , Animais , Aterosclerose/patologia , Linfócitos T CD8-Positivos/imunologia , Técnicas de Cocultura , Células Endoteliais/imunologia , Células Endoteliais/patologia , Feminino , Membro Posterior , Histocitoquímica , Imunidade Celular/imunologia , Isquemia/terapia , Camundongos , Vacinas de DNA/administração & dosagem
8.
J Clin Invest ; 110(2): 185-92, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12122110

RESUMO

Development of tumor immunotherapies focuses on inducing autoimmune responses against tumor-associated self-antigens primarily encoded by normal, unmutated genes. We hypothesized that such responses could be elicited by T cell homeostatic proliferation in the periphery, involving expansion of T cells recognizing self-MHC/peptide ligands. Herein, we demonstrate that sublethally irradiated lymphopenic mice transfused with autologous or syngeneic T cells showed tumor growth inhibition when challenged with melanoma or colon carcinoma cells. Importantly, the antitumor response depended on homeostatic expansion of a polyclonal T cell population within lymph nodes. This response was effective even for established tumors, was characterized by CD8(+) T cell-mediated tumor-specific cytotoxicity and IFN-gamma production, and was associated with long-term memory. The results indicate that concomitant induction of the physiologic processes of homeostatic T cell proliferation and tumor antigen presentation in lymph nodes triggers a beneficial antitumor autoimmune response.


Assuntos
Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Linfócitos T/imunologia , Animais , Apresentação de Antígeno , Antígenos de Neoplasias , Autoantígenos , Autoimunidade , Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Citotoxicidade Imunológica , Homeostase , Memória Imunológica , Imunoterapia , Interferon gama/biossíntese , Ativação Linfocitária , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/transplante , Vitiligo/imunologia , Vitiligo/patologia
9.
Int J Radiat Oncol Biol Phys ; 64(1): 235-41, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16257132

RESUMO

PURPOSE: Intraoperative electron-beam radiotherapy (IOERT) has been applied for local dose escalation in over 1,400 patients in Heidelberg since 1991. Among these were 30 children, in 18 of whom IOERT was employed in radiation treatment with external-beam radiotherapy (EBRT) on account of incomplete resection. We address the question whether IOERT is able to compensate for microscopic or macroscopic tumor residue if employed in the overall radiation regimen. METHODS AND MATERIALS: The data of the aforementioned 18 children were analyzed with regard to local recurrence, overall survival, and complication rates. All children suffered from either sarcomas or neuroblastomas. In all children, IOERT was employed for local dose escalation after or before EBRT. RESULTS: After a median follow-up of 60.5 months, 15 of the treated children are alive. One local failure has been observed. Six children show clinically significant late morbidity, including the loss of a treated limb (Radiation Therapy Oncology Group Grade 4 [RTOG 4]), a severe nerve lesion (RTOG 3), an orthopedic complication (RTOG 2), a ureteral stenosis (not clinically significant), and a kidney hypotrophy (not clinically significant). In 1 child a fracture due to radionecrosis (RTOG 4) was diagnosed; however, in the follow-up, local tumor relapse was diagnosed as another possible reason for the fracture. CONCLUSIONS: Regarding the low incidence of local failure, IOERT seems to be able to compensate incomplete tumor resection in childhood sarcoma and neuroblastoma patients. The incidence of late morbidity is low enough to justify the employment of IOERT as part of the radiation treatment regimen for pediatric patients.


Assuntos
Neoplasias Ósseas/radioterapia , Neuroblastoma/radioterapia , Neoplasias Retroperitoneais/radioterapia , Sarcoma/radioterapia , Adolescente , Criança , Pré-Escolar , Terapia Combinada/métodos , Feminino , Seguimentos , Humanos , Lactente , Período Intraoperatório , Masculino , Neoplasia Residual , Neuroblastoma/cirurgia , Neoplasias Retroperitoneais/cirurgia , Espaço Retroperitoneal , Sarcoma/cirurgia
10.
Int J Radiat Oncol Biol Phys ; 64(5): 1416-23, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16413697

RESUMO

PURPOSE: To analyze long-term prognosis and morbidity after limb-sparing treatment of patients with extremity soft-tissue sarcoma, with intraoperative electron boost radiotherapy (IOERT) followed by a moderate dose of external beam radiotherapy (EBRT). METHODS AND MATERIALS: A total of 153 patients who were treated in a single center from 1991 to 2004 were evaluated. Median IOERT dose was 15 Gy, mean EBRT dose 43 Gy (range, 40-50.4 Gy) in conventional fractionation (1.8-2 Gy). Median duration of follow-up was 33 months. Acute toxicity was assessed with Common Toxicity Criteria; late toxic effects were scored according to European Organization for Research and Treatment of Cancer/Radiation Therapy Oncology Group criteria. RESULTS: Five-year overall survival and 5-year local control rates were 77% and 78%, respectively. Whereas tumor size, patient age, and EBRT dose did not significantly affect outcome, resection status and grading were significant for survival; resection status and IOERT dose were significant for local control. Extremity salvage until death or time of follow-up was achieved in 90% of our patients, 86% of whom showed excellent limb function without impairment in activities of daily life. Acute toxicity Grade 2-4 was observed in 23% and late toxicity Grade 2-4 in 17% of patients. CONCLUSIONS: Treatment with IOERT combined with moderate doses of external beam irradiation yields high local control and extremity preservation rates in resected extremity soft-tissue sarcoma.


Assuntos
Extremidades , Salvamento de Membro/métodos , Sarcoma/radioterapia , Sarcoma/cirurgia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Elétrons/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/cirurgia , Radioterapia/métodos , Sarcoma/mortalidade , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA