Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Int J Cancer ; 134(4): 859-72, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23922012

RESUMEN

Therapeutic cancer vaccines show promise in preclinical studies, yet their clinical efficacy is limited. Increased recruitment of immune cells into tumors and suppression of the immune suppressive tumor environment are critical components toward effective cancer immunotherapies. Here, we report how local low-dose irradiation, alone or with a therapeutic immunization based on Semliki Forest virus (SFV) against human papillomavirus (HPV)-related cancer, influences these immune mechanisms. We first demonstrated that immunization with SFVeE6,7 or SFVeOVA, replicon particles expressing either HPV16 E6/E7 or ovalbumin, resulted in an antigen-specific migration of CD8+ T cells into HPV- and OVA-specific tumors. Local low-dose tumor irradiation alone resulted in a 2-fold increase of intratumoral CD8+ T cells. When 14 Gy irradiation was combined with immunization, intratumoral numbers of CD8+ T cells increased 10-fold and the number of CD8+ T cells specific for the E7- epitope increased more than 20-fold. Irradiation alone however also increased the number of intratumoral myeloid-derived suppressor cells (MDSCs) 3.5-fold. Importantly, this number did not further increase when combined with immunization. As a result, the ratio of antigen-specific CD8+ T cells and MDSCs in tumors increased up to 85-fold compared to the control. We furthermore demonstrated that following irradiation CCR2 and CCL2, CXCR6 and CCL16, chemokines and ligands involved in tumor homing of immune cells, were significantly up regulated. This study demonstrates that local low-dose tumor irradiation influences the intratumoral immune population induced by SFVeE6,7 immunization by a strong increase in the ratio of antitumoral to immune suppressive cells, thus changing the intratumoral immune balance in favor of antitumor activity.


Asunto(s)
Inmunoterapia , Neoplasias Experimentales/prevención & control , Papillomaviridae/inmunología , Infecciones por Papillomavirus/prevención & control , Virus de los Bosques Semliki/fisiología , Linfocitos T Citotóxicos/inmunología , Irradiación Corporal Total , Animales , Presentación de Antígeno , Western Blotting , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Quimiocinas/metabolismo , Terapia Combinada , Femenino , Citometría de Flujo , Humanos , Inmunización , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/virología , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/inmunología , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , ARN Mensajero/genética , Dosis de Radiación , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/inmunología , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
Int J Cancer ; 130(1): 105-12, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21328579

RESUMEN

Vaccine-induced p53-specific immune responses were previously reported to be associated with improved response to secondary chemotherapy in patients with small cell lung cancer. We investigated long-term clinical and immunological effects of the p53-synthetic long peptide (p53-SLP®) vaccine in patients with recurrent ovarian cancer. Twenty patients were immunized with the p53-SLP® vaccine between July 2006 and August 2007. Follow-up information on patients was obtained. Clinical responses to secondary chemotherapy after p53-SLP® immunizations were determined by computerized tomography and/or tumor marker levels (CA125). Disease-specific survival was compared to a matched historical control group. Immune responses were analyzed by flow cytometry, proliferation assay, interferon gamma (IFN-γ) ELISPOT and/or cytokine bead array. Lymphocytes cultured from skin biopsy were analyzed by flow cytometry and proliferation assay. Of 20 patients treated with the p53-SLP® vaccine, 17 were subsequently treated with chemotherapy. Eight of these patients volunteered another blood sample. No differences in clinical response rates to secondary chemotherapy or disease-specific survival were observed between immunized patients and historical controls (p = 0.925, resp. p = 0.601). p53-specific proliferative responses were observed in 5/8 patients and IFN-γ production in 2/7 patients. Lymphocytes cultured from a prior injection site showing inflammation during chemotherapy did not recognize p53-SLP®. Thus, treatment with the p53-SLP® vaccine does not affect responses to secondary chemotherapy or survival, although p53-specific T-cells do survive chemotherapy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Neoplasias Ováricas/terapia , Fragmentos de Péptidos/inmunología , Linfocitos T/inmunología , Proteína p53 Supresora de Tumor/inmunología , Adenocarcinoma de Células Claras/inmunología , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/terapia , Adenocarcinoma Mucinoso/inmunología , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/terapia , Antineoplásicos/uso terapéutico , Antígeno Ca-125/metabolismo , Proliferación Celular , Cistadenocarcinoma Seroso/inmunología , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/terapia , Citocinas , Neoplasias Endometriales/inmunología , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/terapia , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Inmunización , Interferón gamma , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Estudios Prospectivos
3.
Cochrane Database Syst Rev ; (1): CD007287, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20091627

RESUMEN

BACKGROUND: Despite advances in chemotherapy, prognosis of ovarian cancer remains poor. Antigen-specific active immunotherapy aims to induce a tumour-antigen-specific anti-tumour immune responses as an alternative treatment for ovarian cancer. OBJECTIVES: To assess feasibility of antigen-specific active immunotherapy for ovarian cancer. Primary outcomes are clinical efficacy and antigen-specific immunogenicity with carrier-specific immunogenicity and side-effects as secondary outcomes. SEARCH STRATEGY: A systematic search of the Cochrane Central Register of Controlled Trials (CENTRAL) Issue 3, 2009, Cochrane Gynaecological Cancer Group Specialized Register, MEDLINE and EMBASE databases and clinicaltrials.gov was performed (1966 to July 2009). Hand searches were conducted of the proceedings of relevant annual meetings (1996 to July 2009). SELECTION CRITERIA: Randomised controlled trials (RCTs), as well as non-randomised non-controlled studies that included patients with epithelial ovarian cancer, irrespective of stage of disease, and treated with antigen-specific active immunotherapy, irrespective of type of vaccine, antigen used, adjuvant used, route of vaccination, schedule, and reported clinical or immunological outcomes. DATA COLLECTION AND ANALYSIS: Data extraction was performed independently by two review authors. Risk of bias was evaluated with the Delphi-list for RCTs or a selection of quality domains pivotal to the assessment of non-RCTs and deemed best applicable to the non-randomised non-controlled studies. MAIN RESULTS: Thirty-six studies were included. Response definitions showed substantial variation between trials, which makes comparison of trial results unreliable. Information on adverse events was frequently limited. Furthermore, reports of both RCTs and non-RCTs frequently lacked information necessary to assess risk of bias. Serious biases in these trials can thus not be ruled out.The largest body of evidence is currently available for CA-125 targeted antibody therapy (15 studies: 1505 patients). Non-RCTs of this CA-125 targeted antibody therapy suggest increased survival in humoral and/or cellular responders. However, three large randomised placebo-controlled trials did not show any clinical benefit despite induction of immune responses in approximately 60% of patients.Other small studies targeting many different tumour antigens showed promising immunological results. As these strategies have not yet been tested in RCTs, no reliable inferences about clinical efficacy can be made. Given the promising immunological results, limited side effects and toxicity exploration of clinical efficacy in large well-designed RCTs may be worthwhile. AUTHORS' CONCLUSIONS: We conclude that despite promising immunological responses no clinically effective antigen-specific active immunotherapy is yet available for ovarian cancer. Furthermore, the adoption of guidelines to ensure uniformity in trial conduct, response definitions and trial reporting is recommended to improve quality and comparability of immunotherapy trials.


Asunto(s)
Inmunoterapia Activa/métodos , Neoplasias Ováricas/terapia , Anticuerpos Monoclonales/uso terapéutico , Antígeno Ca-125/inmunología , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Femenino , Humanos , Neoplasias Ováricas/inmunología , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Expert Rev Vaccines ; 10(6): 775-84, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21692699

RESUMEN

Ovarian cancer vaccines are one of the new treatment strategies under investigation in epithelial ovarian cancer. This article discusses the results of different immunization strategies, points out potential pitfalls in study designs and provides possible solutions for augmentation of clinical efficacy. Most ovarian cancer vaccines have not yet evolved beyond Phase I/II studies, which do not primarily evaluate clinical efficacy. Although different approaches of antigen-specific immunization generally result in antigen-specific immune responses, clinical benefit is not consistently observed. Based on the currently available results, we emphasize the necessity of multimodal treatment of ovarian cancer, combining classical cytoreductive surgery, (neo)adjuvant chemotherapy, immunotherapy and/or targeted therapy.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Ensayos Clínicos como Asunto , Femenino , Humanos , Resultado del Tratamiento
5.
Int J Cancer ; 121(3): 606-14, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17415711

RESUMEN

Despite intensive treatment, 70% of the ovarian cancer patients will develop recurrent disease, emphasizing the need for new approaches such as immunotherapy. A promising antigenic target for immunotherapy in ovarian cancer is the frequently overexpressed p53 protein. The aim of the study was to evaluate the nature and magnitude of the baseline anti-p53 immune response in ovarian cancer patients. P53-specific T cell responses were detected in both half of the ovarian cancer patients as in the group of control subjects, consisting of women with benign ovarian tumors and healthy controls. Importantly, while in the control group p53-specific immunity was detected among the CD45RA(+) naïve subset of T cells only, the p53-specific T-cell responses in ovarian cancer patients were also present in the CD45RO(+) memory T-cell subset, suggesting that in the cancer patients sufficient amounts of cancer-derived p53 was presented to induce the formation of a p53-specific memory T-cell response. Further characterization of the p53-specific memory T-cell responses revealed that in addition to the type 1 cytokine IFN-gamma also the type 2 cytokines IL-4 and IL-5, as well as the immunosuppressive cytokine IL-10 were produced. Notably, p53-specific T cells were not only detected in the peripheral blood, but also among tumor infiltrating lymphocytes and in tumor-draining lymph nodes. In conclusion, the existence of a weak mixed T-helper type 1 and 2 p53-specific T-cell repertoire supports the rationale of using p53 long peptides in vaccination strategies aiming at the induction of p53-specific Th1/CTL immunity.


Asunto(s)
Neoplasias Ováricas/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Citocinas/metabolismo , Femenino , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Antígenos Comunes de Leucocito , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Persona de Mediana Edad , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína p53 Supresora de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA