Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Clin Cancer Res ; 28(11): 2440-2448, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35294545

RESUMEN

PURPOSE: Despite recent advances in the molecular characterization of gliomas, it remains unclear which patients benefit most from which second-line treatments. The TAVAREC trial was a randomized, open-label phase II trial assessing the benefit of the addition of the angiogenesis inhibitor bevacizumab to treatment with temozolomide in patients with a first enhancing recurrence of World Health Organization grade 2 or 3 glioma without 1p/19q codeletion. We evaluated the prognostic significance of genome-wide DNA methylation profiles and copy-number variations on the TAVAREC trial samples. EXPERIMENTAL DESIGN: Isocitrate dehydrogenase (IDH) mutation status was determined via Sanger sequencing and IHC. DNA methylation analysis was performed using the MethylationEPIC BeadChip (Illumina) from which 1p/19q codeletion, MGMT promoter methylation (MGMT-STP27), and homozygous deletion of CDKN2A/B were determined. DNA methylation classes were determined according to classifiers developed in Heidelberg and The Cancer Genome Atlas (TCGA; "Heidelberg" and "TCGA" classifier respectively). RESULTS: DNA methylation profiles of 122 samples were successfully determined. As expected, most samples were IDH-mutant (89/122) and MGMT promotor methylated (89/122). Methylation classes were prognostic for time to progression. However, Heidelberg methylation classes determined at time of diagnosis were no longer prognostic following enhancing recurrence of the tumor. In contrast, TCGA methylation classes of primary samples remained prognostic also following enhancing recurrence. Homozygous deletions in CDKN2A/B were found in 10 of 87 IDH-mutated samples and were prognostically unfavorable at recurrence. CONCLUSIONS: DNA methylome Heidelberg classification at time of diagnosis is no longer of prognostic value at the time of enhancing recurrence. CDKN2A/B deletion status was predictive of survival from progression of IDH-mutated tumors.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/patología , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Glioma/patología , Homocigoto , Humanos , Isocitrato Deshidrogenasa/genética , Imagen por Resonancia Magnética , Mutación , Recurrencia Local de Neoplasia/genética , Pronóstico , Eliminación de Secuencia
2.
Int J Gynecol Cancer ; 29(9): 1381-1388, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31685557

RESUMEN

BACKGROUND: The behavior of the immune system as a driver in the progression of ovarian cancer has barely been studied. Our knowledge is mainly limited to the intra-tumoral adaptive immune system. Because of the widespread metastases of ovarian cancer, an assessment of the circulating immune system seems more accurate.To demonstrate the presence of immune cells in blood samples of patients with ovarian neoplasms. METHODS: In this exploratory prospective cohort study, peripheral blood mononuclear cells were collected at diagnosis from 143 women, including 62 patients with benign cysts, 13 with borderline tumor, 41 with invasive ovarian cancer, and 27 age-matched healthy controls. Immune profile analyses, based on the presence of CD4 (cluster of differentiation), CD8, natural killer cells, myeloid-derived suppressor cells, and regulatory T cells, were performed by fluorescence activated cell sorting. RESULTS: In a multivariable analysis, six immune cells (activated regulatory T cells, natural killer cells, myeloid-derived suppressor cells, monocytic myeloid-derived suppressor cells, exhausted monocytic myeloid-derived suppressor cells, and total myeloid cells) were selected as independent predictors of malignancy, with an optimism-corrected area under the receiver operating characteristic curve (AUC) of 0.858. In contrast, a profile based on CD8 and regulatory T cells, the current standard in ovarian cancer immunology, resulted in an AUC of 0.639. CONCLUSIONS: Our immune profile in blood suggests an involvement of innate immunosuppression driven by myeloid-derived suppressor cells in the development of ovarian cancer. This finding could contribute to clinical management of patients and in selection of immunotherapy.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Inmunidad Adaptativa , Adulto , Estudios de Casos y Controles , Estudios de Cohortes , Diagnóstico Diferencial , Femenino , Humanos , Inmunidad Innata , Neoplasias Ováricas/sangre , Neoplasias Ováricas/diagnóstico , Pronóstico , Estudios Prospectivos
3.
Lancet Oncol ; 19(9): 1170-1179, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30115593

RESUMEN

BACKGROUND: Bevacizumab is frequently used in the treatment of recurrent WHO grade II and III glioma, but without supporting evidence from randomised trials. Therefore, we assessed the use of bevacizumab in patients with first recurrence of grade II or III glioma who did not have 1p/19q co-deletion. METHODS: The TAVAREC trial was a randomised, open-label phase 2 trial done at 32 centres across Europe in patients with locally diagnosed grade II or III glioma without 1p/19q co-deletion, with a first and contrast-enhancing recurrence after initial radiotherapy or chemotherapy, or both. Previous chemotherapy must have been stopped at least 6 months before enrolment and radiotherapy must have been stopped at least 3 months before enrolment. Random group assignment was done electronically through the European Organisation for Research and Treatment of Cancer web-based system, stratified by a minimisation procedure using institution, initial histology (WHO grade II vs III), WHO performance status (0 or 1 vs 2), and previous treatment (radiotherapy, chemotherapy, or both). Patients were assigned to receive either temozolomide (150-200 mg/m2, orally) monotherapy on days 1-5 every 4 weeks for a maximum of 12 cycles, or the same temozolomide regimen in combination with bevacizumab (10 mg/kg, intravenously) every 2 weeks until progression. The primary endpoint was overall survival at 12 months in the per-protocol population. Safety analyses were done in all patients who started their allocated treatment. The study is registered at EudraCT (2009-017422-39) and ClinicalTrials.gov (NCT01164189), and is complete. FINDINGS: Between Feb 8, 2011, and July 31, 2015, 155 patients were enrolled and randomly assigned to receive either monotherapy (n=77) or combination therapy (n=78). Overall survival in the per-protocol population at 12 months was achieved by 44 (61% [80% CI 53-69]) of 72 patients in the temozolomide group and 38 (55% [47-69]) of 69 in the combination group. The most frequent toxicity was haematological: 17 (23%) of 75 patients in the monotherapy group and 25 (33%) of 76 in the combination group developed grade 3 or 4 haematological toxicity. Other than haematological toxicities, the most common adverse events were nervous system disorders (59 [79%] of 75 patients in the monotherapy group vs 65 [86%] of 76 in the combination group), fatigue (53 [70%] vs 61 [80%]), and nausea (39 [52%] vs 43 [56%]). Infections were more frequently reported in the combination group (29 [38%] of 76 patients) than in the monotherapy group (17 [23%] of 75). One treatment-related death was reported in the combination group (infection after intratumoral haemorrhage during a treatment-related grade 4 thrombocytopenia). INTERPRETATION: We found no evidence of improved overall survival with bevacizumab and temozolomide combination treatment versus temozolomide monotherapy. The findings from this study provide no support for further phase 3 studies on the role of bevacizumab in this disease. FUNDING: Roche Pharmaceuticals.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bevacizumab/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Recurrencia Local de Neoplasia , Temozolomida/administración & dosificación , Adulto , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Inmunológicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bevacizumab/efectos adversos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Deleción Cromosómica , Cromosomas Humanos Par 1 , Cromosomas Humanos Par 19 , Esquema de Medicación , Europa (Continente) , Femenino , Glioma/genética , Glioma/mortalidad , Glioma/patología , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Temozolomida/efectos adversos , Factores de Tiempo , Resultado del Tratamiento
4.
EMBO Mol Med ; 9(12): 1629-1645, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29038312

RESUMEN

Glioma growth and progression are characterized by abundant development of blood vessels that are highly aberrant and poorly functional, with detrimental consequences for drug delivery efficacy. The mechanisms driving this vessel dysmorphia during tumor progression are poorly understood. Using longitudinal intravital imaging in a mouse glioma model, we identify that dynamic sprouting and functional morphogenesis of a highly branched vessel network characterize the initial tumor growth, dramatically changing to vessel expansion, leakage, and loss of branching complexity in the later stages. This vascular phenotype transition was accompanied by recruitment of predominantly pro-inflammatory M1-like macrophages in the early stages, followed by in situ repolarization to M2-like macrophages, which produced VEGF-A and relocate to perivascular areas. A similar enrichment and perivascular accumulation of M2 versus M1 macrophages correlated with vessel dilation and malignancy in human glioma samples of different WHO malignancy grade. Targeting macrophages using anti-CSF1 treatment restored normal blood vessel patterning and function. Combination treatment with chemotherapy showed survival benefit, suggesting that targeting macrophages as the key driver of blood vessel dysmorphia in glioma progression presents opportunities to improve efficacy of chemotherapeutic agents. We propose that vessel dysfunction is not simply a general feature of tumor vessel formation, but rather an emergent property resulting from a dynamic and functional reorganization of the tumor stroma and its angiogenic influences.


Asunto(s)
Vasos Sanguíneos/patología , Neoplasias Encefálicas/patología , Glioma/patología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Vasos Sanguíneos/anomalías , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Glioma/irrigación sanguínea , Glioma/tratamiento farmacológico , Glioma/mortalidad , Humanos , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/patología , Fenotipo , Proteínas Proto-Oncogénicas c-sis/genética , Temozolomida , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Int J Cancer ; 141(9): 1891-1900, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28681455

RESUMEN

Blockade of the immune checkpoint molecule programmed-cell-death-protein-1 (PD-1) yielded promising results in several cancers. To understand the therapeutic potential in human gliomas, quantitative data describing the expression of PD-1 are essential. Moreover, due the immune-specialized region of the brain in which gliomas arise, differences between tumor-infiltrating and circulating lymphocytes should be acknowledged. In this study we have used flow cytometry to quantify PD-1 expression on tumor-infiltrating T cells of 25 freshly resected glioma cell suspensions (10 newly and 5 relapsed glioblastoma, 10 lower grade gliomas) and simultaneously isolated circulating T cells. A strong upregulation of PD-1 expression in the tumor microenvironment compared to the blood circulation was seen in all glioma patients. Additionally, circulating T cells were isolated from 15 age-matched healthy volunteers, but no differences in PD-1 expression were found compared to glioma patients. In the murine GL261 malignant glioma model, there was a similar upregulation of PD-1 on brain-infiltrating lymphocytes. Using a monoclonal PD-1 blocking antibody, we found a marked prolonged survival with 55% of mice reaching long-term survival. Analysis of brain-infiltrating cells 21 days after GL261 tumor implantation showed a shift in infiltrating lymphocyte subgroups with increased CD8+ T cells and decreased regulatory T cells. Together, our results suggest an important role of PD-1 in glioma-induced immune escape, and provide translational evidence for the use of PD-1 blocking antibodies in human malignant gliomas.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Glioma/tratamiento farmacológico , Linfocitos Infiltrantes de Tumor/patología , Receptor de Muerte Celular Programada 1/genética , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/inmunología , Glioma/patología , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Oncoimmunology ; 5(4): e1111505, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27141394

RESUMEN

Neoplastic cells can escape immune control leading to cancer growth. Regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) are crucial in immune escape. TAM are divided based on their immune profile, M1 are immunostimulatory while M2 are immunosuppressive. Research so far has mainly focused on the intratumoral behavior of these cells. This study, on the other hand, explored the systemic changes of the key metabolites [IL-4 (interleukin), IL-13, arginase, IL-10, VEGF-A (vascular endothelial growth factor), CCL-2 (chemokine (C-C) motif ligand 2) and TGF-ß (transforming growth factor)] linked to Treg, MDSC and TAM during the course of the disease in ovarian and fallopian tube cancer patients. Serum samples were therefore analyzed at diagnosis, after (interval)-debulking surgery and after chemotherapy (paclitaxel-carboplatin). We also determined galectin-1 (gal-1), involved in angiogenesis and tumor-mediated immune evasion. We found significantly lower levels of IL-10, VEGF-A, TGF-ß and arginase and higher levels of gal-1 after chemotherapy compared to diagnosis. After debulking surgery, a decrease in IL-10 was significant. Gal-1 and CCL-2 appeared independent prognostic factors for progression-free and overall survival (OS) (multivariate analysis). These results will help us in the decision making of future therapies in order to further modulate the immune system in a positive way.

7.
Oncoimmunology ; 5(2): e1083669, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27057467

RESUMEN

Dendritic cell (DC)-based immunotherapy has yielded promising results against high-grade glioma (HGG). However, the efficacy of DC vaccines is abated by HGG-induced immunosuppression and lack of attention toward the immunogenicity of the tumor lysate/cells used for pulsing DCs. A literature analysis of DC vaccination clinical trials in HGG patients delineated the following two most predominantly applied methods for tumor lysate preparation: freeze-thaw (FT)-induced necrosis or FT-necrosis followed by X-ray irradiation. However, from the available clinical evidence, it is unclear which of both methodologies has superior immunogenic potential. Using an orthotopic HGG murine model (GL261-C57BL/6), we observed that prophylactic vaccination with DCs pulsed with irradiated FT-necrotic cells (compared to FT-necrotic cells only) prolonged overall survival by increasing tumor rejection in glioma-challenged mice. This was associated, both in prophylactic and curative vaccination setups, with an increase in brain-infiltrating Th1 cells and cytotoxic T lymphocytes (CTL), paralleled by a reduced accumulation of regulatory T cells, tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC). Further analysis showed that irradiation treatment of FT-necrotic cells considerably increased the levels of carbonylated proteins - a surrogate-marker of oxidation-associated molecular patterns (OAMPs). Through further application of antioxidants and hydrogen peroxide, we found a striking correlation between the amount of lysate-associated protein carbonylation/OAMPs and DC vaccine-mediated tumor rejection capacity thereby suggesting for the first time a role for protein carbonylation/OAMPs in at least partially mediating antitumor immunity. Together, these data strongly advocate the use of protein oxidation-inducing modalities like irradiation for increasing the immunogenicity of tumor lysate/cells used for pulsing DC vaccines.

8.
Sci Transl Med ; 8(328): 328ra27, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26936504

RESUMEN

The promise of dendritic cell (DC)-based immunotherapy has been established by two decades of translational research. Of the four malignancies most targeted with clinical DC immunotherapy, high-grade glioma (HGG) has shown the highest susceptibility. HGG-induced immunosuppression is a roadblock to immunotherapy, but may be overcome by the application of T helper 1 (T(H)1) immunity-biased, next-generation, DC immunotherapy. To this end, we combined DC immunotherapy with immunogenic cell death (ICD; a modality shown to induce T(H)1 immunity) induced by hypericin-based photodynamic therapy. In an orthotopic HGG mouse model involving prophylactic/curative setups, both biologically and clinically relevant versions of ICD-based DC vaccines provided strong anti-HGG survival benefit. We found that the ability of DC vaccines to elicit HGG rejection was significantly blunted if cancer cell-associated reactive oxygen species and emanating danger signals were blocked either singly or concomitantly, showing hierarchical effect on immunogenicity, or if DCs, DC-associated MyD88 signal, or the adaptive immune system (especially CD8(+) T cells) were depleted. In a curative setting, ICD-based DC vaccines synergized with standard-of-care chemotherapy (temozolomide) to increase survival of HGG-bearing mice by ~300%, resulting in ~50% long-term survivors. Additionally, DC vaccines also induced an immunostimulatory shift in the brain immune contexture from regulatory T cells to T(H)1/cytotoxic T lymphocyte/T(H)17 cells. Analysis of the The Cancer Genome Atlas glioblastoma cohort confirmed that increased intratumor prevalence of T(H)1/cytotoxic T lymphocyte/T(H)17 cells linked genetic signatures was associated with good patient prognosis. Therefore, pending final preclinical checks, ICD-based vaccines can be clinically translated for glioma treatment.


Asunto(s)
Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Glioma/inmunología , Linfocitos T Citotóxicos/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Antracenos , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos/inmunología , Muerte Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glioma/patología , Ratones , Ratones Endogámicos C57BL , Clasificación del Tumor , Perileno/análogos & derivados , Perileno/farmacología , Perileno/uso terapéutico , Fotoquimioterapia , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T Citotóxicos/efectos de los fármacos
9.
J Immunother Cancer ; 3: 57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26676113

RESUMEN

Reliable mouse models are key in the discovery and development of novel anticancer treatments. Non-invasive monitoring techniques such as bioluminescence imaging (BLI) are useful tools to determine tumor engraftment and evaluate tumor growth. However, the development of ascites in ovarian cancer mouse models leads to possible difficulties. Ascites can interfere with the set-up of correct end points and can interfere with the evaluation of tumor volume using BLI. We provide optimized euthanasia criteria and in vivo data underlining the pitfalls of BLI.

10.
Int J Cancer ; 134(4): 873-84, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23929302

RESUMEN

Galectin-1 is a glycan-binding protein, which is involved in the aggressiveness of glioblastoma (GBM) in part by stimulating angiogenesis. In different cancer models, galectin-1 has also been demonstrated to play a pivotal role in tumor-mediated immune evasion especially by modulating cells of the adaptive immune system. It is yet unknown whether the absence or presence of galectin-1 within the glioma microenvironment also causes qualitative or quantitative differences in innate and/or adaptive antitumor immune responses. All experiments were performed in the orthotopic GL261 mouse high-grade glioma model. Stable galectin-1 knockdown was achieved via transduction of parental GL261 tumor cells with a lentiviral vector encoding a galectin-1-targeting miRNA. We demonstrated that the absence of tumor-derived but not of host-derived galectin-1 significantly prolonged the survival of glioma-bearing mice as such and in combination with dendritic cell (DC)-based immunotherapy. Both flow cytometric and pathological analysis revealed that the silencing of glioma-derived galectin-1 significantly decreased the amount of brain-infiltrating macrophages and myeloid-derived suppressor cells (MDSC) in tumor-bearing mice. Additionally, we revealed a pro-angiogenic role for galectin-1 within the glioma microenvironment. The data provided in this study reveal a pivotal role for glioma-derived galectin-1 in the regulation of myeloid cell accumulation within the glioma microenvironment, the most abundant immune cell population in high-grade gliomas. Furthermore, the prolonged survival observed in untreated and DC-vaccinated glioma-bearing mice upon the silencing of tumor-derived galectin-1 strongly suggest that the in vivo targeting of tumor-derived galectin-1 might offer a promising and realistic adjuvant treatment modality in patients diagnosed with GBM.


Asunto(s)
Inmunidad Adaptativa/inmunología , Neoplasias Encefálicas/inmunología , Células Dendríticas/inmunología , Galectina 1/fisiología , Glioma/inmunología , Inmunidad Innata/inmunología , Células Mieloides/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Glioma/metabolismo , Glioma/patología , Técnicas para Inmunoenzimas , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/patología , Neovascularización Patológica/prevención & control , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
11.
Anticancer Res ; 33(12): 5495-500, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24324087

RESUMEN

AIM: Treatment options are limited in uterine cancer, leading to a poor prognosis. Overexpression of Wilms' tumor gene 1 (WT1), the highest ranked tumor antigen, is attractive for immunotherapy. PATIENTS AND METHODS: Six pre-treated patients with uterine cancer received four weekly vaccines of autologous dendritic cells (DCs) electroporated with WT1 mRNA. Safety, feasibility and immunogenicity were assessed. In cases of response, patients received monthly booster vaccines. RESULTS: The technique was feasible. One patient had a local allergic reaction. Three out of four Human Leucocyte Antigen-A2 (HLA-A2)-positive patients showed an oncological response; an enrichment of WT1-specific T-cells was observed in two of them. Two HLA-A2-negative patients did not show an oncological or an immunological response. CONCLUSION: A first series of six patients with uterine cancer treated with WT1 mRNA-electroporated DCs is presented herein. Oncological and immunological responses were observed and are supportive for further research.


Asunto(s)
Células Dendríticas/metabolismo , Inmunoterapia , Neoplasias Uterinas/terapia , Proteínas WT1/genética , Anciano , Electroporación , Estudios de Factibilidad , Femenino , Humanos , Inmunoterapia/efectos adversos , Persona de Mediana Edad , ARN Mensajero/genética
12.
Anticancer Res ; 33(9): 3855-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24023319

RESUMEN

BACKGROUND: Dendritic cell (DC)-based immunotherapy is an emerging new treatment option in ovarian cancer, an important cause of cancer-related mortality. PATIENTS AND METHODS: One patient with ovarian carcinosarcoma (OCS) and one with serous ovarian cancer (SOC) received four weekly vaccinations of autologous DCs electroporated with mRNA coding for the Wilms' tumor gene 1 (WT1). Safety, feasibility and immunogenicity were assessed. RESULTS: Vaccination was feasible without toxicity. In an ex vivo antigen re-stimulation assay of peripheral blood mononuclear cells, both patients showed increasing cluster of differentiation 137 (CD137+) antigen-specific T-cells and interleukin 10 (IL-10) production post-vaccination. Moreover, interleukin-2 (IL-2) production increased (OCS) as well as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) (SOC). Disease in patients progressed after four vaccines and patients continued with conventional therapies. After cessation of immunotherapy, they had an extended survival of 19 (OCS) and 12 (SOC) months. CONCLUSION: To our knowledge, we report for the first time the feasibility and T-cell immunogenicity of WT1 mRNA-loaded DC immunotherapy in ovarian cancer.


Asunto(s)
Carcinosarcoma/terapia , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias Ováricas/terapia , ARN Mensajero/genética , Proteínas WT1/genética , Carcinosarcoma/inmunología , Carcinosarcoma/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Electroporación , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Análisis de Supervivencia
13.
J Neurooncol ; 115(1): 9-17, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23824536

RESUMEN

High-grade gliomas (HGG) are the most common and most aggressive intrinsic human brain tumors in adults. Galectin-1, a glycan-binding protein that is overexpressed in HGG, has been shown to contribute significantly to the aggressive nature of HGG. It is unknown whether increased galectin-1 expression levels are exclusively found at the tumor site or whether galectin-1 can also be detected in the serum of HGG patients. Galectin-1 serum levels were analyzed in a prospective dataset of 43 healthy controls and 125 patients with newly diagnosed or recurrent HGG. Samples were taken at the moment of surgical resection and/or 2-3 weeks after surgery. Galectin-1 serum levels were determined using an ELISA for galectin-1. Galectin-1 serum levels depended significantly on age and sex in the control group. Age- and sex-adjusted galectin-1 serum levels were significantly higher in all patient subgroups compared to healthy controls with a high discriminative ability that increased with age. We did not observe a significant decrease in the galectin-1 serum levels upon surgical resection of the tumor. Collectively, the data presented here may represent a first step to establish galectin-1 as a biomarker in HGG disease monitoring. Further longitudinal evaluation is required and ongoing to investigate the value of galectin-1 serum levels in HGG patients as an additional diagnostic marker, but more importantly as a predictor of treatment response and prognosis. Furthermore, galectin-1 serum levels could also provide an important tool for the identification of HGG patients that could benefit from galectin-1 directed therapies that are currently under development.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/diagnóstico , Galectina 1/sangre , Glioma/sangre , Glioma/diagnóstico , Adolescente , Adulto , Anciano , Encéfalo/metabolismo , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/diagnóstico , Pronóstico , Estudios Prospectivos , Adulto Joven
14.
Clin Dev Immunol ; 2013: 952469, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23710206

RESUMEN

The recruitment and activation of regulatory T cells (Tregs) in the micro-environment of malignant brain tumors has detrimental effects on antitumoral immune responses. Hence, local elimination of Tregs within the tumor micro-environment represents a highly valuable tool from both a fundamental and clinical perspective. In the syngeneic experimental GL261 murine glioma model, Tregs were prophylactically eliminated through treatment with PC61, an anti-CD25 mAb. This resulted in specific elimination of CD4+CD25hiFoxp3+ Treg within brain-infiltrating lymphocytes and complete protection against subsequent orthotopic GL261 tumor challenge. Interestingly, PC61-treated mice also showed a pronounced infiltration of CD11b+ myeloid cells in the brain. Phenotypically, these cells could not be considered as Gr-1+ myeloid-derived suppressor cells (MDSC) but were identified as F4/80+ macrophages and granulocytes.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Glioma/tratamiento farmacológico , Depleción Linfocítica , Células Mieloides/efectos de los fármacos , Animales , Encéfalo/inmunología , Encéfalo/patología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Glioma/inmunología , Glioma/patología , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Ratones , Células Mieloides/inmunología , Células Mieloides/patología , Neoplasias Experimentales , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
15.
J Invest Dermatol ; 132(9): 2245-54, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22622427

RESUMEN

Aggressiveness of advanced melanomas relates in part to their marked propensity to develop neoangiogenesis and metastases. Among its numerous pro-cancer roles, galectin (gal)-1 expressed and/or secreted by both cancer and endothelial cells stimulates proliferation and angiogenesis. This study first shows that gal-1 is more highly expressed at both mRNA and protein levels than its congeners in melanomas and particularly in advanced lesions. The roles of gal-1 were further investigated in vivo in the highly proliferating and vascularized pseudometastatic B16F10 mouse melanoma model using stable knockdown B16F10 cells and wild-type versus gal-1 knockout mice, and then in vitro in B16F10 tumoral and lung microvascular cells. Gal-1 depletion in the B16F10 tumor cells but not in the tumor-bearing mice significantly increased melanoma-bearing mice survival. Tumor-derived gal-1 thus seems to have more critical roles than the host-derived one. In fact, gal-1 displays distinct effects on the H-Ras-dependent p53/p21 pathways: in primary lung microvessel endothelial cells, gal-1 seems to be involved in the maintenance of senescent status through the induction of both p53 and p21 while it stimulates B16F10 cancer cell proliferation through a p53/p21 decrease. Altogether, these data point to gal-1 as a potential target to combat melanomas.


Asunto(s)
Galectina 1/metabolismo , Melanoma Experimental/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Técnicas de Silenciamiento del Gen , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Melanoma Experimental/genética , Melanoma Experimental/secundario , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
16.
Cancer Immunol Immunother ; 61(11): 2033-44, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22527250

RESUMEN

PURPOSE: Dendritic cell (DC)-based tumor vaccination has rendered promising results in relapsed high-grade glioma patients. In the HGG-2006 trial (EudraCT 2006-002881-20), feasibility, toxicity, and clinical efficacy of the full integration of DC-based tumor vaccination into standard postoperative radiochemotherapy are studied in 77 patients with newly diagnosed glioblastoma. PATIENTS AND METHODS: Autologous DC are generated after leukapheresis, which is performed before the start of radiochemotherapy. Four weekly induction vaccines are administered after the 6-week course of concomitant radiochemotherapy. During maintenance chemotherapy, 4 boost vaccines are given. Feasibility and progression-free survival (PFS) at 6 months (6mo-PFS) are the primary end points. Overall survival (OS) and immune profiling, rather than monitoring, as assessed in patients' blood samples, are the secondary end points. Analysis has been done on intent-to-treat basis. RESULTS: The treatment was feasible without major toxicity. The 6mo-PFS was 70.1 % from inclusion. Median OS was 18.3 months. Outcome improved significantly with lower EORTC RPA classification. Median OS was 39.7, 18.3, and 10.7 months for RPA classes III, IV, and V, respectively. Patients with a methylated MGMT promoter had significantly better PFS (p = 0.0027) and OS (p = 0.0082) as compared to patients with an unmethylated status. Exploratory "immunological profiles" were built to compare to clinical outcome, but no statistical significant evidence was found for these profiles to predict clinical outcome. CONCLUSION: Full integration of autologous DC-based tumor vaccination into standard postoperative radiochemotherapy for newly diagnosed glioblastoma seems safe and possibly beneficial. These results were used to power the currently running phase IIb randomized clinical trial.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/trasplante , Glioblastoma/terapia , Inmunoterapia , Nivel de Atención , Adulto , Anciano , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Quimioradioterapia , Terapia Combinada/métodos , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Células Dendríticas/inmunología , Supervivencia sin Enfermedad , Femenino , Glioblastoma/inmunología , Glioblastoma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Trasplante Autólogo , Resultado del Tratamiento , Proteínas Supresoras de Tumor/genética
17.
Expert Rev Neurother ; 11(4): 533-43, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21469926

RESUMEN

The prognosis of patients diagnosed with high-grade glioma continues to be dismal in spite of multimodal treatment. Active specific immunotherapy by means of dendritic cell vaccination is considered to be a new promising concept that aims at generating an anti-tumoral immune response. However, it is now widely accepted that the success of immunotherapeutic strategies to promote tumor regression will rely not only on enhancing the effector arm of the immune response but also on downregulation of the counteracting tolerogenic signals. In this article, we summarize evidence that galectin-1, an evolutionarily conserved glycan-binding protein that is abundantly expressed in high-grade glioma, is an important player in glioma-mediated immune escape.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Galectina 1/fisiología , Glioma/terapia , Inmunoterapia Activa , Escape del Tumor , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Glioma/inmunología , Glioma/patología , Humanos
18.
Brain Pathol ; 19(4): 694-712, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19744041

RESUMEN

The prognosis of patients with malignant glioma is poor in spite of multimodal treatment approaches consisting of neurosurgery, radiochemotherapy and maintenance chemotherapy. Among innovative treatment strategies like targeted therapy, antiangiogenesis and gene therapy approaches, immunotherapy emerges as a meaningful and feasible treatment approach for inducing long-term survival in at least a subpopulation of these patients. Setting up immunotherapy for an inherent immunosuppressive tumor located in an immune-privileged environment requires integration of a lot of scientific input and knowledge of both tumor immunology and neuro-oncology. The field of immunotherapy is moving into the direction of active specific immunotherapy using autologous dendritic cells (DCs) as vehicle for immunization. In the translational research program of the authors, the whole cascade from bench to bed to bench of active specific immunotherapy for malignant glioma is covered, including proof of principle experiments to demonstrate immunogenicity of patient-derived mature DCs loaded with autologous tumor lysate, preclinical in vivo experiments in a murine orthotopic glioma model, early phase I/II clinical trials for relapsing patients, a phase II trial for patients with newly diagnosed glioblastoma (GBM) for whom immunotherapy is integrated in the current multimodal treatment, and laboratory analyses of patient samples. The strategies and results of this program are discussed in the light of the internationally available scientific literature in this fast-moving field of basic science and translational clinical research.


Asunto(s)
Neoplasias Encefálicas/terapia , Células Dendríticas/inmunología , Glioma/terapia , Animales , Protocolos Antineoplásicos , Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Terapia Combinada , Modelos Animales de Enfermedad , Glioma/inmunología , Humanos , Inmunoterapia , Ratones , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA