Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Cancer ; 143(3): 686-698, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29464699

RESUMEN

Improved understanding of cancer immunology has provided insight into the phenomenon of frequent tumor recurrence after initially successful immunotherapy. A delicate balance exists between the capacity of the immune system to control tumor growth and various resistance mechanisms that arise to avoid or even counteract the host's immune system. These resistance mechanisms include but are not limited to (i) adaptive expression of inhibitory checkpoint molecules in response to the proinflammatory environment and (ii) amplification of cancer stem cells, a small fraction of tumor cells possessing the capacity for self-renewal and mediating treatment resistance and formation of metastases after long periods of clinical remission. Several individual therapeutic agents have so far been developed to revert T-cell exhaustion or disrupt the cross-talk between cancer stem cells and the tumor-promoting microenvironment. Here, we demonstrate that a three-arm combination therapy-consisting of an mRNA-based vaccine to induce antigen-specific T-cell responses, monoclonal antibodies blocking inhibitory checkpoint molecules (PD-1, TIM-3, LAG-3), and antibodies targeting IL-6 and TGF-ß-improves the therapeutic outcome in subcutaneous TC-1 tumors and significantly prolongs survival of treated mice. Our findings point to a need for a rational development of multidimensional anticancer therapies, aiming at the induction of tumor-specific immunity and simultaneously targeting multiple resistance mechanisms.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Interleucina-6/antagonistas & inhibidores , Neoplasias/genética , Neoplasias/metabolismo , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunoterapia , Interleucina-6/metabolismo , Melanoma Experimental , Ratones , Neoplasias/patología , Neoplasias/terapia , Recurrencia , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
2.
Oncotarget ; 8(26): 41932-41946, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28410210

RESUMEN

Blockade of the inhibitory PD-1/PD-L1 immune checkpoint axis is a promising cancer treatment. Nonetheless, a significant number of patients and malignancies do not respond to this therapy. To develop a screen for response to PD-1/PD-L1 inhibition, it is critical to develop a non-invasive tool to accurately assess dynamic immune checkpoint expression. Here we evaluated non-invasive SPECT/CT imaging of PD-L1 expression, in murine tumor models with varying PD-L1 expression, using high affinity PD-L1-specific nanobodies (Nbs). We generated and characterized 37 Nbs recognizing mouse PD-L1. Among those, four Nbs C3, C7, E2 and E4 were selected and evaluated for preclinical imaging of PD-L1 in syngeneic mice. We performed SPECT/CT imaging in wild type versus PD-L1 knock-out mice, using Technetium-99m (99mTc) labeled Nbs. Nb C3 and E2 showed specific antigen binding and beneficial biodistribution. Through the use of CRISPR/Cas9 PD-L1 knock-out TC-1 lung epithelial cell lines, we demonstrate that SPECT/CT imaging using Nb C3 and E2 identifies PD-L1 expressing tumors, but not PD-L1 non-expressing tumors, thereby confirming the diagnostic potential of the selected Nbs. In conclusion, these data show that Nbs C3 and E2 can be used to non-invasively image PD-L1 levels in the tumor, with the strength of the signal correlating with PD-L1 levels. These findings warrant further research into the use of Nbs as a tool to image inhibitory signals in the tumor environment.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Trazadores Radiactivos , Anticuerpos de Dominio Único , Animales , Biomarcadores , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones , Ratones Noqueados , Neoplasias/patología , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único
3.
Methods Mol Biol ; 1499: 179-191, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27987150

RESUMEN

Recent advances in molecular biology have led to dramatic enhancement of the stability of in vitro transcribed (IVT) messenger RNA (mRNA) and improvement in its translational efficacy. Nowadays, mRNA-based vaccines represent a promising approach in the field of anticancer immunotherapy, gaining attention over the earlier-established bacteria-, virus-, or cell-based vaccination approaches. Here, we present the experimental procedures employed in our laboratory to induce anticancer immune responses in different murine tumor models using IVT mRNA encoding for immune activation signals and antigens of interest.


Asunto(s)
Vacunas contra el Cáncer/inmunología , ARN Mensajero/inmunología , Animales , Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Femenino , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Vacunación/métodos
4.
Sci Rep ; 6: 22509, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26931556

RESUMEN

The lack of appropriate mouse models is likely one of the reasons of a limited translational success rate of therapeutic vaccines against cervical cancer, as rapidly growing ectopic tumours are commonly used for preclinical studies. In this work, we demonstrate that the tumour microenvironment of TC-1 tumours differs significantly depending on the anatomical location of tumour lesions (i.e. subcutaneously, in the lungs and in the genital tract). Our data demonstrate that E7-TriMix mRNA vaccine-induced CD8(+) T lymphocytes migrate into the tumour nest and control tumour growth, although they do not express mucosa-associated markers such as CD103 or CD49a. We additionally show that despite the presence of the antigen-specific T cells in the tumour lesions, the therapeutic outcomes in the genital tract model remain limited. Here, we report that such a hostile tumour microenvironment can be reversed by cisplatin treatment, leading to a complete regression of clinically relevant tumours when combined with mRNA immunization. We thereby demonstrate the necessity of utilizing clinically relevant models for preclinical evaluation of anticancer therapies and the importance of a simultaneous combination of anticancer immune response induction with targeting of tumour environment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Membrana Mucosa/patología , ARN Mensajero/inmunología , Neoplasias del Cuello Uterino/inmunología , Vacunas/administración & dosificación , Animales , Femenino , Humanos , Memoria Inmunológica , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral , Neoplasias del Cuello Uterino/patología , Vacunas/inmunología
5.
Cancer Immunol Res ; 4(2): 146-56, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26659303

RESUMEN

Modulating the activity of tumor-infiltrating dendritic cells (TiDC) provides opportunities for novel cancer interventions. In this article, we report on our study of the uptake of mRNA by CD8α(+) cross-presenting TiDCs upon its intratumoral (i.t.) delivery. We exploited this property to deliver mRNA encoding the costimulatory molecule CD70, the activation stimuli CD40 ligand, and constitutively active Toll-like receptor 4, referred to as TriMix mRNA. We show that TiDCs are reprogrammed to mature antigen-presenting cells that migrate to tumor-draining lymph nodes (TDLN). TriMix stimulated antitumor T-cell responses to spontaneously engulfed cancer antigens, including a neoepitope. We show in various mouse cancer models that i.t. delivery of TriMix mRNA results in systemic therapeutic antitumor immunity. Finally, we show that the induction of antitumor responses critically depends on TiDCs, whereas it only partially depends on TDLNs. As such, we provide a platform and a mechanistic rationale for the clinical testing of i.t. administration of TriMix mRNA.


Asunto(s)
Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos , Neoplasias/genética , Neoplasias/inmunología , ARN Mensajero/genética , Linfocitos T/inmunología , Animales , Biomarcadores , Ligando CD27/genética , Ligando de CD40/genética , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones , Neoplasias/mortalidad , Neoplasias/patología , Fenotipo , ARN Mensajero/administración & dosificación , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/metabolismo , Receptor Toll-Like 4/genética
6.
Oncotarget ; 6(3): 1359-81, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25682197

RESUMEN

The identification of tumor-specific antigens and the immune responses directed against them has instigated the development of therapies to enhance antitumor immune responses. Most of these cancer immunotherapies are administered systemically rather than directly to tumors. Nonetheless, numerous studies have demonstrated that intratumoral therapy is an attractive approach, both for immunization and immunomodulation purposes. Injection, recruitment and/or activation of antigen-presenting cells in the tumor nest have been extensively studied as strategies to cross-prime immune responses. Moreover, delivery of stimulatory cytokines, blockade of inhibitory cytokines and immune checkpoint blockade have been explored to restore immunological fitness at the tumor site. These tumor-targeted therapies have the potential to induce systemic immunity without the toxicity that is often associated with systemic treatments. We review the most promising intratumoral immunotherapies, how these affect systemic antitumor immunity such that disseminated tumor cells are eliminated, and which approaches have been proven successful in animal models and patients.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Humanos
7.
Expert Rev Vaccines ; 14(2): 235-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25263094

RESUMEN

About 25 years ago, mRNA became a tool of interest in anticancer vaccination approaches. However, due to its rapid degradation in situ, direct application of mRNA was confronted with considerable skepticism during its early use. Consequently, mRNA was for a long time mainly used for the ex vivo transfection of dendritic cells, professional antigen-presenting cells known to stimulate immunity. The interest in direct application of mRNA experienced a revival, as researchers became aware of the many advantages mRNA offers. Today, mRNA is considered to be an ideal vehicle for the induction of strong immune responses against cancer. The growing numbers of preclinical trials and as a consequence the increasing clinical application of mRNA as an off-the-shelf anticancer vaccine signifies a renaissance for transcript-based antitumor therapy. In this review, we highlight this renaissance using a timeline providing all milestones in the application of mRNA for anticancer vaccination.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Neoplasias/terapia , ARN Mensajero/inmunología , Vacunación , Vacunas de ADN/administración & dosificación , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Células Dendríticas/inmunología , Humanos , Inmunoterapia , Neoplasias/inmunología , Vacunas de ADN/inmunología
8.
Oncoimmunology ; 4(4): e998107, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26137411

RESUMEN

Melanoma patients are at a high risk of developing brain metastases, which are strongly vascularized and therefore have a significant risk of spontaneous bleeding. VEGF not only plays a role in neo-angiogenesis but also in the antitumor immune response. VEGFR-targeted therapy might not only have an impact on the tumor vascularization but also on tumor-infiltrating immune cells. In this study, we investigated the effect of axitinib, a small molecule TKI of VEGFR-1, -2, and -3, on tumor growth and on the composition of tumor-infiltrating immune cells in subcutaneous and intracranial mouse melanoma models. In vivo treatment with axitinib induced a strong inhibition of tumor growth and significantly improved survival in both tumor models. Characterization of the immune cells within the spleen and tumor of tumor-bearing mice respectively showed a significant increase in the number of CD3+CD8+ T cells and CD11b+ cells of axitinib-treated mice. More specifically, we observed a significant increase of intratumoral monocytic myeloid-derived suppressor cells (moMDSCs; CD11b+Ly6ChighLy6G-). Interestingly, in vitro proliferation assays showed that moMDSCs isolated from spleen or tumor of axitinib-treated mice had a reduced suppressive capacity on a per cell basis as compared to those isolated from vehicle-treated mice. Moreover, MDSCs from axitinib-treated animals displayed the capacity to stimulate allogeneic T cells. Thus, treatment with axitinib induces differentiation of moMDSC toward an antigen-presenting phenotype. Based on these observations, we conclude that the impact of axitinib on tumor growth and survival is most likely not restricted to direct anti-angiogenic effects but also involves important effects on tumor immunity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA