Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Intervalo de año de publicación
1.
Clin Cancer Res ; 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630754

RESUMEN

PURPOSE: Cancer patients frequently undergo radiotherapy in their clinical management with unintended irradiation of blood vessels and copiously irrigated organs in which polymorphonuclear leukocytes circulate. Following the observation that such low doses of ionizing radiation are able to induce neutrophils to extrude neutrophil extracellular traps (NETs), we have investigated the mechanisms, consequences and the occurrence of such phenomena in patients undergoing radiotherapy. EXPERIMENTAL DESIGN: NETosis was analyzed in cultures of neutrophils isolated from healthy donors, cancer patients and cancer-bearing mice under confocal microscopy. Cocultures of radiation-induced NETs, immune effector lymphocytes and tumor cells were used to study the effects of irradiation-induced NETs on immune cytotoxicity. Radiation-induced NETs were intravenously injected to mice assessing their effects on metastasis. Circulating NETs in irradiated cancer patients were measured by ELISA methods detecting MPO-DNA complexes and citrullinated H3. RESULTS: Very low γ-radiation doses (0.5-1 Gy) given to neutrophils elicit NET formation in a manner dependent on oxidative stress, NADPH oxidase activity and autocrine interleukin-8. Radiation-induced NETs interfere with NK- and T-cell cytotoxicity. As a consequence, pre-injection of irradiation-induced NETs increases the number of successful metastases in mouse tumor models. Increases in circulating NETs were readily detected in two prospective series of patients following the first fraction of their radiotherapy courses. CONCLUSIONS: NETosis is induced by low-dose ionizing irradiation in a neutrophil-intrinsic fashion and radiation-induced NETs are able to interfere with immune-mediated cytotoxicity. Radiation-induced NETs foster metastasis in mouse models and can be detected in the circulation of patients undergoing conventional radiotherapy treatments.

2.
Mol Ther Nucleic Acids ; 33: 599-616, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37637207

RESUMEN

IL-12 is a potent cytokine for cancer immunotherapy. However, its systemic delivery as a recombinant protein has shown unacceptable toxicity in the clinic. Currently, the intratumoral injection of IL-12-encoding mRNA or DNA to avoid such side effects is being evaluated in clinical trials. In this study, we aimed to improve this strategy by further favoring IL-12 tethering to the tumor. We generated in vitro transcribed mRNAs encoding murine single-chain IL-12 fused to diabodies binding to CSF1R and/or PD-L1. These targeted molecules are expressed in the tumor microenvironment, especially on myeloid cells. The binding capacity of chimeric constructs and the bioactivity of IL-12 were demonstrated in vitro and in vivo. Doses as low as 0.5 µg IL-12-encoding mRNA achieved potent antitumor effects in subcutaneously injected B16-OVA and MC38 tumors. Treatment delivery was associated with increases in IL-12p70 and IFN-γ levels in circulation. Fusion of IL-12 to the diabodies exerted comparable efficacy against bilateral tumor models. However, it achieved tethering to myeloid cells infiltrating the tumor, resulting in nearly undetectable systemic levels of IL-12 and IFN-γ. Overall, tethering IL-12 to intratumoral myeloid cells in the mRNA-transferred tumors achieves similar efficacy while reducing the dangerous systemic bioavailability of IL-12.

3.
Mol Ther Nucleic Acids ; 33: 668-682, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37650116

RESUMEN

Intratumoral immunotherapy strategies for cancer based on interleukin-12 (IL-12)-encoding cDNA and mRNA are under clinical development in combination with anti-PD-(L)1 monoclonal antibodies. To make the most of these approaches, we have constructed chimeric mRNAs encoding single-chain IL-12 fused to single-chain fragment variable (scFv) antibodies that bind to transforming growth factor ß (TGF-ß) and CD137 (4-1BB). Several neutralizing TGF-ß agents and CD137 agonists are also undergoing early-phase clinical trials. To attain TGF-ß and CD137 binding by the constructions, we used bispecific tandem scFv antibodies (taFvs) derived from the specific 1D11 and 1D8 monoclonal antibodies (mAbs), respectively. Transfection of mRNAs encoding the chimeric constructs achieved functional expression of the proteins able to act on their targets. Upon mRNA intratumoral injections in the transplantable mouse cancer models CT26, MC38, and B16OVA, potent therapeutic effects were observed following repeated injections into the tumors. Efficacy was dependent on the number of CD8+ T cells able to recognize tumor antigens that infiltrated the malignant tissue. Although the abscopal effects on concomitant uninjected lesions were modest, such distant effects on untreated lesions were markedly increased when combined with systemic PD-1 blockade.

4.
Clin Cancer Res ; 29(23): 4711-4727, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37522874

RESUMEN

In cancer pathogenesis, soluble mediators are responsible for a type of inflammation that favors the progression of tumors. The mechanisms chiefly involve changes in the cellular composition of the tumor tissue stroma and in the functional modulation of myeloid and lymphoid leukocytes. Active immunosuppression, proangiogenesis, changes in leukocyte traffic, extracellular matrix remodeling, and alterations in tumor-antigen presentation are the main mechanisms linked to the inflammation that fosters tumor growth and metastasis. Soluble inflammatory mediators and their receptors are amenable to various types of inhibitors that can be combined with other immunotherapy approaches. The main proinflammatory targets which can be interfered with at present and which are under preclinical and clinical development are IL1ß, IL6, the CXCR1/2 chemokine axis, TNFα, VEGF, leukemia inhibitory factor, CCL2, IL35, and prostaglandins. In many instances, the corresponding neutralizing agents are already clinically available and can be repurposed as a result of their use in other areas of medicine such as autoimmune diseases and chronic inflammatory conditions.


Asunto(s)
Mediadores de Inflamación , Neoplasias , Humanos , Neoplasias/terapia , Inmunoterapia , Inflamación , Quimiocinas
5.
Oncoimmunology ; 12(1): 2147317, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36531687

RESUMEN

Previous studies have shown that local delivery of tumor antigen-specific CD8+ T lymphocytes engineered to transiently express single-chain IL-12 mRNA is highly efficacious. Peritoneal dissemination of cancer is a frequent and often fatal patient condition usually diagnosed when the tumor burden is too large and hence uncontrollable with current treatment options. In this study, we have modeled intracavitary adoptive T cell therapy with OVA-specific OT-I T cells electroporated with IL-12 mRNA to treat B16-OVA and PANC02-OVA tumor spread in the peritoneal cavity. Tumor localization in the omentum and the effects of local T-cell encounter with the tumor antigens were monitored, the gene expression profile evaluated, and the phenotypic reprogramming of several immune subsets was characterized. Intraperitoneal administration of T cells promoted homing to the omentum more effectively than intravenous administration. Transient IL-12 expression was responsible for a favorable reprogramming of the tumor immune microenvironment, longer persistence of transferred T lymphocytes in vivo, and the development of immunity to endogenous antigens following primary tumor eradication. The efficacy of the strategy was at least in part recapitulated with the adoptive transfer of lower affinity transgenic TCR-bearing PMEL-1 T lymphocytes to treat the aggressive intraperitoneally disseminated B16-F10 tumor. Locoregional adoptive transfer of transiently IL-12-armored T cells appears to offer promising therapeutic advantages in terms of anti-tumor efficacy to treat peritoneal carcinomatosis.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias Peritoneales , Ratones , Animales , Interleucina-12/genética , ARN Mensajero/genética , Neoplasias Peritoneales/terapia , Traslado Adoptivo , Antígenos de Neoplasias/genética , Modelos Animales de Enfermedad , Microambiente Tumoral
6.
Cancer Immunol Res ; 11(2): 184-198, 2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-36478221

RESUMEN

IL12-based local gene therapy of cancer constitutes an active area of clinical research using plasmids, mRNAs, and viral vectors. To improve antitumor effects, we have experimentally tested the combination of mRNA constructs encoding IL12 and IL18. Moreover, we have used a form of IL18 [decoy-resistant IL18 (DR-18)] which has preserved bioactivity but does not bind to the IL18 binding protein decoy receptor. Both cytokines dramatically synergize to induce IFNγ release from mouse splenocytes, and, if systemically cotransferred to the liver, they mediate lethal toxicity. However, if given intratumorally to B16OVA tumor-bearing mice, the combination attains efficacy against the directly treated tumor and moderate tumor-delaying activity on distant noninjected lesions. Cotreatment was conducive to the presence of more activated CD8+ T cells in the treated and noninjected tumors. In keeping with these findings, the efficacy of treatment was contingent on the integrity of CD8+ T cells and cDC1 dendritic cells in the treated mice. Furthermore, efficacy of IL12 plus DR-18 local mRNA coinjection against distant concomitant tumors could be enhanced upon combination with anti-PD-1 mAb systemic treatment, thus defining a feasible synergistic immunotherapy strategy.


Asunto(s)
Interleucina-18 , Neoplasias , Animales , Ratones , Neoplasias/genética , Neoplasias/terapia , Linfocitos T CD8-positivos , Inmunoterapia , Interleucina-12/metabolismo
7.
Pharmacol Ther ; 239: 108189, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35430292

RESUMEN

Interleukin-12 is considered a potent agent to enhance antitumor immune responses. It belongs to a family of heterodimeric cytokines with key roles in the up-regulation and down-regulation of cellular immunity. Since its discovery, recombinant IL-12 was found to exert potent antitumor effects in rodent tumor models and was rapidly tested in the clinic with an unfavorable benefit/toxicity profile. Localized delivery of IL-12 dramatically improves the therapeutic index and this approach is being applied in the clinic based on in-vivo electroporation of naked plasmid DNA encoding IL-12, mRNA formulations, viral vectors and tumor-targeted fusion proteins. Other biotechnology strategies such as IL-12-engineered local adoptive cell therapy and pro-cytokines can also be used to improve results and broaden the therapeutic window. Combination strategies of such localized IL-12-based approaches with checkpoint inhibitors are yielding promising results both preclinically and in the early-phase clinical trials.


Asunto(s)
Interleucina-12 , Neoplasias , Humanos , Interleucina-12/genética , Inmunoterapia/métodos , Vectores Genéticos , Inmunoterapia Adoptiva , Factores Inmunológicos , Neoplasias/terapia
8.
Inmunología (1987) ; 29(1): 50-55, ene.-mar. 2010. ilus
Artículo en Inglés | IBECS | ID: ibc-108266

RESUMEN

La división de tareas entre las células del sistema inmunitario es undenominador común en la organización de las diferentes respuestas quelleva a cabo. Como excepción a este paradigma, una subpoblación leucocitaria de ratones parece estar equipada para matar células transformadas einfectadas con virus y, a la vez, con la capacidad de procesamiento antigé-nico y presentación a linfocitos T. Estas funciones se adscriben clásicamente de forma separada a linfocitos NK y a células dendríticas. Las IKDC (“Interferon-producing killer dendritic cells”) fueron definidas en ratón como células que expresan en su superficie CD11c con menor intensidad que las células dendríticas convencionales mientras que coexpresan B220, NK1.1, CD49b(VLA-2) y MHC de Clase II. La ausencia de CD3, CD19 y Gr1 (Ly49C) caracteriza también esta subpoblación cuando se estudia mediante citometría de (..) (AU)


Division of labour among cells of the immune system is a prevailingtheme in the orchestration of immune responses. Contrary to this paradigm, a particular leukocyte population in the mouse seems to be equipped with the ability to kill transformed and virally infected cells and withthe capability to mediate antigen processing and presentation to T cells.Those two functions are classically ascribed separately to Natural Killer(NK) cells and Dendritic cells (DC). IKDC (interferon-producing killerdendritic cell) were defined in mice as cells expressing CD11c, althoughto a lesser extent than conventional DCs (cDCs), while coexpressing B220,NK1.1, CD49b (VLA-2) and MHC Class II molecules. Absence of CD3,CD19 and Gr1(Ly49c) expression also featured this minor subset uponmulticolour FACS characterization. These cells kill a variety tumor celllines in a TRAIL-dependent fashion. Furthermore, IKDC produce high (..) (AU)


Asunto(s)
Humanos , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Interferón gamma/inmunología , Antígenos/inmunología , Tejido Linfoide/inmunología
9.
Expert Opin Biol Ther ; 7(5): 599-615, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17477799

RESUMEN

There is compelling evidence for the role of natural killer (NK) cells in tumor immunosurveillance and their beneficial effects on many experimentally successful immunotherapy strategies. NK cells mediate cell contact-dependent cellular cytotoxicity and produce pro-inflammatory cytokines, but do not rearrange antigen receptors. Their activation depends on various germline-encoded receptors, including CD16, which mediates recognition of antibody-coated target cells. NK cytotoxicity is checked by a repertoire of inhibitory receptors that scan adequate expression of major histocompatibility complex class I molecules on the potential target cell. Functional cross-talk of NK and dendritic cells suggests a critical role for NK cells in the initiation and regulation of cellular immunity. Considerable knowledge on the molecular basis of NK recognition/activation contrasts with a lack of successful translational research on these matters. However, there is plenty of opportunity for targeted intervention of inhibitory/activatory surface receptors and for adoptive cell therapy with autologous or allogeneic NK cells.


Asunto(s)
Comunicación Celular/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Citotoxicidad Inmunológica , Humanos , Inmunidad Celular , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Ligandos , Ratones , Neoplasias/metabolismo , Neoplasias/terapia , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Receptores de Células Asesinas Naturales
10.
Int J Cancer ; 116(2): 275-81, 2005 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-15800914

RESUMEN

In the course of a clinical trial consisting of intratumoral injections of dendritic cells (DCs) transfected to produce interleukin-12, the use of (111)In-labeled tracing doses of DCs showed that most DCs remained inside tumor tissue, instead of migrating out. In search for factors that could explain this retention, it was found that tumors from patients suffering hepatocellular carcinoma, colorectal or pancreatic cancer were producing IL-8 and that this chemokine attracted monocyte-derived dendritic cells that uniformly express both IL-8 receptors CXCR1 and CXCR2. Accordingly, neutralizing antihuman IL-8 monoclonal antibodies blocked the chemotactic attraction of DCs by recombinant IL-8, as well as by the serum of the patients or culture supernatants of human colorectal carcinomas. In addition, tissue culture supernatants of colon carcinoma cells inhibited DC migration induced by MIP-3beta in an IL-8-dependent fashion. IL-8 production in malignant tissue and the responsiveness of DCs to IL-8 are a likely explanation of the clinical images, which suggest retention of DCs inside human malignant lesions. Impairment of DC migration toward lymphoid tissue could be involved in cancer immune evasion.


Asunto(s)
Movimiento Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Células Dendríticas/inmunología , Interleucina-8/biosíntesis , Interleucina-8/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Anticuerpos Monoclonales , Quimiotaxis , Humanos , Inmunoterapia , Interleucina-12/biosíntesis , Transfección
11.
Expert Opin Biol Ther ; 5(1): 7-22, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15709906

RESUMEN

Like paratroopers in special operations, dendritic cells (DCs) can be deployed behind the enemy borders of malignant tissue to ignite an antitumour immune response. 'Cross-priming T cell responses' is the code name for their mission, which consists of taking up antigen from transformed cells or their debris, migrating to lymphoid tissue ferrying the antigenic cargo, and meeting specific T cells. This must be accomplished in such an immunogenic manner that specific T lymphocytes would mount a robust enough response as to fully reject the malignancy. To improve their immunostimulating activity, local gene therapy can be very beneficial, either by transfecting DCs with genes enhancing their performance, or by preparing tumour tissue with pro-inflammatory mediators. In addition, endogenous DCs from the tumour host can be attracted into the malignant tissue following transfection of certain chemokine genes into tumour cells. On their side, tumour stroma and malignant cells set up a hostile immunosuppressive environment for artificially released or attracted DCs. This milieu is usually rich in transforming growth factor-beta, vascular endothelial growth factor, and IL-10, -6 and -8, among other substances that diminish DC performance. Several molecular strategies are being devised to interfere with the immunosuppressive actions of these substances and to further enhance the level of anticancer immunity achieved after artificial release of DCs intratumourally.


Asunto(s)
Células Dendríticas/inmunología , Ambiente , Terapia Genética/métodos , Neoplasias/inmunología , Animales , Ingeniería Genética/métodos , Humanos , Neoplasias/genética
12.
Clin Cancer Res ; 9(15): 5454-64, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14654524

RESUMEN

Immunotherapeutic monoclonal antibodies (mAbs) can be defined as those that exert their functions by tampering with immune system cell molecules, causing an enhancement of antitumor immune responses. Some of these antibodies are agonistic ligands for surface receptors involved in the activation of lymphocytes and/or antigen-presenting cells, whereas others are antagonists of mechanisms that normally limit the intensity of immune reactions. Several mAbs of this category have been described to display in vivo antitumor activity in mouse models. Only anti-CTLA-4 (CD152) mAb has entered clinical trials, but the preclinical effects described for anti-CD40, anti-CD137 (4-1BB), anti-CD102 (intercellular adhesion molecule-2), and regulatory T cell-depleting mAbs should lead to their prompt clinical development. Their use in combination with immunizations against tumor antigens has been reported to be endowed with synergistic properties. This new group of antitumor agents holds promise for at least additive effects with conventional therapies of cancer and deserves intensive translational research.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias/inmunología , Antígenos CD/inmunología , Antígenos de Diferenciación/inmunología , Antígeno CTLA-4 , Moléculas de Adhesión Celular/inmunología , Humanos , Inmunidad Celular , Modelos Inmunológicos , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral
13.
Arch Immunol Ther Exp (Warsz) ; 50(1): 13-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11916305

RESUMEN

Cellular immune responses can destroy cancer cells, achieving the cure of experimental malignancies. An expanding wealth of knowledge on the molecular basis of how to prime and amplify a T cell response has fueled a number of strategies successful at treating established tumors (rather than merely preventing tumor grafting). The most efficacious approaches operate at different stages, including: 1) priming the immune response using tumor antigen-expressing dendritic cells or tumor cells transfected with genes that render them immunogenic, 2) sustaining and amplifying immunity using agonistic monoclonal antibodies against costimulatory molecules or immune-potentiating cytokines, and 3) eliminating mechanisms that self-regulate the strength of the immune response, such as inhibitory receptors or regulatory T cells. A rational combination of such approaches holds great hope for cumulative and synergistic effects, but there is also evidence that they can open the flood-gates for unwanted inflammatory reactions. The next decade can be envisioned as the time when the first reproducibly efficacious combination regimes for cancer immunotherapy will become available and widely used in the clinic, as clinicians learn the best strategies and try to harness their potentially damaging effects.


Asunto(s)
Inmunoconjugados , Inmunoterapia , Neoplasias/terapia , Abatacept , Animales , Antígenos CD , Antígenos de Diferenciación/metabolismo , Autoinmunidad , Antígenos CD40/metabolismo , Antígeno CTLA-4 , Células Dendríticas/inmunología , Humanos , Inmunoterapia/efectos adversos , Receptores de Interleucina-2/metabolismo , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...