Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunity ; 49(3): 490-503.e4, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30170810

RESUMEN

The NF-κB pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF-κB pathway in radiation-induced anti-tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF-κB pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non-canonical NF-κB deficiency promoted IR-induced anti-tumor immunity. Mechanistic studies revealed that non-canonical NF-κB signaling in dendritic cells (DCs) was activated by the STING sensor-dependent DNA-sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non-canonical NF-κB pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non-canonical NF-κB pathway enhanced the anti-tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non-canonical NF-κB pathways in IR-induced STING-IFN production and provide an alternative strategy to improve cancer radiotherapy.


Asunto(s)
Neoplasias del Colon/radioterapia , Células Dendríticas/inmunología , Melanoma/radioterapia , FN-kappa B/metabolismo , Neoplasias Experimentales/radioterapia , Radioterapia/métodos , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Neoplasias del Colon/inmunología , ADN/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular , Melanoma/inmunología , Melanoma Experimental , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Tolerancia a Radiación , Radiación Ionizante , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Immunity ; 41(5): 843-52, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25517616

RESUMEN

Ionizing radiation-mediated tumor regression depends on type I interferon (IFN) and the adaptive immune response, but several pathways control I IFN induction. Here, we demonstrate that adaptor protein STING, but not MyD88, is required for type I IFN-dependent antitumor effects of radiation. In dendritic cells (DCs), STING was required for IFN-? induction in response to irradiated-tumor cells. The cytosolic DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS) mediated sensing of irradiated-tumor cells in DCs. Moreover, STING was essential for radiation-induced adaptive immune responses, which relied on type I IFN signaling on DCs. Exogenous IFN-? treatment rescued the cross-priming by cGAS or STING-deficient DCs. Accordingly, activation of STING by a second messenger cGAMP administration enhanced antitumor immunity induced by radiation. Thus radiation-mediated antitumor immunity in immunogenic tumors requires a functional cytosolic DNA-sensing pathway and suggests that cGAMP treatment might provide a new strategy to improve radiotherapy.


Asunto(s)
ADN/inmunología , Proteínas de la Membrana/genética , Neoplasias/radioterapia , Nucleotidiltransferasas/inmunología , Inmunidad Adaptativa , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Antineoplásicos/farmacología , Células Cultivadas , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Interferón beta/biosíntesis , Interferón beta/inmunología , Interferón beta/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Neoplasias/inmunología , Nucleótidos Cíclicos/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Radiación Ionizante , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Xantonas/farmacología
3.
Mol Ther ; 20(5): 1046-55, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22334019

RESUMEN

Radiotherapy offers an effective treatment for advanced cancer but local and distant failures remain a significant challenge. Here, we treated melanoma and pancreatic carcinoma in syngeneic mice with ionizing radiation (IR) combined with the poly(ADP-ribose) polymerase inhibitor (PARPi) veliparib to inhibit DNA repair and promote accelerated senescence. Based on prior work implicating cytotoxic T lymphocytes (CTLs) as key mediators of radiation effects, we discovered that senescent tumor cells induced by radiation and veliparib express immunostimulatory cytokines to activate CTLs that mediate an effective antitumor response. When these senescent tumor cells were injected into tumor-bearing mice, an antitumor CTL response was induced which potentiated the effects of radiation, resulting in elimination of established tumors. Applied to human cancers, radiation-inducible immunotherapy may enhance radiotherapy responses to prevent local recurrence and distant metastasis.


Asunto(s)
Bencimidazoles/farmacología , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Pancreáticas/terapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Vacunas contra el Cáncer/inmunología , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Terapia Combinada , Citocinas/biosíntesis , Citocinas/inmunología , Citotoxicidad Inmunológica , Femenino , Humanos , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Trasplante de Neoplasias , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas
4.
Am J Respir Cell Mol Biol ; 44(3): 415-22, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20508068

RESUMEN

Novel therapies are desperately needed for radiation-induced lung injury (RILI), which, despite aggressive corticosteroid therapy, remains a potentially fatal and dose-limiting complication of thoracic radiotherapy. We assessed the utility of simvastatin, an anti-inflammatory and lung barrier-protective agent, in a dose- and time-dependent murine model of RILI (18-(25 Gy). Simvastatin reduced multiple RILI indices, including vascular leak, leukocyte infiltration, and histological evidence of oxidative stress, while reversing RILI-associated dysregulated gene expression, including p53, nuclear factor-erythroid-2-related factor, and sphingolipid metabolic pathway genes. To identify key regulators of simvastatin-mediated RILI protection, we integrated whole-lung gene expression data obtained from radiated and simvastatin-treated mice with protein-protein interaction network analysis (single-network analysis of proteins). Topological analysis of the gene product interaction network identified eight top-prioritized genes (Ccna2a, Cdc2, fcer1 g, Syk, Vav3, Mmp9, Itgam, Cd44) as regulatory nodes within an activated RILI network. These studies identify the involvement of specific genes and gene networks in RILI pathobiology, and confirm that statins represent a novel strategy to limit RILI.


Asunto(s)
Regulación de la Expresión Génica , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lesión Pulmonar/metabolismo , Pulmón/metabolismo , Pulmón/efectos de la radiación , Traumatismos por Radiación/tratamiento farmacológico , Simvastatina/farmacología , Animales , Lavado Broncoalveolar , Receptores de Hialuranos/biosíntesis , Lesión Pulmonar/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Mapeo de Interacción de Proteínas , Neumonitis por Radiación , Transcripción Genética
5.
Mol Ther ; 18(5): 912-20, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20197756

RESUMEN

Ad.Egr-TNF is a radioinducible adenovector currently in phase 3 trials for inoperable pancreatic cancer. The combination of Ad.Egr-TNF and ionizing radiation (IR) contributes to local tumor control through the production of tumor necrosis factor-alpha (TNFalpha) in the tumor microenvironment. Moreover, clinical and preclinical studies with Ad.Egr-TNF/IR have suggested that this local approach suppresses the growth of distant metastatic disease; however, the mechanisms responsible for this effect remain unclear. These studies have been performed in wild-type (WT) and TNFR1,2(-/-) mice to assess the role of TNFalpha-induced signaling in the suppression of draining lymph node (DLN) metastases. The results demonstrate that production of TNFalpha in the tumor microenvironment induces expression of interferon (IFNbeta). In turn, IFNbeta stimulates the production of chemokines that recruit CD8(+) T cells to the tumor. The results further demonstrate that activation of tumor antigen-specific CD8(+) CTLs contributes to local antitumor activity and suppression of DLN metastases. These findings support a model in which treatment of tumors with Ad.Egr-TNF and IR is mediated by local and distant immune-mediated antitumor effects that suppress the development of metastases.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Metástasis de la Neoplasia/prevención & control , Metástasis de la Neoplasia/terapia , Radiación Ionizante , Factor de Necrosis Tumoral alfa/metabolismo , Adenoviridae/genética , Animales , Proliferación Celular , Vectores Genéticos/genética , Humanos , Interferón beta/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Metástasis de la Neoplasia/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/genética
6.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1041-52, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19833862

RESUMEN

Intestinal injury following abdominal radiation therapy or accidental exposure remains a significant clinical problem that can result in varying degrees of mucosal destruction such as ulceration, vascular sclerosis, intestinal wall fibrosis, loss of barrier function, and even lethal gut-derived sepsis. We determined the ability of a high-molecular-weight polyethylene glycol-based copolymer, PEG 15-20, to protect the intestine against the early and late effects of radiation in mice and rats and to determine its mechanism of action by examining cultured rat intestinal epithelia. Rats were exposed to fractionated radiation in an established model of intestinal injury, whereby an intestinal segment is surgically placed into the scrotum and radiated daily. Radiation injury score was decreased in a dose-dependent manner in rats gavaged with 0.5 or 2.0 g/kg per day of PEG 15-20 (n = 9-13/group, P < 0.005). Complementary studies were performed in a novel mouse model of abdominal radiation followed by intestinal inoculation with Pseudomonas aeruginosa (P. aeruginosa), a common pathogen that causes lethal gut-derived sepsis following radiation. Mice mortality was decreased by 40% in mice drinking 1% PEG 15-20 (n = 10/group, P < 0.001). Parallel studies were performed in cultured rat intestinal epithelial cells treated with PEG 15-20 before radiation. Results demonstrated that PEG 15-20 prevented radiation-induced intestinal injury in rats, prevented apoptosis and lethal sepsis attributable to P. aeruginosa in mice, and protected cultured intestinal epithelial cells from apoptosis and microbial adherence and possible invasion. PEG 15-20 appeared to exert its protective effect via its binding to lipid rafts by preventing their coalescence, a hallmark feature in intestinal epithelial cells exposed to radiation.


Asunto(s)
Íleon/efectos de los fármacos , Enfermedades Intestinales/prevención & control , Mucosa Intestinal/efectos de los fármacos , Microdominios de Membrana/efectos de los fármacos , Polietilenglicoles/administración & dosificación , Pseudomonas aeruginosa/efectos de los fármacos , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/administración & dosificación , Sepsis/prevención & control , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Adhesión Bacteriana/efectos de los fármacos , Línea Celular , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Íleon/microbiología , Íleon/patología , Íleon/efectos de la radiación , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Microdominios de Membrana/metabolismo , Microdominios de Membrana/microbiología , Microdominios de Membrana/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Pseudomonas aeruginosa/patogenicidad , Traumatismos Experimentales por Radiación/microbiología , Traumatismos Experimentales por Radiación/patología , Ratas , Ratas Sprague-Dawley , Sepsis/microbiología , Factores de Tiempo , Virulencia/efectos de los fármacos
7.
Magn Reson Med ; 62(2): 348-56, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19449382

RESUMEN

Imaging techniques are under development to facilitate early analysis of spatial patterns of tumor response to combined radiation and antivascular gene therapy. A genetically modified, replication defective adenoviral vector (Ad.EGR-TNFalpha), injected intratumorally, mediates infected cells to express tumor necrosis factor alpha (TNFalpha), which is increased after exposure to radiation. The goal of this study was to characterize an image based "signature" for response to this combined radiation and gene therapy in mice with human prostate xenografts. This study is part of an imaged guided therapy project where such a signature would be useful in guiding subsequent treatments. Changes in the tumor micro-environment were assessed using MRI registered with electron paramagnetic resonance imaging which provides images of tissue oxygenation. Dynamic contrast-enhanced MRI was used to assess tissue perfusion. When compared with null vector (control) treatment, the ratio of contrast agent (Gd-DTPA-BMA) washout rate to uptake rate was lower (P = 0.001) after treatment, suggesting a more balanced perfusion. Concomitantly, oxygenation significantly increased in the treated animals and decreased or did not change in the control animals (P < 0.025). This is the first report of minimally invasive, quantitative, absolute oxygen measurements correlated with tissue perfusion in vivo.


Asunto(s)
Terapia Genética/métodos , Imagen por Resonancia Magnética/métodos , Imagen de Perfusión/métodos , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Radioterapia Conformacional/métodos , Técnica de Sustracción , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Pronóstico , Resultado del Tratamiento
8.
Cancer Res ; 67(19): 9214-20, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17909027

RESUMEN

Elsewhere, we reported that multiple serial in vivo passage of a squamous cell carcinoma cells (SCC61) concurrent with ionizing radiation (IR) treatment resulted in the selection of radioresistant tumor (nu61) that overexpresses the signal transducer and activator of transcription 1 (Stat1)/IFN-dependent pathway. Here, we report that (a) the Stat1 pathway is induced by IR, (b) constitutive overexpression of Stat1 is linked with failure to transmit a cytotoxic signal by radiation or IFNs, (c) selection of parental cell line SCC61 against IFN-alpha and IFN-gamma leads to the same IR- and IFN-resistant phenotype as was obtained by IR selection, and (d) suppression of Stat1 by short hairpin RNA renders the IR-resistant nu61 cells radiosensitive to IR. We propose a model that transient induction of Stat1 by IFN, IR, or other stress signals activates cytotoxic genes and cytotoxic response. Constitutive overexpression of Stat1 on the other hand leads to the suppression of the cytotoxic response and induces prosurvival genes that, at high levels of Stat1, render the cells resistant to IR or other inducers of cell death.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Resistencia a Antineoplásicos , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Humanos , Interferón-alfa/farmacología , Interferón gamma/farmacología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , ARN Interferente Pequeño/genética , Tolerancia a Radiación , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/genética , Trasplante Heterólogo
9.
Nat Commun ; 10(1): 3959, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477729

RESUMEN

Successful combinations of radiotherapy and immunotherapy depend on the presence of live T cells within the tumor; however, radiotherapy is believed to damage T cells. Here, based on longitudinal in vivo imaging and functional analysis, we report that a large proportion of T cells survive clinically relevant doses of radiation and show increased motility, and higher production of interferon gamma, compared with T cells from unirradiated tumors. Irradiated intratumoral T cells can mediate tumor control without newly-infiltrating T cells. Transcriptomic analysis suggests T cell reprogramming in the tumor microenvironment and similarities with tissue-resident memory T cells, which are more radio-resistant than circulating/lymphoid tissue T cells. TGFß is a key upstream regulator of T cell reprogramming and contributes to intratumoral Tcell radio-resistance. These findings have implications for the design of radio-immunotherapy trials in that local irradiation is not inherently immunosuppressive, and irradiation of multiple tumors might optimize systemic effects of radiotherapy.


Asunto(s)
Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias Experimentales/terapia , Radioterapia/métodos , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Supervivencia Celular/genética , Terapia Combinada , Perfilación de la Expresión Génica/métodos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Tolerancia a Radiación/genética
10.
Mol Cancer Ther ; 17(2): 407-418, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29030460

RESUMEN

Despite significant advances in combinations of radiotherapy and chemotherapy, altered fractionation schedules and image-guided radiotherapy, many cancer patients fail to benefit from radiation. A prevailing hypothesis is that targeting repair of DNA double strand breaks (DSB) can enhance radiation effects in the tumor and overcome therapeutic resistance without incurring off-target toxicities. Unrepaired DSBs can block cancer cell proliferation, promote cancer cell death, and induce cellular senescence. Given the slow progress to date translating novel DSB repair inhibitors as radiosensitizers, we have explored drug repurposing, a proven route to improving speed, costs, and success rates of drug development. In a prior screen where we tracked resolution of ionizing radiation-induced foci (IRIF) as a proxy for DSB repair, we had identified pitavastatin (Livalo), an HMG-CoA reductase inhibitor commonly used for lipid lowering, as a candidate radiosensitizer. Here, we report that pitavastatin and other lipophilic statins are potent inhibitors of DSB repair in breast and melanoma models both in vitro and in vivo When combined with ionizing radiation, pitavastatin increased persistent DSBs, induced senescence, and enhanced acute effects of radiation on radioresistant melanoma tumors. shRNA knockdown implicated HMG-CoA reductase, farnesyl diphosphate synthase, and protein farnesyl transferase in IRIF resolution, DSB repair, and senescence. These data confirm on-target activity of statins, although via inhibition of protein prenylation rather than cholesterol biosynthesis. In light of prior studies demonstrating enhanced efficacy of radiotherapy in patients taking statins, this work argues for clinical evaluation of lipophilic statins as nontoxic radiosensitizers to enhance the benefits of image-guided radiotherapy. Mol Cancer Ther; 17(2); 407-18. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."


Asunto(s)
Reparación del ADN/efectos de los fármacos , Acilcoenzima A/farmacología , Animales , Senescencia Celular , Femenino , Humanos , Ratones
11.
J Clin Invest ; 110(3): 403-10, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12163460

RESUMEN

Ionizing radiation (IR) and radical oxygen intermediates (ROIs) activate the early growth response-1 (Egr1) promoter through specific cis-acting sequences termed CArG elements. Ad.Egr.TNF.11D, a replication-deficient adenoviral vector containing CArG elements cloned upstream of the cDNA for human recombinant TNF-alpha was used to treat human esophageal adenocarcinoma and rat colon adenocarcinoma cells in culture and as xenografts in athymic nude mice. Cisplatin, a commonly used chemotherapeutic agent, causes tumor cell death by producing DNA damage and generating ROIs. The present studies demonstrate induction of TNF-alpha production in tumor cells and xenografts treated with the combination of Ad.Egr.TNF.11D and cisplatin. The results show that the Egr1 promoter is induced by cisplatin and that this induction is mediated in part through the CArG elements. These studies also demonstrate an enhanced antitumor response without an increase in toxicity following treatment with Ad.Egr.TNF.11D and cisplatin, compared with either agent alone. Chemo-inducible cancer gene therapy thus provides a means to control transgene expression while enhancing the effectiveness of commonly used chemotherapeutic agents.


Asunto(s)
Cisplatino/farmacología , Proteínas Inmediatas-Precoces , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Adenocarcinoma/terapia , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Neoplasias del Colon , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz , Neoplasias Esofágicas/terapia , Femenino , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Ratas , Factores de Transcripción/genética , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis
12.
Clin Exp Metastasis ; 24(7): 521-31, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17653822

RESUMEN

TNFerade is a replication incompetent adenovector designed to express human TNFalpha under control of the Egr-1 radiation and chemotherapy enhanced promoter, and is currently in Phase II/III clinical testing. Data from Phase I clinical testing of TNFerade in a limited set of melanoma patients suggested the potential to impact distal metastases following intratumoral injections of TNFerade. These clinical observations and the multiple potential mechanisms of TNFerade led us to hypothesize local treatment with TNFerade + radiation may impact metastatic disease. We explored this hypothesis in preclinical models using the spontaneously metastatic, syngeneic B16F10 murine melanoma model. Established subcutaneous B16F10 tumors were treated with intratumoral injections of TNFerade and localized 2 Gy fractionated radiation therapy, modeling the clinical treatment regimen. Following 10-14 days of treatment, mice were evaluated for metastases development in the iliac and axillary lymph nodes. Comparisons of metastatic burden to control groups indicated TNFerade +/- radiation suppressed the formation of metastases in the lymph nodes. Additional experiments in TNF receptor knockout mice, where the only possible effects are on tumor cells containing the TNFalpha receptor, indicate TNFerade's local and distal activities are critically dependent on a host-mediated response. These data provide direct preclinical evidence local therapy of a solid tumor with TNFerade can also reduce metastatic disease, in addition to effects on the treated lesion. Furthermore, our finding of a host dependant response(s) for TNFerade at both the treated tumor and on lymph node metastases suggest the potential for broad activity independent of tumor histology.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Melanoma/terapia , Neoplasias/terapia , Factor de Necrosis Tumoral alfa/genética , Adenoviridae/genética , Adulto , Animales , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Femenino , Estudios de Seguimiento , Vectores Genéticos , Humanos , Metástasis Linfática/prevención & control , Masculino , Melanoma/inmunología , Melanoma/mortalidad , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Trasplante de Neoplasias , Receptores del Factor de Necrosis Tumoral/genética , Células Tumorales Cultivadas
13.
Nat Commun ; 8(1): 1736, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29170400

RESUMEN

Radiotherapy induces and promotes innate and adaptive immunity in which host STING plays an important role. However, radioresistance in irradiated tumors can also develop, resulting in relapse. Here we report a mechanism by which extrinsic resistance develops after local ablative radiation that relies on the immunosuppressive action of STING. The STING/type I interferon pathway enhances suppressive inflammation in tumors by recruiting myeloid cells in part via the CCR2 pathway. Germ-line knockouts of CCR2 or treatment with an anti-CCR2 antibody results in blockade of radiation-induced MDSC infiltration. Treatment with anti-CCR2 antibody alleviates immunosuppression following activation of the STING pathway, enhancing the anti-tumor effects of STING agonists and radiotherapy. We propose that radiation-induced STING activation is immunosuppressive due to (monocytic) M-MDSC infiltration, which results in tumor radioresistance. Furthermore, the immunosuppressive effects of radiotherapy and STING agonists can be abrogated in humans by a translational strategy involving anti-CCR2 antibody treatment to improve radiotherapy.


Asunto(s)
Proteínas de la Membrana/inmunología , Células Supresoras de Origen Mieloide/inmunología , Tolerancia a Radiación/inmunología , Inmunidad Adaptativa , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/radioterapia , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/inmunología , Transducción de Señal
14.
Cancer Res ; 63(2): 308-11, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12543780

RESUMEN

Ionizing radiation (IR) and concomitant angiostatin (AS) produce greater than additive local antitumor effects. We examined whether prolonged AS treatment added to IR reduces proliferation of lung metastases from LLC primary tumors. Flank tumors were treated with 40 Gy with or without AS (25 mg/kg/day). IR plus a 14-day course of AS improved local tumor control and blocked the increase in lung weights observed in the group receiving IR plus a 2-day course of AS group. Animals treated with prolonged AS exhibited no increase in lung weight and no macrometastases. These findings suggest that long-term treatment with antiangiogenic compounds may be effective in preventing metastases from IR-treated tumors as well as increasing the local antitumor effects of radiotherapy.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/radioterapia , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Fragmentos de Péptidos/farmacología , Plasminógeno/farmacología , Angiostatinas , Animales , Carcinoma Pulmonar de Lewis/secundario , Terapia Combinada , Femenino , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Ratones , Ratones Endogámicos C57BL
15.
Oncotarget ; 7(23): 33919-33, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27129153

RESUMEN

Radiation therapy remains a significant therapeutic modality in the treatment of cancer. An attractive strategy would be to enhance the benefits of ionizing radiation (IR)with radiosensitizers. A high-content drug repurposing screen of approved and investigational agents, natural products and other small molecules has identified multiple candidates that blocked repair of IR damage in vitro. Here, we validated a subset of these hits in vitro and then examined effects on tumor growth after IR in a murine tumor model. Based on robust radiosensitization in vivo and other favorable properties of cephalexin, we conducted additional studies with other beta-lactam antibiotics. When combined with IR, each cephalosporin tested increased DNA damage and slowed tumor growth without affecting normal tissue toxicity. Our data implicate reactive oxygen species in the mechanism by which cephalosporins augment the effects of IR. This work provides a rationale for using commonly prescribed beta-lactam antibiotics as non-toxic radiosensitizers to enhance the therapeutic ratio of radiotherapy.


Asunto(s)
Antibacterianos/farmacología , Neoplasias de la Mama/radioterapia , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Cefalosporinas/farmacología , Reposicionamiento de Medicamentos , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Células MCF-7 , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Chemother Pharmacol ; 56(3): 317-21, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15887016

RESUMEN

We examined the interaction between forphenicinol (FPL) and cyclophosphamide (CPA) or ionizing radiation (IR) on the growth of murine squamous cell carcinoma tumors SCCVII. Primary tumors were established in C3H mice by injecting SCCVII tumor cells subcutaneously into the right hind limb. FPL (100 mg/kg for 8 days) and/or CPA (25 mg/kg twice) were administered by intraperitoneal injection. Tumors were irradiated to a total dose of 40 Gy (eight 5-Gy fractions). SCCVII tumor growth was inhibited by FPL (P=0.054), IR (P=0.003) and CPA (P<0.001) compared with control. The combination of FPL and CPA inhibited tumor growth additively compared with either treatment alone in both small- and large-volume tumors. FPL did not significantly enhance the antitumor effects of IR, however, when CPA+FPL were combined with IR, significant tumor growth inhibition was observed compared with FPL alone (P<0.001), CPA alone (P=0.002) and IR alone (P=0.002). Due to its low toxicity profile, FPL may be combined with CPA, IR and other cytotoxic therapies to potentially enhance the therapeutic ratio.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Ciclofosfamida/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Animales , Carcinoma de Células Escamosas/radioterapia , Terapia Combinada , Sinergismo Farmacológico , Femenino , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Radioterapia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Mol Cancer Ther ; 3(9): 1167-75, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15367711

RESUMEN

A replication-defective adenoviral vector, Ad.Egr-TNF.11D, was engineered by ligating the CArG (CC(A/T)6GG) elements of the Egr-1 gene promoter upstream to a cDNA encoding human tumor necrosis factor-alpha. We report here that Ad.Egr-TNF.11D is activated by the clinically important anticancer agents cisplatin, cyclophosphamide, doxorubicin, 5-fluorouracil, gemcitabine, and paclitaxel. N-acetylcysteine, a free radical scavenger, blocked induction of tumor necrosis factor-alpha by anticancer agents, supporting a role for reactive oxygen intermediates in activation of the CArG sequences. Importantly, resistance of PC-3 human prostate carcinoma and PROb rat colon carcinoma tumors to doxorubicin in vivo was reversed by combining doxorubicin with Ad.Egr-TNF and resulted in significant antitumor effects. Treatment with Ad.Egr-TNF.11D has been associated with inhibition of tumor angiogenesis. In this context, a significant decrease in tumor microvessel density was observed following combined treatment with doxorubicin and Ad.Egr-TNF.11D as compared with either agent alone. These data show that Ad.Egr-TNF.11D is activated by diverse anticancer drugs.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Terapia Genética , Neoplasias/terapia , Factor de Necrosis Tumoral alfa/genética , Acetilcisteína/farmacología , Adenoviridae/genética , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Proteínas de Unión al ADN/genética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Proteína 1 de la Respuesta de Crecimiento Precoz , Terapia Genética/métodos , Vectores Genéticos , Humanos , Proteínas Inmediatas-Precoces/genética , Masculino , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/terapia , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Ratas , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
FEBS Lett ; 565(1-3): 167-70, 2004 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-15135073

RESUMEN

We demonstrate that human umbilical vein endothelial cells (HUVEC) grown in co-culture (CC) with U87 glioblastoma cells transfected with green fluorescent protein (GFP-U87) exhibit resistance to radiation-mediated apoptosis. cDNA macroarray analysis reveals increases in the accumulation of RNAs for HUVEC genes encoding cell adhesion molecules, growth factor-related proteins, and cell cycle regulatory/DNA repair proteins. An increase in protein expression of integrin alphav, integrin beta1, MAPK(p42), Rad51, DNA-PK(CS), and ataxia telangiectasia gene (ATM) was detected in HUVEC grown in CC with GFP-U87 cells compared with HUVEC grown in mono-culture. Treatment with anti-VEGF antibody decreases the expression of integrin alphav, integrin beta1, DNA-PK(CS) and ATM with a corresponding increase in ionizing radiation (IR)-induced apoptosis. These data support the concept that endothelial cells growing in the tumor microenvironment may develop resistance to cytotoxic therapies due to the up-regulation by tumor cells of endothelial cells genes associated with survival.


Asunto(s)
Apoptosis , Células Endoteliales/patología , Glioblastoma/patología , Proteínas de la Ataxia Telangiectasia Mutada , Western Blotting , Adhesión Celular , Ciclo Celular , Proteínas de Ciclo Celular , División Celular , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Reparación del ADN , ADN Complementario/metabolismo , Proteína Quinasa Activada por ADN , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/efectos de la radiación , Endotelio Vascular/citología , Glioblastoma/radioterapia , Proteínas Fluorescentes Verdes , Humanos , Rayos Infrarrojos , Integrina alfaV/biosíntesis , Integrina alfaVbeta3/metabolismo , Integrina beta1/biosíntesis , Proteínas Luminiscentes/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Nucleares , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/metabolismo , Recombinasa Rad51 , Radioterapia , Recombinación Genética , Transcripción Genética , Transfección , Proteínas Supresoras de Tumor , Venas Umbilicales/citología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Int J Oncol ; 24(3): 731-6, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14767560

RESUMEN

The promising benefits of cancer gene therapy have been limited by the inability to deliver therapeutic genes homogeneously throughout the tumor mass and to control gene expression within the tumor cells. Transcriptional targeting, the use of DNA regulatory promoter sequences to localize transgene expression, has been employed as a solution to circumvent these limitations. TNF-alpha is a cytokine that exhibits potent anticancer properties, but its utility following systemic administration is limited by toxicity. We review a strategy whereby ligating TNF-alpha to segments of the chemo-inducible EGR1 or MDR1 promoters activates expression of TNF-alpha cDNA and enhances effectiveness of gene therapy and chemotherapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Terapia Genética/métodos , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Transcripción Genética , Animales , Ensayos Clínicos como Asunto , Citotoxinas , ADN/metabolismo , Daño del ADN , Proteína 1 de la Respuesta de Crecimiento Precoz , Humanos , Modelos Biológicos , Radiación Ionizante , Factor de Necrosis Tumoral alfa/metabolismo
20.
Cancer Chemother Pharmacol ; 50(5): 412-8, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12439600

RESUMEN

PURPOSE: We examined the interaction between cyclophosphamide (CPA) and angiostatin (AS) on the growth of primary Lewis lung carcinoma (LLC) tumors and on the development of LLC pulmonary metastases. We studied the effects of AS and CPA on the stages of angiogenesis employing in vitro assays. METHODS: Primary tumor growth and pulmonary metastases were measured to evaluate the effects of treatment with AS alone, CPA alone or the combination of CPA and AS. We examined the effects of CPA plus AS on endothelial cell (HUVEC) survival, migration and tube formation. RESULTS: Combined treatment with CPA and AS did not significantly affect primary tumor growth when compared with CPA treatment alone. However, a significant decrease in the number of pulmonary metastases was observed following CPA plus AS treatment when compared with CPA treatment alone ( P<0.001). AS did not enhance CPA-mediated HUVEC cytotoxicity, and CPA failed to enhance AS-mediated inhibition of migration. However, tube formation was inhibited following combined treatment with CPA and AS when compared with either treatment alone. CONCLUSIONS: AS enhanced the antimetastatic effects of CPA without significantly influencing the effects of CPA on primary tumor growth. CPA plus AS inhibited tube formation, suggesting that interrupting specific steps in the angiogenesis process might be an effective approach to the treatment of subclinical distant metastases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacología , Metástasis de la Neoplasia/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Plasminógeno/farmacología , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Angiostatinas , Animales , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/secundario , Movimiento Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Ciclofosfamida/administración & dosificación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Modelos Lineales , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Plasminógeno/administración & dosificación , Inhibidores de la Síntesis de la Proteína/administración & dosificación , Inhibidores de la Síntesis de la Proteína/farmacología , Método Simple Ciego , Células Tumorales Cultivadas/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA