Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Immunity ; 51(3): 548-560.e4, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31471106

RESUMEN

Immunotherapy can reinvigorate dormant responses to cancer, but response rates remain low. Oncolytic viruses, which replicate in cancer cells, induce tumor lysis and immune priming, but their immune consequences are unclear. We profiled the infiltrate of aggressive melanomas induced by oncolytic Vaccinia virus using RNA sequencing and found substantial remodeling of the tumor microenvironment, dominated by effector T cell influx. However, responses to oncolytic viruses were incomplete due to metabolic insufficiencies induced by the tumor microenvironment. We identified the adipokine leptin as a potent metabolic reprogramming agent that supported antitumor responses. Leptin metabolically reprogrammed T cells in vitro, and melanoma cells expressing leptin were immunologically controlled in mice. Engineering oncolytic viruses to express leptin in tumor cells induced complete responses in tumor-bearing mice and supported memory development in the tumor infiltrate. Thus, leptin can provide metabolic support to tumor immunity, and oncolytic viruses represent a platform to deliver metabolic therapy.


Asunto(s)
Leptina/inmunología , Melanoma/inmunología , Virus Oncolíticos/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Inmunoterapia/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral/inmunología , Virus Vaccinia/inmunología
2.
Immunity ; 50(1): 51-63.e5, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635239

RESUMEN

Interferon-inducible human oligoadenylate synthetase-like (OASL) and its mouse ortholog, Oasl2, enhance RNA-sensor RIG-I-mediated type I interferon (IFN) induction and inhibit RNA virus replication. Here, we show that OASL and Oasl2 have the opposite effect in the context of DNA virus infection. In Oasl2-/- mice and OASL-deficient human cells, DNA viruses such as vaccinia, herpes simplex, and adenovirus induced increased IFN production, which resulted in reduced virus replication and pathology. Correspondingly, ectopic expression of OASL in human cells inhibited IFN induction through the cGAS-STING DNA-sensing pathway. cGAS was necessary for the reduced DNA virus replication observed in OASL-deficient cells. OASL directly and specifically bound to cGAS independently of double-stranded DNA, resulting in a non-competitive inhibition of the second messenger cyclic GMP-AMP production. Our findings define distinct mechanisms by which OASL differentially regulates host IFN responses during RNA and DNA virus infection and identify OASL as a negative-feedback regulator of cGAS.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Infecciones por Virus ADN/inmunología , Virus ADN/fisiología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , 2',5'-Oligoadenilato Sintetasa/genética , Animales , AMP Cíclico/metabolismo , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nucleotidiltransferasas/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Células THP-1 , Replicación Viral
3.
Breast Cancer Res Treat ; 157(1): 41-54, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27097807

RESUMEN

The present study offers novel insights into the molecular circuitry of accelerated in vivo tumor growth by Notch2 knockdown in triple-negative breast cancer (TNBC) cells. Therapeutic vulnerability of Notch2-altered growth to a small molecule (withaferin A, WA) is also demonstrated. MDA-MB-231 and SUM159 cells were used for the xenograft studies. A variety of technologies were deployed to elucidate the mechanisms underlying tumor growth augmentation by Notch2 knockdown and its reversal by WA, including Fluorescence Molecular Tomography for measurement of tumor angiogenesis in live mice, Seahorse Flux analyzer for ex vivo measurement of tumor metabolism, proteomics, and Luminex-based cytokine profiling. Stable knockdown of Notch2 resulted in accelerated in vivo tumor growth in both cells reflected by tumor volume and/or latency. For example, the wet tumor weight from mice bearing Notch2 knockdown MDA-MB-231 cells was about 7.1-fold higher compared with control (P < 0.0001). Accelerated tumor growth by Notch2 knockdown was highly sensitive to inhibition by a promising steroidal lactone (WA) derived from a medicinal plant. Molecular underpinnings for tumor growth intensification by Notch2 knockdown included compensatory increase in Notch1 activation, increased cellular proliferation and/or angiogenesis, and increased plasma or tumor levels of growth stimulatory cytokines. WA administration reversed many of these effects providing explanation for its remarkable anti-cancer efficacy. Notch2 functions as a tumor growth suppressor in TNBC and WA offers a novel therapeutic strategy for restoring this function.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Receptor Notch2/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Witanólidos/administración & dosificación , Animales , Antineoplásicos Fitogénicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Receptor Notch1/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Witanólidos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Ther ; 23(1): 202-14, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25292189

RESUMEN

Oncolytic viral therapy utilizes a tumor-selective replicating virus which preferentially infects and destroys cancer cells and triggers antitumor immunity. The Western Reserve strain of vaccinia virus (VV) is the most virulent strain of VV in animal models and has been engineered for tumor selectivity through two targeted gene deletions (vvDD). We performed the first-in-human phase 1, intratumoral dose escalation clinical trial of vvDD in 16 patients with advanced solid tumors. In addition to safety, we evaluated signs of vvDD replication and spread to distant tumors, pharmacokinetics and pharmacodynamics, clinical and immune responses to vvDD. Dose escalation proceeded without dose-limiting toxicities to a maximum feasible dose of 3 × 10(9) pfu. vvDD replication in tumors was reproducible. vvDD genomes and/or infectious particles were recovered from injected (n = 5 patients) and noninjected (n = 2 patients) tumors. At the two highest doses, vvDD genomes were detected acutely in blood in all patients while delayed re-emergence of vvDD genomes in blood was detected in two patients. Fifteen of 16 patients exhibited late symptoms, consistent with ongoing vvDD replication. In summary, intratumoral injection of the oncolytic vaccinia vvDD was well-tolerated in patients and resulted in selective infection of injected and noninjected tumors and antitumor activity.


Asunto(s)
Neoplasias de la Mama/terapia , Neoplasias del Colon/terapia , Melanoma/terapia , Neoplasias Pancreáticas/terapia , Neoplasias Cutáneas/terapia , Virus Vaccinia/inmunología , Replicación Viral/genética , Anciano , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Relación Dosis-Respuesta Inmunológica , Femenino , Eliminación de Gen , Humanos , Inyecciones Intralesiones , Masculino , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Virus Vaccinia/genética , Virus Vaccinia/crecimiento & desarrollo
5.
Int J Cancer ; 135(5): 1238-46, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24474587

RESUMEN

Oncolytic vaccinia virus has been shown to induce a profound, rapid and tumor-specific vascular collapse in both preclinical models and clinical studies; however, a complete examination of the kinetics and levels of collapse and revascularization has not been described previously. Contrast-enhanced ultrasound was used to follow tumor perfusion levels in mouse tumor models at times after vaccinia therapy. It was observed that revascularization after viral therapy was dramatically delayed and did not occur until after viral clearance. This indicated that oncolytic vaccinia may possess a previously undescribed antiangiogenic potential that might synergize with the reported anti-vascular effects. Despite a rapid loss of perfusion and widespread hypoxia within the tumor, it was observed that VEGF levels in the tumor were suppressed throughout the period of active viral infection. Although tumor vasculature could eventually reform after the viral therapy was cleared in mouse models, anti-tumor effects could be significantly enhanced through additional combination with anti-VEGF therapies. This was initially examined using a gene therapy approach (Ad-Flk1-Fc) to target VEGF directly, demonstrating that the timing of application of the antiangiogenic therapy was critical. However, it is also known that oncolytic vaccinia sensitizes tumors to tyrosine kinase inhibitors (TKI) in the clinic through an unknown mechanism. It is possible this phenomenon may be mediated through the antiangiogenic effects of the TKIs. This was modeled in mouse tumors using sunitinib in combination with oncolytic vaccinia. It was observed that prevention of angiogenesis mediated by oncolytic vaccinia can be utilized to enhance the TKI therapy.


Asunto(s)
Neoplasias/terapia , Neovascularización Patológica/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Virus Vaccinia , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/uso terapéutico , Terapia Genética , Humanos , Indoles/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirroles/uso terapéutico , Sunitinib , Factor A de Crecimiento Endotelial Vascular/genética
6.
Mol Ther ; 21(1): 167-74, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23281444

RESUMEN

Genetic therapies, including transfected immune cells and viral vectors, continue to show clinical responses as systemically deliverable and targeted therapeutics, with the first such approaches having been approved for cancer treatment. The majority of these employ cytokine transgenes. However, expression of cytokines early after systemic delivery can result in increased toxicity and nonspecific induction of the immune response. In addition, premature immune-mediated clearance of the therapy may result, especially for viral-based approaches. Here, it was initially verified that cytokine (interleukin (IL)2) or chemokine (CCL5) expression from a systemically delivered oncolytic virus resulted in reduced oncolytic activity and suboptimal immune activation, while IL2 also resulted in increased toxicity. However, all these limitations could be overcome through incorporation of exogenous regulation of cytokine or chemokine transgene function through fusion of a small and externally controllable destabilizing domain to the protein of interest. Regulation allowed an initial phase without cytokine function, permitting enhanced delivery and oncolytic activity before activation of cytokine function and a subsequent phase of enhanced and tumor-targeted immunotherapeutic activity. As a result of this exogenous regulation of cytokine function, both oncolytic and immune-mediated mechanisms of action were optimized, greatly enhancing therapeutic activity, while toxicity was significantly reduced.


Asunto(s)
Citocinas/fisiología , Terapia Genética , Neoplasias/terapia , Animales , Citocinas/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Viroterapia Oncolítica , Transgenes , Virus Vaccinia/metabolismo , Virus Vaccinia/fisiología , Replicación Viral
7.
Zhonghua Zhong Liu Za Zhi ; 36(11): 828-33, 2014 Nov.
Artículo en Zh | MEDLINE | ID: mdl-25620479

RESUMEN

OBJECTIVE: To investigate the expression and significance of CCL5 in patients with esophageal carcinoma. METHODS: Using reverse transcriptase polymerase chain reaction (RT-PCR), the expressions of CCL5/CD8/granzyme B/perforin in tumor and corresponding adjacent tissues from esophageal carcinoma patients were examined. Flow cytometry (FACS) was used to detect the percentages of CD8(+) T cells and CCR5(+)CD8(+) T cells in TIL and PBMC from the patients. Transwell assay was performed to study the effect of CCL5 on the migration of T cells in vitro. T test and Spearman correlation analysis were performed. RESULTS: The mRNA expressions of CCL5 and perforin were 0.348 2 ± 0.300 1 and 0.181 9 ± 0.118 6, respectively, in the tumor samples, while their expressions in adjacent samples were 0.279 6 ± 0.138 0 and 0.118 0 ± 0.109 8, respectively, with no statistically significant differences between them (P > 0.05 for both). The mRNA expressions of CD8 and granzyme B were significantly higher in the tumor tissues than in adjacent tissues (0.464 9 ± 0.300 8 vs. 0.279 0 ± 0.173 4, 0.648 7 ± 0.516 0 vs. 0.469 7 ± 0.259 1; P < 0.05 for both). The relative expression of CCL5 was positively correlated with that of CD8, perforin and granzyme B (r(CD8) = 0.272, P = 0.034; r(perforin) = 0.305, P = 0.026; r(granzymeB) = 0.108, P = 0.012) in the tumor sites. FACS data revealed that the proportions of CD8(+) T cells in TIL and PBMC were (45.86 ± 16.09)% and (34.05 ± 15.07)%, respectively, showing a significant difference (P = 0.022). Similarly, CCR5(+)CD8(+) T cells fraction in TIL (48.12 ± 26.75)% was much higher than that in PBMC (19.53 ± 13.67) % (P < 0.001). Transwell assay showed that CCL5 protein enhanced the migration of T cells, supporting that CCL5 is crucial for CD8(+) T cells recruitment in vivo. Intriguingly, CCL5 expression was down-regulated in advanced patients (stage IIb-IV). The accumulation of CD8(+) T cells and CCR5(+)CD8(+) T cells was strongly reduced in advanced patients, suggesting that CCL5 expression may be involved in the local control of the disease and its reduction may be involved in disease progression. CONCLUSIONS: The current data indicate the involvement of CCL5 in the regulation of CD8(+) T cell entry into tumor lesions in esophageal carcinoma patients. This process may affect the disease status and potentially as a prognostic factor for cancer patients. Enhancing local CCL5 expression in tumor lesions may represent a novel strategy in esophageal cancer therapy.


Asunto(s)
Quimiocina CCL5/metabolismo , Neoplasias Esofágicas/metabolismo , Linfocitos T CD8-positivos , Progresión de la Enfermedad , Citometría de Flujo , Humanos , Leucocitos Mononucleares , Linfocitos Infiltrantes de Tumor
8.
Cancer Immunol Res ; 12(6): 779-790, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38517470

RESUMEN

IFN regulatory factor 1 (IRF1) can promote antitumor immunity. However, we have shown previously that in the tumor cell, IRF1 can promote tumor growth, and IRF1-deficient tumor cells exhibit severely restricted tumor growth in several syngeneic mouse tumor models. Here, we investigate the potential of functionally modulating IRF1 to reduce tumor progression and prolong survival. Using inducible IRF1 expression, we established that it is possible to regulate IRF1 expression to modulate tumor progression in established B16-F10 tumors. Expression of IRF2, which is a functional antagonist of IRF1, downregulated IFNγ-induced expression of inhibitory ligands, upregulated MHC-related molecules, and slowed tumor growth and extended survival. We characterized the functional domain(s) of IRF2 needed for this antitumor activity, showing that a full-length IRF2 was required for its antitumor functions. Finally, using an oncolytic vaccinia virus as a delivery platform, we showed that IRF2-expressing vaccinia virus suppressed tumor progression and prolonged survival in multiple tumor models. These results suggest the potency of targeting IRF1 and using IRF2 to modulate immunotherapy.


Asunto(s)
Factor 1 Regulador del Interferón , Factor 2 Regulador del Interferón , Virus Oncolíticos , Animales , Factor 2 Regulador del Interferón/metabolismo , Factor 2 Regulador del Interferón/genética , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Ratones , Línea Celular Tumoral , Factor 1 Regulador del Interferón/metabolismo , Factor 1 Regulador del Interferón/genética , Viroterapia Oncolítica/métodos , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Ratones Endogámicos C57BL , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Modelos Animales de Enfermedad , Femenino
9.
Hepatology ; 55(6): 1863-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22234969

RESUMEN

UNLABELLED: Hypoxia is often found in solid tumors and is associated with tumor progression and poor clinical outcomes. The exact mechanisms related to hypoxia-induced invasion and metastasis remain unclear. We elucidated the mechanism by which the nuclear-damage-associated molecular pattern molecule, high-mobility group box 1 (HMGB1), released under hypoxic stress, can induce an inflammatory response to promote invasion and metastasis in hepatocellular carcinoma (HCC) cells. Caspase-1 activation was found to occur in hypoxic HCC cells in a process that was dependent on the extracellular release of HMGB1 and subsequent activation of both Toll-like receptor 4 (TLR4)- and receptor for advanced glycation endproducts (RAGE)-signaling pathways. Downstream from hypoxia-induced caspase-1 activation, cleavage and release of proinflammatory cytokines interleukin (IL)-1ß and -18 occurred. We further demonstrate that overexpression of HMGB1 or treatment with recombinant HMGB1 enhanced the invasiveness of HCC cells, whereas stable knockdown of HMGB1 remarkably reduced HCC invasion. Moreover, in a murine model of HCC pulmonary metastasis, stable knockdown of HMGB1 suppressed HCC invasion and metastasis. CONCLUSION: These results suggest that in hypoxic HCC cells, HMGB1 activates TLR4- and RAGE-signaling pathways to induce caspase-1 activation with the subsequent production of multiple inflammatory mediators, which, in turn, promote cancer invasion and metastasis.


Asunto(s)
Carcinoma Hepatocelular/patología , Caspasa 1/metabolismo , Proteína HMGB1/fisiología , Neoplasias Hepáticas/patología , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Activación Enzimática , Humanos , Interleucina-1beta/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/fisiología , Transducción de Señal , Receptor Toll-Like 4/fisiología
10.
Mol Ther ; 20(4): 736-48, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22008913

RESUMEN

Since previous work using a nonreplicating adenovirus-expressing mouse interferon-ß (Ad.mIFNß) showed promising preclinical activity, we postulated that a vector-expressing IFNß at high levels that could also replicate would be even more beneficial. Accordingly a replication competent, recombinant vaccinia viral vector-expressing mIFNß (VV.mIFNß) was tested. VV.mIFNß-induced antitumor responses in two syngeneic mouse flank models of lung cancer. Although VV.mIFNß had equivalent in vivo efficacy in both murine tumor models, the mechanisms of tumor killing were completely different. In LKRM2 tumors, viral replication was minimal and the tumor killing mechanism was due to activation of immune responses through induction of a local inflammatory response and production of antitumor CD8 T-cells. In contrast, in TC-1 tumors, the vector replicated well, induced an innate immune response, but antitumor activity was primarily due to a direct oncolytic effect. However, the VV.mIFNß vector was able to augment the efficacy of an antitumor vaccine in the TC-1 tumor model in association with increased numbers of infiltrating CD8 T-cells. These data show the complex relationships between oncolytic viruses and the immune system which, if understood and harnessed correctly, could potentially be used to enhance the efficacy of immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Interferón beta/metabolismo , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Femenino , Interferón beta/genética , Neoplasias Pulmonares/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Virus Vaccinia/inmunología , Replicación Viral/genética , Replicación Viral/fisiología
11.
J Exp Med ; 220(10)2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37552475

RESUMEN

While checkpoint blockade immunotherapies have widespread success, they rely on a responsive immune infiltrate; as such, treatments enhancing immune infiltration and preventing immunosuppression are of critical need. We previously generated αPD-1 resistant variants of the murine HNSCC model MEER. While entirely αPD-1 resistant, these tumors regress after single dose of oncolytic vaccinia virus (VV). We then generated a VV-resistant MEER line to dissect the immunologic features of sensitive and resistant tumors. While treatment of both tumor types induced immune infiltration and IFNγ, we found a defining feature of resistance was elevation of immunosuppressive cytokines like TGFß, which blunted IFNγ signaling, especially in regulatory T cells. We engineered VV to express a genetically encoded TGFßRII inhibitor. Inhibitor-expressing VV produced regressions in resistant tumor models and showed impressive synergy with checkpoint blockade. Importantly, tumor-specific, viral delivery of TGFß inhibition had no toxicities associated with systemic TGFß/TGFßR inhibition. Our data suggest that aside from stimulating immune infiltration, oncolytic viruses are attractive means to deliver agents to limit immunosuppression in cancer.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Ratones , Línea Celular Tumoral , Inmunosupresores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Microambiente Tumoral , Virus Vaccinia/genética
12.
Mol Cancer ; 11: 76, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23043612

RESUMEN

Serum lactate dehydrogenase (LDH) is a prognostic factor for patients with stage IV melanoma. To gain insights into the biology underlying this prognostic factor, we analyzed total serum LDH, serum LDH isoenzymes, and serum lactate in up to 49 patients with metastatic melanoma. Our data demonstrate that high serum LDH is associated with a significant increase in LDH isoenzymes 3 and 4, and a decrease in LDH isoenzymes 1 and 2. Since LDH isoenzymes play a role in both glycolysis and oxidative phosphorylation (OXPHOS), we subsequently determined using tissue microarray (TMA) analysis that the levels of proteins associated with mitochondrial function, lactate metabolism, and regulators of glycolysis were all elevated in advanced melanomas compared with nevic melanocytes. To investigate whether in advanced melanoma, the glycolysis and OXPHOS pathways might be linked, we determined expression of the monocarboxylate transporters (MCT) 1 and 4. Analysis of a nevus-to-melanoma progression TMA revealed that MCT4, and to a lesser extend MCT1, were elevated with progression to advanced melanoma. Further analysis of human melanoma specimens using the Seahorse XF24 extracellular flux analyzer indicated that metastatic melanoma tumors derived a large fraction of energy from OXPHOS. Taken together, these findings suggest that in stage IV melanomas with normal serum LDH, glycolysis and OXPHOS may provide metabolic symbiosis within the same tumor, whereas in stage IV melanomas with high serum LDH glycolysis is the principle source of energy.


Asunto(s)
Glucólisis , Melanoma/metabolismo , Fosforilación Oxidativa , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isoenzimas/sangre , L-Lactato Deshidrogenasa/sangre , Melanoma/sangre , Melanoma/patología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Estadificación de Neoplasias , Nevo/metabolismo , Factores de Acoplamiento de la Fosforilación Oxidativa/metabolismo
13.
Mol Ther ; 18(9): 1698-705, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20606649

RESUMEN

A major limitation to the use of immunotherapy in the treatment of cancer has been the localized immune suppressive environment within the tumor. Although there is evidence that tumor-selective (oncolytic) viruses may help to overcome this immune suppression, a primary limitation to their use has been limited systemic delivery potential, especially in the face of antiviral immunity. We recently demonstrated that tumor-trafficking immune cells can efficiently deliver oncolytic viral therapies to their tumor targets. These cells act as both a therapeutic agent and also a carrier vehicle for the oncolytic virus. Here, we demonstrate that such delivery is also possible in the face of pre-existing antiviral immunity, so overcoming the limited systemic delivery of naked, cell-free virus. It was also found that treatment of previously immunized mice or repeat treatments leading to immunization resulted in a switch from a primarily oncolytic to an immunotherapeutic mechanism of action. Furthermore, repeat cycles of treatment with combination immune cell-viral therapy resulted in increased tumor infiltration of effector T-cells and a general reduction in the levels of known immune suppressive lymphocyte populations. This therefore represents a novel and effective means to overcome localized immune suppression within the tumor microenvironment.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Viroterapia Oncolítica/enfermería , Animales , Línea Celular Tumoral , Células Asesinas Inducidas por Citocinas/inmunología , Perros , Ratones , Ratones Endogámicos C57BL
14.
J Immunother Cancer ; 8(1)2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32098828

RESUMEN

BACKGROUND: Natural killer (NK) cells have potent antitumor activities. Nevertheless, adoptive transfer therapy of NK cells has gained very limited success in patients with solid tumors as most infused NK cells remain circulating in the peripheral blood instead of entering tumor sites. Chemokines and their receptors play important roles in NK cell distribution. Enhancing chemokine receptors on immune cells to match and be driven to tumor-specific chemokines may improve the therapeutic efficacy of NK cells. METHODS: The CCR5-CCL5 axis is critical in NK cell homing to tumor sites. Thus, we analyzed CCR5 expression on NK cells from patients with cancer and healthy donors. We then upregulated CCR5 and CCL5 with lentiviruses and oncolytic viruses in NK and tumor cells, respectively. Animal experiments were also carried out to test the efficacy of the combination of oncolytic virus with NK cells. RESULTS: In NK cells from patients with various solid tumors or healthy subjects, CCR5 was expressed at low levels before and after expansion in vitro. CCR5-engineered NK cells showed enhanced tumor infiltration and antitumor effects, but no complete regressions were noted in the in vivo tumor models. To further improve therapeutic efficacy, we constructed CCL5-expressing oncolytic vaccinia virus. In vitro data demonstrated that vaccinia virus can produce CCL5 in tumor cells while infectivity remained unaffected. Supernatants from tumor cells infected by CCL5-modified vaccinia virus enhanced the directional movement of CCR5-overexpressed NK cells but not green fluorescent protein (GFP)-expressing cells. More importantly, NK cells were resistant to the vaccinia virus and their functions were not affected after being in contact. In vivo assays demonstrated that CCL5-expressing vaccinia virus induced a greater accumulation of NK cells within tumor lesions compared with that of the prototype virus. CONCLUSION: Enhancement of matched chemokines and chemokine receptors is a promising method of increasing NK cell homing and therapeutic effects. Oncolytic vaccinia viruses that express specific chemokines can synergistically augment the efficacies of NK cell-based therapy.


Asunto(s)
Quimiocina CCL5/metabolismo , Células Asesinas Naturales/inmunología , Virus Oncolíticos/inmunología , Receptores CCR5/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos
15.
Biochim Biophys Acta ; 1785(2): 217-31, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18328829

RESUMEN

Tremendous advances have been made in developing oncolytic viruses (OVs) in the last few years. By taking advantage of current knowledge in cancer biology and virology, specific OVs have been genetically engineered to target specific molecules or signal transduction pathways in cancer cells in order to achieve efficient and selective replication. The viral infection and amplification eventually induce cancer cells into cell death pathways and elicit host antitumor immune responses to further help eliminate cancer cells. Specifically targeted molecules or signaling pathways (such as RB/E2F/p16, p53, IFN, PKR, EGFR, Ras, Wnt, anti-apoptosis or hypoxia) in cancer cells or tumor microenvironment have been studied and dissected with a variety of OVs such as adenovirus, herpes simplex virus, poxvirus, vesicular stomatitis virus, measles virus, Newcastle disease virus, influenza virus and reovirus, setting the molecular basis for further improvements in the near future. Another exciting new area of research has been the harnessing of naturally tumor-homing cells as carrier cells (or cellular vehicles) to deliver OVs to tumors. The trafficking of these tumor-homing cells (stem cells, immune cells and cancer cells), which support proliferation of the viruses, is mediated by specific chemokines and cell adhesion molecules and we are just beginning to understand the roles of these molecules. Finally, we will highlight some avenues deserving further study in order to achieve the ultimate goals of utilizing various OVs for effective cancer treatment.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenovirus Humanos , Herpesvirus Humano 1 , Neoplasias/virología , Virus Oncolíticos , Transducción de Señal , Virus Vaccinia , Replicación Viral
16.
17.
Cancer Immunol Res ; 7(8): 1258-1266, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31239318

RESUMEN

Multiple studies have associated the transcription factor IRF1 with tumor-suppressive activities. Here, we report an opposite tumor cell-intrinsic function of IRF1 in promoting tumor growth. IRF1-deficient tumor cells showed reduced tumor growth in MC38 and CT26 colon carcinoma and B16 melanoma mouse models. This reduction in tumor growth was dependent on host CD8+ T cells. Detailed profiling of tumor-infiltrating leukocytes did not show changes in the various T-cell and myeloid cell populations. However, CD8+ T cells that had infiltrated IRF1-deficieint tumors in vivo exhibited enhanced cytotoxicity. IRF1-deficient tumor cells lost the ability to upregulate PD-L1 expression in vitro and in vivo and were more susceptible to T-cell-mediated killing. Induced expression of PD-L1 in IRF1-deficient tumor cells restored tumor growth. These results indicate differential activity of IRF1 in tumor escape.


Asunto(s)
Antígeno B7-H1/genética , Regulación Neoplásica de la Expresión Génica , Inmunomodulación , Factor 1 Regulador del Interferón/metabolismo , Animales , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Memoria Inmunológica , Inmunomodulación/genética , Factor 1 Regulador del Interferón/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo
18.
Methods Mol Biol ; 433: 243-58, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18679628

RESUMEN

The use of replication-competent oncolytic viruses has largely advanced cancer gene therapy. Oncolytic virus not only possesses unique mechanisms of action that are distinct from other treatment modalities, its self-perpetuating nature provides an ideal platform for therapeutic transgene insertion. Tumor selectivity can be achieved by deleting viral genes that are critical for growth in normal cells but dispensable in tumor cells, transcriptional control under tumor-specific promoters, fiber modification targeting tumor-specific cellular receptors, or the use of inherent tumor-specific viruses. Transgene products can be amplified along with viral replication, thus maximizing therapeutic effect. Using adenovirus as a template, this chapter describes common assays used for the study of oncolytic viruses, with special emphasis on in vitro and in vivo viral replication determination.


Asunto(s)
Adenoviridae/metabolismo , Terapia Genética/métodos , Neoplasias/genética , Neoplasias/terapia , Virus Oncolíticos/metabolismo , Línea Celular , Humanos , Imagenología Tridimensional , Proteínas Luminiscentes/metabolismo , Coloración y Etiquetado , Virión , Virosis/virología , Inactivación de Virus
19.
Oncoimmunology ; 7(3): e1395997, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29399394

RESUMEN

T cell trafficking into tumors depends on a "match" between chemokine receptors on effector cells (e.g., CXCR3 and CCR5) and tumor-secreted chemokines. There is often a chemokine/chemokine receptor "mismatch", with tumors producing minute amounts of chemokines, resulting in inefficient targeting of effectors to tumors. We aimed to alter tumors to produce higher levels of CXCL11, a CXCR3 ligand, to attract more effector cells following immunotherapy. Mice bearing established subcutaneous tumors were studied. In our first approach, we used modified chimeric antigen receptor (CAR)-transduced human T cells to deliver CXCL11 (CAR/CXCL11) into tumors. In our second approach, we intravenously (iv) administered a modified oncolytic vaccinia virus (VV) engineered to produce CXCL11 (VV.CXCL11). The effect of these treatments on T cell trafficking into the tumors and anti-tumor efficacy after subsequent CAR T cell injections or anti-tumor vaccines was determined. CAR/CXCL11 and VV.CXCL11 significantly increased CXCL11 protein levels within tumors. For CAR/CXCL11, injection of a subsequent dose of CAR T cells did not result in increased intra-tumoral trafficking, and appeared to decrease the function of the injected CAR T cells. In contrast, VV.CXCL11 increased the number of total and antigen-specific T cells within tumors after CAR T cell injection or vaccination and significantly enhanced anti-tumor efficacy. Both approaches were successful in increasing CXCL11 levels within the tumors; however, only the vaccinia approach was successful in recruiting T cells and augmenting anti-tumor efficacy. VV.CXCL11 should be considered as a potential approach to augment adoptive T cell transfer or vaccine immunotherapy.

20.
J Psychiatr Pract ; 13(4): 261-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17667740

RESUMEN

Violence associated with personality disorders is usually best viewed separately from psychiatric diagnosis, as a syndrome of violence rather than a syndrome of diagnosis. The authors describe eight categories of violence associated with personality disorders that may help clinicians choose treatment or management techniques: purposeful, instrumental violence; purposeful, non-instrumental violence; purposeful, targeted, defensive violence; targeted, impulsive violence; nontargeted, impulsive violence incidental to emotional escape; random but purposeful violence; violence related to perceived or feared loss or abandonment; and violence related to chronic paranoia or related misconceptions. The categories are not completely mutually exclusive, nor do they represent a "decision tree." We also point out three important principles about the relationship between personality disorders and violence: 1) Personality disorders are rarely ego dystonic; 2) Most patients and violent situations that come to clinical attention involve comorbid conditions. 3) Violence and violence risk are often associated with intoxication.


Asunto(s)
Trastornos de la Personalidad/diagnóstico , Violencia/psicología , Comorbilidad , Psiquiatría Forense , Humanos , Trastornos de la Personalidad/psicología , Trastornos de la Personalidad/terapia , Medición de Riesgo , Violencia/clasificación , Violencia/legislación & jurisprudencia , Violencia/prevención & control
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda