Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1036: 157-172, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29275471

RESUMO

Oncolytic viral therapy is a promising approach to treat many malignancies, including breast, colorectal, hepatocellular, and melanoma. The best results are seen when using "targeted and armed" viruses. These are viruses that have been genetically modified to selectively replicate within cancer cells and express specific transgenes that alter the tumor microenvironment to inhibit tumor progression. The products of these transgenes induce cell death, make the virus less virulent, compromise tumor vascularity, and are capable of modulating or enhancing the immune system-such as cytokines and chemokines. In addition, oncolytic viruses can induce anti-vascular effects and disrupt the extracellular matrix to improve viral spread within the tumor. Oncolytic viruses also improve crosstalk between fibroblasts, cytokine-induced killer cells, and cancer cells within the microenvironment, leading to enhanced tumor cell death.


Assuntos
Neoplasias , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Microambiente Tumoral/imunologia , Animais , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
2.
Mol Ther ; 21(3): 620-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23229093

RESUMO

The combination of an oncolytic virus, that directly destroys tumor cells and mediates an acute immune response, with an immune cell therapy, capable of further enlisting and enhancing the host immune response, has the potential to create a potent therapeutic effect. We have previously developed several strategies for optimizing the delivery of oncolytic vaccinia virus vectors to their tumor targets, including the use of immune cell-based carrier vehicles and the incorporation of mutations that increase production of the enveloped form of vaccinia (extracellular enveloped viral (EEV)) that is better adapted to spread within a host. Here, we initially combine these approaches to create a novel therapeutic, consisting of an immune cell (cytokine-induced killer, CIK) preloaded with an oncolytic virus that is EEV enhanced. This resulted in direct interaction between the viral and immune cell components with each assisting the other in directing the therapy to the tumor and so enhancing the antitumor effects. This effect could be further improved through CCL5 expression from the virus. The resulting multicomponent therapy displays the ability for synergistic crosstalk between components, so significantly enhancing tumor trafficking and antitumor effects.


Assuntos
Antineoplásicos/farmacologia , Células Matadoras Induzidas por Citocinas/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Animais , Linhagem Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Replicação Viral
3.
Blood ; 127(21): 2509, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27231392
4.
J Virol ; 85(1): 21-31, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20962097

RESUMO

Vaccinia virus (VacV) enters mammalian cells, replicates extranuclearly, and produces virions that move to the cell surface along microtubules, fuse with the plasma membrane, and move from infected cells toward apposing cells on actin-filled membranous protrusions or actin tails. To form actin tails, cell-associated enveloped virions (CEV) require Abl and Src family tyrosine kinases. Furthermore, release of CEV from the cell requires Abl but not Src family tyrosine kinases and is blocked by imatinib mesylate (STI-571; Gleevec), an Abl family kinase inhibitor used to treat chronic myelogenous leukemia in humans. Here we demonstrate that the Poxviridae family members monkeypox virus (MPX) and variola virus (VarV) use conserved mechanisms for actin motility and extracellular enveloped virion (EEV) release. Furthermore, we show that imatinib mesylate is effective in a mouse model of infection with VacV, whether delivered prophylactically or postinfection, and restricts spread of virions from the site of inoculation. While inhibitors of both Src and Abl family kinases, such as dasatinib (BMS-354825; Sprycel), are effective in limiting dissemination of VacV, VarV, and MPX in vitro, members of this class of drugs appear to have immunosuppressive effects in vivo that preclude their use as anti-infectives. Together, these data suggest a possible utility for imatinib mesylate in treating smallpox or MPX infections or complications associated with vaccination.


Assuntos
Monkeypox virus/enzimologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Vírus da Varíola/enzimologia , Vírion/fisiologia , Liberação de Vírus/fisiologia , Quinases da Família src/metabolismo , Células 3T3 , Actinas/metabolismo , Animais , Benzamidas , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Mesilato de Imatinib , Camundongos , Camundongos Endogâmicos BALB C , Monkeypox virus/efeitos dos fármacos , Monkeypox virus/fisiologia , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Vacínia/tratamento farmacológico , Vacínia/prevenção & controle , Vacínia/virologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/enzimologia , Vírus da Varíola/efeitos dos fármacos , Vírus da Varíola/fisiologia , Liberação de Vírus/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores
5.
Mol Ther ; 19(4): 650-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21266959

RESUMO

Tumor vaccines can induce robust immune responses targeting tumor antigens in the clinic, but antitumor effects have been disappointing. One reason for this is ineffective tumor infiltration of the cytotoxic T lymphocytes (CTLs) produced. Oncolytic viruses are capable of selectively replicating within tumor tissue and can induce a strong immune response. We therefore sought to determine whether these therapies could be rationally combined such that modulation of the tumor microenvironment by the viral therapy could help direct beneficial CTLs induced by the vaccine. As such, we examined the effects of expressing chemokines from oncolytic vaccinia virus, including CCL5 (RANTES), whose receptors are expressed on CTLs induced by different vaccines, including type-1-polarized dendritic cells (DC1). vvCCL5, an oncolytic vaccinia virus expressing CCL5, induced chemotaxis of lymphocyte populations in vitro and in vivo, and displayed improved safety in vivo. Interestingly, enhanced therapeutic benefits with vvCCL5 in vivo correlated with increased persistence of the viral agent exclusively within the tumor. When tumor-bearing mice were both vaccinated with DC1 and treated with vvCCL5 a further significant enhancement in tumor response was achieved which correlated with increased levels of tumor infiltrating lymphocytes. This approach therefore represents a novel means of combining biological therapies for cancer treatment.


Assuntos
Quimiocina CCL5/metabolismo , Células Dendríticas/imunologia , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Animais , Linhagem Celular Tumoral , Quimiocina CCL5/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Camundongos , Camundongos Mutantes , Camundongos Nus , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Vaccinia virus/genética , Vaccinia virus/metabolismo
6.
J Gene Med ; 13(12): 692-701, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22028274

RESUMO

BACKGROUND: Preclinical studies have demonstrated that, unlike oncolytic adenoviruses, oncolytic vaccinia viruses can reach implanted tumors upon systemic injection. However, the biodistribution of this oncolytic agent in in situ autochthonous tumor models remains poorly characterized. In the present study, we assessed this biodistribution in a model of mouse hepatocellular carcinoma (HCC) obtained after injection of the carcinogen diethylnitrosamine (DEN). METHODS: Twelve months after DEN administration, histology, quantitative reverse transcription-polymerase chain reaction, in situ hybridization and viral titration were used to characterize tumors, as well as to assess the viral load of the livers upon either intravenous or intraperitoineal injection. RESULTS: The results obtained showed that the architecture of the liver was lost, with a noticeable absence of sinusoids, as well as the presence of steatosis and α-fetoprotein-positive HCC tumor nodules. Bioluminescence imaging and measures of the infective virus load demonstrated that intravenous injection of 10(8) plaque-forming units of the recombinant vaccinia virus led to a predominant transduction of the liver, whereas intraperitoneal injection resulted in a lower level of liver transduction accompanied by an increased infection of the lungs, spleen, kidneys and bowels. Immunohistochemical analysis of liver sections of animals injected intravenously with the virus revealed a preferential localization of vaccinia-specific immunoreactivity in the tumors. CONCLUSIONS: The findings of the present study emphasize the importance of the route of administration of the vector and highlight the relevance of systemic injection of oncolytic vaccinia virus in the context of hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias Experimentais , Vírus Oncolíticos/genética , Poxviridae/genética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virologia , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Neoplasias Experimentais/virologia , Terapia Viral Oncolítica , Distribuição Tecidual
7.
J Immunol ; 183(1): 332-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542444

RESUMO

An early reaction of CD4(+) T lymphocytes to Ag is the production of cytokines, notably IL-2. To detect cytokine-dependent responses, naive Ag-specific T cells were stimulated in vivo and the presence of phosphorylated STAT5 molecules was used to identify the cell populations responding to IL-2. Within hours of T cell priming, IL-2-dependent STAT5 phosphorylation occurred primarily in Foxp3(+) regulatory T cells. In contrast, the Ag-specific T cells received STAT5 signals only after repeated Ag exposure or memory differentiation. Regulatory T cells receiving IL-2 signals proliferated and developed enhanced suppressive activity. These results indicate that one of the earliest events in a T cell response is the activation of endogenous regulatory cells, potentially to prevent autoimmunity.


Assuntos
Ativação Linfocitária/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Proliferação de Células , Interleucina-2/biossíntese , Interleucina-2/fisiologia , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Fosforilação , Fase de Repouso do Ciclo Celular/genética , Fase de Repouso do Ciclo Celular/imunologia , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/virologia , Linfócitos T Reguladores/virologia , Fatores de Tempo , Vaccinia virus/imunologia
8.
Proc Natl Acad Sci U S A ; 105(34): 12128-33, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18713866

RESUMO

Many cancer therapeutic agents elicit resistance that renders them ineffective and often produces cross-resistance to other drugs. One of the most common mechanisms of resistance involves P-glycoprotein (Pgp)-mediated drug efflux. To address this problem, new agents have been sought that are less prone to inducing resistance and less likely to serve as substrates for Pgp efflux. An alternative to this approach is to deliver established agents as molecular transporter conjugates into cells through a mechanism that circumvents Pgp-mediated efflux and allows for release of free drug only after cell entry. Here we report that the widely used chemotherapeutic agent Taxol, ineffective against Taxol-resistant human ovarian cancer cell lines, can be incorporated into a releasable octaarginine conjugate that is effective against the same Taxol-resistant cell lines. It is significant that the ability of the Taxol conjugates to overcome Taxol resistance is observed both in cell culture and in animal models of ovarian cancer. The generality and mechanistic basis for this effect were also explored with coelenterazine, a Pgp substrate. Although coelenterazine itself does not enter cells because of Pgp efflux, its octaarginine conjugate does so readily. This approach shows generality for overcoming the multidrug resistance elicited by small-molecule cancer chemotherapeutics and could improve the prognosis for many patients with cancer and fundamentally alter search strategies for novel therapeutic agents that are effective against resistant disease.


Assuntos
Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Oligopeptídeos/farmacologia , Paclitaxel/farmacocinética , Transporte Biológico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/química
9.
J Clin Invest ; 117(11): 3350-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17965776

RESUMO

Replication-selective oncolytic viruses (virotherapeutics) are being developed as novel cancer therapies with unique mechanisms of action, but limitations in i.v. delivery to tumors and systemic efficacy have highlighted the need for improved agents for this therapeutic class to realize its potential. Here we describe the rational, stepwise design and evaluation of a systemically effective virotherapeutic (JX-963). We first identified a highly potent poxvirus strain that also trafficked efficiently to human tumors after i.v. administration. This strain was then engineered to target cancer cells with activation of the transcription factor E2F and the EGFR pathway by deletion of the thymidine kinase and vaccinia growth factor genes. For induction of tumor-specific cytotoxic T lymphocytes, we further engineered the virus to express human GM-CSF. JX-963 was more potent than the previously used virotherapeutic Onyx-015 adenovirus and as potent as wild-type vaccinia in all cancer cell lines tested. Significant cancer selectivity of JX-963 was demonstrated in vitro in human tumor cell lines, in vivo in tumor-bearing rabbits, and in primary human surgical samples ex vivo. Intravenous administration led to systemic efficacy against both primary carcinomas and widespread organ-based metastases in immunocompetent mice and rabbits. JX-963 therefore holds promise as a rationally designed, targeted virotherapeutic for the systemic treatment of cancer in humans and warrants clinical testing.


Assuntos
Neoplasias/metabolismo , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Poxviridae/fisiologia , Animais , Linhagem Celular Tumoral , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/genética , Vírus Oncolíticos/genética , Poxviridae/genética , Coelhos
10.
Expert Rev Mol Med ; 12: e2, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20064281

RESUMO

The ability to externally regulate the expression or function of a gene product has proven to be a powerful tool in the study of proteins and disease in vitro, and more recently in transgenic animal models. The transfer of these technologies to regulate a therapeutic, adoptively transferred gene product in a clinical setting may provide a means to exert additional control over a large variety of therapies for many diseases, leading to increased safety and effectiveness. This could be applied to any biological therapy, including gene therapy, viral therapies, cellular therapies (such as immune cell therapies, stem cell therapies and bone marrow transplant), some vaccines and even organ transplant. A variety of systems have been used in a basic research setting to conditionally regulate the function of a protein, including control of transcription and mRNA stability, and the use of protein inhibitors. However, most of these have disadvantages for medical use, where a simple, specific, tunable, reversible and broadly applicable means to regulate protein function is needed. Recent advances in controlling the stability or function of proteins through the interaction of small-molecule effectors and fusion domains on the protein have raised the possibility that direct and highly specific external control of therapeutic protein function in humans will be feasible.


Assuntos
Terapia Biológica/métodos , Estabilidade Proteica , Animais , Doença , Humanos , Proteínas/química , Proteínas/genética , Proteínas/metabolismo
11.
BMC Cancer ; 10: 146, 2010 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-20398414

RESUMO

BACKGROUND: Bacterial targeting of tumours is an important anti-cancer strategy. We previously showed that strain SL7838 of Salmonella typhimurium targets and kills cancer cells. Whether NO generation by the bacteria has a role in SL7838 lethality to cancer cells is explored. This bacterium has the mechanism for generating NO, but also for decomposing it. METHODS: Mechanism underlying Salmonella typhimurium tumour therapy was investigated through in vitro and in vivo studies. NO measurements were conducted either by chemical assays (in vitro) or using Biosensors (in vivo). Cancer cells cytotoxic assay were done by using MTS. Bacterial cell survival and tumour burden were determined using molecular imaging techniques. RESULTS: SL7838 generated nitric oxide (NO) in anaerobic cell suspensions, inside infected cancer cells in vitro and in implanted 4T1 tumours in live mice, the last, as measured using microsensors. Thus, under these conditions, the NO generating pathway is more active than the decomposition pathway. The latter was eliminated, in strain SL7842, by the deletion of hmp- and norV genes, making SL7842 more proficient at generating NO than SL7838. SL7842 killed cancer cells more effectively than SL7838 in vitro, and this was dependent on nitrate availability. This strain was also ca. 100% more effective in treating implanted 4T1 mouse tumours than SL7838. CONCLUSIONS: NO generation capability is important in the killing of cancer cells by Salmonella strains.


Assuntos
Terapia Biológica/métodos , Neoplasias/terapia , Óxido Nítrico/metabolismo , Salmonella typhimurium/metabolismo , Animais , Técnicas Biossensoriais , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação Bacteriana da Expressão Gênica , Humanos , Hidrazinas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/microbiologia , Neoplasias/patologia , Doadores de Óxido Nítrico/farmacologia , Salmonella typhimurium/genética , Fatores de Tempo , Carga Tumoral
13.
Mol Cancer Ther ; 8(2): 333-41, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19190118

RESUMO

We report the discovery of a new prodrug, 6-chloro-9-nitro-5-oxo-5H-benzo(a)phenoxazine (CNOB). This prodrug is efficiently activated by ChrR6, the highly active prodrug activating bacterial enzyme we have previously developed. The CNOB/ChrR6 therapy was effective in killing several cancer cell lines in vitro. It also efficiently treated tumors in mice with up to 40% complete remission. 9-Amino-6-chloro-5H-benzo(a)phenoxazine-5-one (MCHB) was the only product of CNOB reduction by ChrR6. MCHB binds DNA; at nonlethal concentration, it causes cell accumulation in the S phase, and at lethal dose, it induces cell surface Annexin V and caspase-3 and caspase-9 activities. Further, MCHB colocalizes with mitochondria and disrupts their electrochemical potential. Thus, killing by CNOB involves MCHB, which likely induces apoptosis through the mitochondrial pathway. An attractive feature of the CNOB/ChrR6 regimen is that its toxic product, MCHB, is fluorescent. This feature proved helpful in in vitro studies because simple fluorescence measurements provided information on the kinetics of CNOB activation within the cells, MCHB killing mechanism, its generally efficient bystander effect in cells and cell spheroids, and its biodistribution. The emission wavelength of MCHB also permitted its visualization in live animals, allowing noninvasive qualitative imaging of MCHB in mice and the tumor microenvironment. This feature may simplify exploration of barriers to the penetration of MCHB in tumors and their amelioration.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Nitrorredutases/uso terapêutico , Oxazinas/uso terapêutico , Pró-Fármacos/uso terapêutico , Animais , Anexina A5/metabolismo , Antineoplásicos/farmacologia , Efeito Espectador/efeitos dos fármacos , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Fluorescência , Humanos , Cinética , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/enzimologia , Neoplasias/patologia , Oxazinas/farmacologia , Pró-Fármacos/farmacologia , Distribuição Tecidual/efeitos dos fármacos , Resultado do Tratamento
14.
Methods Mol Biol ; 542: 635-47, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19565925

RESUMO

Oncolytic or replication-selective viruses have been used as powerful tools for the delivery of therapeutic genes to tumors. Because these vectors are capable of replicating within the tumor, the therapeutic gene is amplified within the target tissue itself, resulting in the spread of the virus both within the tumor, and sometimes also between tumors. Vaccinia virus holds many advantages when serving as the backbone for oncolytic viral strains, including a large cloning capacity (at least 25 kbp) (1); a short life-cycle (2, 3); extensive previous use in humans, with contraindications and adverse reactions well described and antivirals available (4); the potential for systemic (intravenous) delivery to distant tumors; and vaccinia strains have previously demonstrated antitumor benefits in clinical trials (5). Because vaccinia has no known receptor and is capable of infecting almost any cell type, tumor selectivity has to be engineered into vaccinia at steps after infection. We will therefore discuss potential viral virulence genes and metabolic targets that result in tumor-selective vaccinia strains. Because the virus has limited natural requirements for host cell proteins, and, instead, contains a large genome and multiple genes involved in virulence, a large number of possible attenuating gene deletions can result in the production of viral strains reliant on inherent properties of the host cell for replication. The protocols for producing viral gene deletions and constructing viral gene expression vectors have been well established for vaccinia and are summarized briefly in this chapter. Basic assays for testing the tumor selectivity and therapeutic index of new oncolytic constructs in vitro will be covered. In addition, we describe how bioluminescence imaging can be incorporated into preclinical testing of vaccinia gene expression strains to examine the timing, biodistribution, and kinetics of viral gene expression noninvasively after delivery of the viral agents to tumor-bearing mice via different routes.


Assuntos
Biologia Molecular/métodos , Vírus Oncolíticos/fisiologia , Vaccinia virus/classificação , Vaccinia virus/fisiologia , Animais , Linhagem Celular , Humanos , Medições Luminescentes , Replicação Viral
15.
Curr Opin Mol Ther ; 10(4): 387-92, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18683104

RESUMO

The field of oncolytic viral therapy has undergone a major shift in focus in the last few years. Less research has been directed at making incremental improvements in original vectors based mainly on strains of adenovirus and HSV; instead a variety of different viral strains have been suggested as potential backbones for future oncolytic viruses (including Newcastle disease virus, reovirus, vesicular stomatitis virus, polio virus, retrovirus, Sindbis virus, picornavirus, mumps and measles virus), with many of these progressing to clinical trials. Of these, vaccinia virus represents a particularly promising candidate. It possesses a variety of intrinsic molecular properties suitable for an oncolytic virus (such as rapid life cycle and lysis of infected cells, and an ability to infect various cell types), in addition to undergoing extensive study both in the laboratory and in the clinic. Although not a natural human pathogen, there are extensive data on the effects of vaccinia infection in humans. Preclinical models incorporating new oncolytic vaccinia strains, as well as data from the first clinical trials that have utilized the next-generation oncolytic vaccinia strains for the potential treatment of cancer have been described.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Vaccinia virus , Animais , Ensaios Clínicos como Assunto , Deleção de Genes , Humanos , Neoplasias/patologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Vaccinia virus/genética , Vaccinia virus/metabolismo
16.
PLoS Med ; 4(12): e353, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18162040

RESUMO

BACKGROUND: Oncolytic viruses hold much promise for clinical treatment of many cancers, but a lack of systemic delivery and insufficient tumor cell killing have limited their usefulness. We have previously demonstrated that vaccinia virus strains are capable of systemic delivery to tumors in mouse models, but infection of normal tissues remains an issue. We hypothesized that interferon-beta (IFN-beta) expression from an oncolytic vaccinia strain incapable of responding to this cytokine would have dual benefits as a cancer therapeutic: increased anticancer effects and enhanced virus inactivation in normal tissues. We report the construction and preclinical testing of this virus. METHODS AND FINDINGS: In vitro screening of viral strains by cytotoxicity and replication assay was coupled to cellular characterization by phospho-flow cytometry in order to select a novel oncolytic vaccinia virus. This virus was then examined in vivo in mouse models by non-invasive imaging techniques. A vaccinia B18R deletion mutant was selected as the backbone for IFN-beta expression, because the B18R gene product neutralizes secreted type-I IFNs. The oncolytic B18R deletion mutant demonstrated IFN-dependent cancer selectivity and efficacy in vitro, and tumor targeting and efficacy in mouse models in vivo. Both tumor cells and tumor-associated vascular endothelial cells were targeted. Complete tumor responses in preclinical models were accompanied by immune-mediated protection against tumor rechallenge. Cancer selectivity was also demonstrated in primary human tumor explant tissues and adjacent normal tissues. The IFN-beta gene was then cloned into the thymidine kinase (TK) region of this virus to create JX-795 (TK-/B18R-/IFN-beta+). JX-795 had superior tumor selectivity and systemic intravenous efficacy when compared with the TK-/B18R- control or wild-type vaccinia in preclinical models. CONCLUSIONS: By combining IFN-dependent cancer selectivity with IFN-beta expression to optimize both anticancer effects and normal tissue antiviral effects, we were able to achieve, to our knowledge for the first time, tumor-specific replication, IFN-beta gene expression, and efficacy following systemic delivery in preclinical models.


Assuntos
Interferon beta/metabolismo , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/metabolismo , Vaccinia virus/metabolismo , Animais , Sobrevivência Celular , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Genes Reporter , Células HCT116 , Haplorrinos , Humanos , Interferon-alfa/metabolismo , Interferon beta/genética , Luciferases , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Vírus Oncolíticos/genética , Deleção de Sequência , Timidina Quinase/genética , Timidina Quinase/metabolismo , Fatores de Tempo , Distribuição Tecidual , Vaccinia virus/enzimologia , Vaccinia virus/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
17.
Clin Cancer Res ; 12(6): 1859-67, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16551871

RESUMO

Cytokine-induced killer (CIK) cells are ex vivo activated and expanded CD8+ natural killer T cells that have been shown to have antitumor activity. This is the first study exploring cell killing of primary ovarian carcinoma cells with and without bispecific antibodies. Primary cancer cells and autologous CIK cells were collected from women with epithelial ovarian cancer. Bispecific antibodies against cancer antigen-125 (BSAbxCA125) and Her2 (BSAbxHer2) were developed using chemical heteroconjugation. On fluorescence-activated cell sorting analysis, the expansion of CIK cells resulted in a significant increase of CD3+CD8+ and CD3+CD56+ T cells. With enhancement by bispecific antibodies, the mean percent lysis in a 51Cr release assay of fresh ovarian cancer cells exposed to autologous CIK cells increased from 21.7 +/- 0.3% to 89.4 +/- 2.1% at an E:T ratio of 100:1 (P < 0.001). Anti-NKG2D antibodies attenuated the CIK activity by 56.8% on primary cells (P < 0.001). In a xenograft severe combined immunodeficient mouse model, real-time tumor regression and progression was visualized using a noninvasive in vivo bioluminescence imaging system. Four hours after CIK cell injection, we were able to visualize CD8+NKG2D+ CIK cells infiltrating Her2-expressing cancer cells on fluorescence microscopy. Mice that underwent adoptive transfer of CIK cells redirected with BSAbxCA125 and BSAbxHer2 had significant reduction in tumor burden (P < 0.001 and P < 0.001) and improvement in survival (P = 0.05 and P = 0.006) versus those treated with CIK cells alone. Bispecific antibodies significantly enhanced the cytotoxicity of CIK cells in primary ovarian cancer cells and in our in vivo mouse model. The mechanism of cytolysis seems to be mediated in part by the NKG2D receptor.


Assuntos
Anticorpos Biespecíficos/farmacologia , Linfócitos T CD8-Positivos/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Ovarianas/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Linfócitos T CD8-Positivos/citologia , Linhagem Celular Tumoral , Citocinas/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/imunologia , Feminino , Humanos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/citologia , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos SCID , Microscopia de Fluorescência , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
18.
Trends Cancer ; 2(2): 67-68, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27004260

RESUMO

The identification of STING as a key cytoplasmic innate recognition molecule for DNA viruses whose function is lost in a variety of cancers has coincided with the approval of IMLYGIC for metastatic melanoma. This represents the first replication competent viral therapy approved for the treatment of any cancer in the US. The role of STING pathway in the selectivity of HSV has been addressed for the first time in Xia et al (1).

19.
Methods Mol Biol ; 1444: 67-72, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27283418

RESUMO

The capacity to combine noninvasive whole animal imaging of genetic reporters and exogenously added probes in a single animal makes fluorescence imaging a powerful tool for investigating molecular events in live animals in preclinical research. However, the adsorption and diffraction properties of light passing through tissues mean that the choice of reporters, models, and imaging systems needs to be carefully considered. Here, we describe approaches to design and run experiments incorporating noninvasive whole animal fluorescence imaging into small animal imaging studies.


Assuntos
Genes Reporter , Tomografia de Coerência Óptica/instrumentação , Imagem Corporal Total/instrumentação , Animais , Corantes Fluorescentes/administração & dosagem , Camundongos , Modelos Animais , Tomografia de Coerência Óptica/métodos , Imagem Corporal Total/métodos
20.
Cancer Cell ; 30(1): 108-119, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27374223

RESUMO

Immunotherapies are highly promising cancer treatments, but understanding the factors mediating their resistance remains critical. Successes in randomized clinical testing have supported the growing appreciation that oncolytic virotherapies primarily act as immunotherapies. Here we identified prostaglandin E2 (PGE2) in the tumor as a key mediator of resistance to immunotherapies, including oncolytic vaccinia virotherapy. Elevated levels of PGE2 coupled to suppressive chemokine profiles and high levels of granulocytic myeloid-derived suppressor cells resulted in loss of immunotherapeutic potential. Viral vectors engineered to target PGE2 were capable of overcoming localized immunosuppression leading to profound changes in the tumor's immune status. This allowed the viral vectors to raise robust anti-tumor adaptive immune responses and sensitized established and previously resistant tumors to immunotherapies.


Assuntos
Quimiocinas/metabolismo , Dinoprostona/antagonistas & inibidores , Marcação de Genes/métodos , Hidroxiprostaglandina Desidrogenases/genética , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica/métodos , Animais , Vacinas Anticâncer , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Vetores Genéticos/administração & dosagem , Hidroxiprostaglandina Desidrogenases/farmacologia , Imunoterapia , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Análise de Sobrevida , Resultado do Tratamento , Vaccinia virus/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa