Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Cancer ; 135(5): 1238-46, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24474587

RESUMO

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.


Assuntos
Neoplasias/terapia , Neovascularização Patológica/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Vaccinia virus , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/uso terapêutico , Terapia Genética , Humanos , Indóis/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirróis/uso terapêutico , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/genética
2.
Mol Ther ; 21(1): 167-74, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23281444

RESUMO

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.


Assuntos
Citocinas/fisiologia , Terapia Genética , Neoplasias/terapia , Animais , Citocinas/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica , Transgenes , Vaccinia virus/metabolismo , Vaccinia virus/fisiologia , Replicação Viral
3.
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
4.
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
5.
Mol Ther ; 18(9): 1698-705, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20606649

RESUMO

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.


Assuntos
Imunoterapia/métodos , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Terapia Viral Oncolítica/enfermagem , Animais , Linhagem Celular Tumoral , Células Matadoras Induzidas por Citocinas/imunologia , Cães , Camundongos , Camundongos Endogâmicos C57BL
6.
J Acoust Soc Am ; 128(1): 456-65, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20649239

RESUMO

Speech sound patterns can be discerned using multiple acoustic cues. The relative weighting of these cues is known to be language-specific. Speech-sound training in adults induces changes in cue-weighting such that relevant acoustic cues are emphasized. In the current study, the extent to which individual variability in cue weighting contributes to differential success in learning to use foreign sound patterns was examined. Sixteen English-speaking adult participants underwent a sound-to-meaning training paradigm, during which they learned to incorporate Mandarin linguistic pitch contours into words. In addition to cognitive tests, measures of pitch pattern discrimination and identification were collected from all participants. Reaction time data from the discrimination task was subjected to 3-way multidimensional scaling to extract dimensions underlying tone perception. Two dimensions relating to pitch height and pitch direction were found to underlie non-native tone space. Good learners attended more to pitch direction relative to poor learners, before and after training. Training increased the ability to identify and label pitch direction. The results demonstrate that variability in the ability to successfully learn to use pitch in lexical contexts can be explained by pre-training differences in cue-weighting.


Assuntos
Sinais (Psicologia) , Idioma , Fonética , Reconhecimento Psicológico , Detecção de Sinal Psicológico , Acústica da Fala , Percepção da Fala , Estimulação Acústica , Adulto , Cognição , Feminino , Humanos , Masculino , Reconhecimento Fisiológico de Modelo , Discriminação da Altura Tonal , Psicoacústica , Tempo de Reação , Fatores de Tempo , Adulto Jovem
7.
J Immunother Cancer ; 8(1)2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32098828

RESUMO

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.


Assuntos
Quimiocina CCL5/metabolismo , Células Matadoras Naturais/imunologia , Vírus Oncolíticos/imunologia , Receptores CCR5/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus
8.
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
9.
Cell Rep ; 15(2): 264-73, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27050526

RESUMO

The immune response plays a key role in enhancing the therapeutic activity of oncolytic virotherapies. However, to date, investigators have relied on inherent interactions between the virus and the immune system, often coupled to the expression of a single cytokine transgene. Recently, the importance of TLR activation in mediating adaptive immunity has been demonstrated. We therefore sought to influence the type and level of immune response raised after oncolytic vaccinia therapy through manipulation of TLR signaling. Vaccinia naturally activates TLR2, associated with an antibody response, whereas a CTL response is associated with TLR3-TRIF-signaling pathways. We manipulated TLR signaling by vaccinia through deglycosylation of the viral particle to block TLR2 activation and expression of a TRIF transgene. The resulting vector displayed greatly reduced production of anti-viral neutralizing antibody as well as an increased anti-tumor CTL response. Delivery in both naive and pre-treated mice was enhanced and immunotherapeutic activity dramatically improved.


Assuntos
Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica , Transdução de Sinais , Linfócitos T/metabolismo , Receptor 2 Toll-Like/metabolismo , Vaccinia virus/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Glicosilação , Imunoterapia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Necrose , Linfócitos T Citotóxicos/metabolismo , Timidina Quinase/metabolismo
10.
Oncolytic Virother ; 4: 75-82, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27512672

RESUMO

Results from randomized clinical trials over the last several years have finally begun to demonstrate the potential of oncolytic viral therapies to treat a variety of cancers. One reason for these successes has been the realization that this platform is most effective when considered primarily as an immunotherapy. Cancer immunotherapy has also made dramatic strides recently with antibodies capable of blocking immune checkpoint inhibitors and adoptive T-cell therapies, notably CAR T-cells, leading a panel of novel and highly clinically effective therapies. It is clear therefore that an understanding of how and when these complementary approaches can most effectively be combined offers the real hope of moving beyond simply treating the disease and toward starting to talk about curative therapies. In this review we discuss approaches to combining these therapeutic platforms, both through engineering the viral vectors to more beneficially interact with the host immune response during therapy, as well as through the direct combinations of different therapeutics. This primarily, but not exclusively focuses on strains of oncolytic vaccinia virus. Some of the results reported to date, primarily in pre-clinical models but also in early clinical trials, are dramatic and hold great promise for the future development of similar therapies and their translation into cancer therapies.

11.
Clin Cancer Res ; 21(24): 5543-51, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26187615

RESUMO

PURPOSE: Recent data from randomized clinical trials with oncolytic viral therapies and with cancer immunotherapies have finally recapitulated the promise these platforms demonstrated in preclinical models. Perhaps the greatest advance with oncolytic virotherapy has been the appreciation of the importance of activation of the immune response in therapeutic activity. Meanwhile, the understanding that blockade of immune checkpoints (with antibodies that block the binding of PD1 to PDL1 or CTLA4 to B7-2) is critical for an effective antitumor immune response has revitalized the field of immunotherapy. The combination of immune activation using an oncolytic virus and blockade of immune checkpoints is therefore a logical next step. EXPERIMENTAL DESIGN: Here, we explore such combinations and demonstrate their potential to produce enhanced responses in mouse tumor models. Different combinations and regimens were explored in immunocompetent mouse models of renal and colorectal cancer. Bioluminescence imaging and immune assays were used to determine the mechanisms mediating synergistic or antagonistic combinations. RESULTS: Interaction between immune checkpoint inhibitors and oncolytic virotherapy was found to be complex, with correct selection of viral strain, antibody, and timing of the combination being critical for synergistic effects. Indeed, some combinations produced antagonistic effects and loss of therapeutic activity. A period of oncolytic viral replication and directed targeting of the immune response against the tumor were required for the most beneficial effects, with CD8(+) and NK, but not CD4(+) cells mediating the effects. CONCLUSIONS: These considerations will be critical in the design of the inevitable clinical translation of these combination approaches. Clin Cancer Res; 21(24); 5543-51. ©2015 AACR.See related commentary by Slaney and Darcy, p. 5417.


Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/metabolismo , Terapia Viral Oncolítica , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno CTLA-4/antagonistas & inibidores , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Imunidade Celular , Imunomodulação/efeitos dos fármacos , Imunoterapia/métodos , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Camundongos , Terapia de Alvo Molecular , Neoplasias/diagnóstico , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Carga Tumoral/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/genética , Replicação Viral/efeitos dos fármacos
12.
J Clin Invest ; 125(10): 3915-27, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26348895

RESUMO

Optical imaging of whole, living animals has proven to be a powerful tool in multiple areas of preclinical research and has allowed noninvasive monitoring of immune responses, tumor and pathogen growth, and treatment responses in longitudinal studies. However, fluorescence-based studies in animals are challenging because tissue absorbs and autofluoresces strongly in the visible light spectrum. These optical properties drive development and use of fluorescent labels that absorb and emit at longer wavelengths. Here, we present a far-red absorbing fluoromodule-based reporter/probe system and show that this system can be used for imaging in living mice. The probe we developed is a fluorogenic dye called SC1 that is dark in solution but highly fluorescent when bound to its cognate reporter, Mars1. The reporter/probe complex, or fluoromodule, produced peak emission near 730 nm. Mars1 was able to bind a variety of structurally similar probes that differ in color and membrane permeability. We demonstrated that a tool kit of multiple probes can be used to label extracellular and intracellular reporter-tagged receptor pools with 2 colors. Imaging studies may benefit from this far-red excited reporter/probe system, which features tight coupling between probe fluorescence and reporter binding and offers the option of using an expandable family of fluorogenic probes with a single reporter gene.


Assuntos
Compostos de Anilina/análise , Corantes Fluorescentes/análise , Genes Reporter , Microscopia Intravital , Neoplasias Experimentais/ultraestrutura , Imagem Óptica/métodos , Anticorpos de Cadeia Única/análise , Ativação Metabólica , Compostos de Anilina/farmacocinética , Animais , Linhagem Celular , Permeabilidade da Membrana Celular , Cor , Desamino Arginina Vasopressina/farmacologia , Endocitose/efeitos dos fármacos , Fluorescência , Corantes Fluorescentes/farmacocinética , Proteínas de Fluorescência Verde/análise , Células HCT116/transplante , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/química , Neoplasias Peritoneais/química , Neoplasias Peritoneais/ultraestrutura , Receptores de Vasopressinas/análise , Receptores de Vasopressinas/genética , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/metabolismo , Relação Estrutura-Atividade , Frações Subcelulares/metabolismo , Frações Subcelulares/ultraestrutura , Transdução Genética
13.
Nat Med ; 21(5): 530-6, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25894825

RESUMO

Tumors are complex ecosystems composed of networks of interacting 'normal' and malignant cells. It is well recognized that cytokine-mediated cross-talk between normal stromal cells, including cancer-associated fibroblasts (CAFs), vascular endothelial cells, immune cells, and cancer cells, influences all aspects of tumor biology. Here we demonstrate that the cross-talk between CAFs and cancer cells leads to enhanced growth of oncolytic virus (OV)-based therapeutics. Transforming growth factor-ß (TGF-ß) produced by tumor cells reprogrammed CAFs, dampened their steady-state level of antiviral transcripts and rendered them sensitive to virus infection. In turn, CAFs produced high levels of fibroblast growth factor 2 (FGF2), initiating a signaling cascade in cancer cells that reduced retinoic acid-inducible gene I (RIG-I) expression and impeded the ability of malignant cells to detect and respond to virus. In xenografts derived from individuals with pancreatic cancer, the expression of FGF2 correlated with the susceptibility of the cancer cells to OV infection, and local application of FGF2 to resistant tumor samples sensitized them to virotherapy both in vitro and in vivo. An OV engineered to express FGF2 was safe in tumor-bearing mice, showed improved therapeutic efficacy compared to parental virus and merits consideration for clinical testing.


Assuntos
Fibroblastos/metabolismo , Vírus Oncolíticos/metabolismo , Microambiente Tumoral , Idoso , Animais , Antivirais/química , Linhagem Celular Tumoral , Chlorocebus aethiops , Técnicas de Cocultura , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Microscopia de Fluorescência , Pessoa de Meia-Idade , Transplante de Neoplasias , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Vero
14.
Oncolytic Virother ; 3: 1-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27512659

RESUMO

The field of oncolytic virology has made great strides in recent years. However, one key finding has been that the use of viral agents that replicate selectively in tumors is usually insufficient to achieve anything beyond small and transient responses. Instead, like most cancer therapies, oncolytic viruses are most effective in combination with other therapies, which is where they have proven therapeutic effects in clinical and preclinical studies. In cases of some of the smaller RNA viruses, effects can only be achieved through combination regimens with chemotherapy, radiotherapy, or targeted conventional therapies. However, larger DNA viruses are able to express one or more transgenes; thus, therapeutic mechanisms can be built into the viral vector itself. The incorporated approaches into arming oncolytic viruses through transgene expression will be the main focus of this review, including use of immune activators, prodrug converting enzymes, anti-angiogenic factors, and targeting of the stroma. This will focus on poxviruses as model systems with large cloning capacities, which have routinely been used as transgene expression vectors in different settings, including vaccine and oncolytic viral therapy.

15.
Neoplasia ; 14(12): 1115-21, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23308044

RESUMO

Promising phase II clinical results have been reported recently for several oncolytic viral therapeutics, including strains based on vaccinia virus. One reason for this has been an increased appreciation of the critical therapeutic importance of the immune response raised by these viruses. However, the most commonly used approaches to enhance these immunotherapeutic effects in oncolytic viruses, typically though expression of cytokine transgenes, often also result in a reduction in oncolytic activity and premature clearance of the virotherapy from the tumor. Approaches that enhance the immunotherapeutic effects while maintaining oncolytic activity would therefore be beneficial. Here, it is demonstrated that the expression of the chemokine CCL19 (ELC) from an oncolytic vaccinia virus (vvCCL19) results in increased antitumor effects in syngeneic mouse tumor models. This corresponded with increased t cell and dendritic cell infiltration into the tumor. However, vvCCL19 persisted in the tumor at equivalent levels to a control virus without CCL19, demonstrating that oncolytic activity was not curtailed. Instead, vvCCL19 was cleared rapidly and selectively from normal tissues and organs, indicating a potentially increased safety profile. The therapeutic activity of vvCCL19 could be further significantly increased through combination with adoptive transfer of therapeutic immune cells expressing CCR7, the receptor for CCL19. This approach therefore represents a means to increase the safety and therapeutic benefit of oncolytic viruses, used alone or in combination with immune cell therapies.


Assuntos
Adenocarcinoma/terapia , Quimiocina CCL19/metabolismo , Neoplasias do Colo/terapia , Imunoterapia , Células T Matadoras Naturais/imunologia , Vírus Oncolíticos/metabolismo , Vaccinia virus/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/virologia , Animais , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/virologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Células T Matadoras Naturais/metabolismo , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Receptores CCR7/metabolismo , Baço/metabolismo , Vaccinia virus/genética , Vaccinia virus/fisiologia , Carga Viral
16.
J Immunother ; 34(3): 289-96, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21389869

RESUMO

A variety of immune cell therapies proposed for use in the treatment of cancer, including both autologous cells (Lymphokine Activated Killer, Cytokine Induced Killer) or cell lines (TALL-104, NK-92), rely on recognition of NKG2D ligands on malignant cells for targeting. These ligands, such as MICA and MICB in humans are stress response ligands and are commonly, but not ubiquitously expressed within tumors. Several tumor escape mechanisms have been reported, including ligand downregulation and internalization, or proteolytic cleavage and shedding of their exposed portions (releasing soluble MICA and MICB; sMICA, sMICB). Therefore, an ability to prescreen patients for the level of tumor cell surface expression and shedding of these ligands would prevent needless treatment of patients that are unable to respond, whereas targeted pretreatment of patients to increase surface expression and/or block shedding would enhance the subsequent effectiveness of these therapies. Here, we report that serum tests of sMICA and sMICB in conjunction with tumor measurements might be used to determine rates of shedding from a tumor and that treatment with a selected combination of histone deacetylase inhibitors (to upregulate cell surface MICA/B in some tumors), and metalloproteinase inhibitors (to block MICA/B shedding in others) can be incorporated to regulate cell surface MICA/B levels before immune cell therapy, significantly enhancing their effectiveness (either used alone or as carrier vehicles for oncolytic viruses). Ultimately prescreening patients undergoing such immune cell therapies might be used to personalize cancer treatment regimens based on the NKG2D-ligand status of the tumor.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Imunoterapia , Células Matadoras Naturais/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Animais , Linhagem Celular Tumoral , Feminino , Antígenos de Histocompatibilidade Classe I/sangue , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , Ligantes , Metaloproteases/metabolismo , Camundongos , Camundongos Nus , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Medicina de Precisão/métodos , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Solubilidade , Evasão Tumoral/genética , Evasão Tumoral/imunologia
17.
Cell Host Microbe ; 8(2): 174-85, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20709294

RESUMO

Vaccinia immunization was pivotal to successful smallpox eradication. However, the early immune responses that distinguish poxvirus immunization from pathogenic infection remain unknown. To address this, we developed a strategy to map the activation of key signaling networks in vivo and applied this approach to define and compare the earliest signaling events elicited by immunizing (vaccinia) and lethal (ectromelia) poxvirus infections in mice. Vaccinia induced rapid TLR2-dependent responses, leading to IL-6 production, which then initiated STAT3 signaling in dendritic and T cells. In contrast, ectromelia did not induce TLR2 activation, and profound mouse strain-dependent responses were observed. In resistant C57BL/6 mice, the STAT1 and STAT3 pathways were rapidly activated, whereas in susceptible BALB/c mice, IL-6-dependent STAT3 activation did not occur. These data link early immune signaling events to infection outcome and suggest that activation of different pattern-recognition receptors early after infection may be important in determining vaccine efficacy.


Assuntos
Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Vaccinia virus/imunologia , Vacínia/imunologia , Animais , Linhagem Celular , Células Dendríticas/imunologia , Predisposição Genética para Doença , Especificidade de Hospedeiro/imunologia , Humanos , Imunização , Interleucina-6/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Receptor 2 Toll-Like/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA