Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
BMC Cancer ; 20(1): 226, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32183732

RESUMO

BACKGROUND: Simeoni and colleagues introduced a compartmental model for tumor growth that has proved quite successful in modeling experimental therapeutic regimens in oncology. The model is based on a system of ordinary differential equations (ODEs), and accommodates a lag in therapeutic action through delay compartments. There is some ambiguity in the appropriate number of delay compartments, which we examine in this note. METHODS: We devised an explicit delay differential equation model that reflects the main features of the Simeoni ODE model. We evaluated the original Simeoni model and this adaptation with a sample data set of mammary tumor growth in the FVB/N-Tg(MMTVneu)202Mul/J mouse model. RESULTS: The experimental data evinced tumor growth heterogeneity and inter-individual diversity in response, which could be accommodated statistically through mixed models. We found little difference in goodness of fit between the original Simeoni model and the delay differential equation model relative to the sample data set. CONCLUSIONS: One should exercise caution if asserting a particular mathematical model uniquely characterizes tumor growth curve data. The Simeoni ODE model of tumor growth is not unique in that alternative models can provide equivalent representations of tumor growth.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Modelos Biológicos , Algoritmos , Animais , Feminino , Camundongos
2.
Mol Ther ; 22(7): 1320-1332, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24695102

RESUMO

This study characterizes the ability of novel oncolytic rhabdoviruses (Maraba MG1) to boost natural killer (NK) cell activity. Our results demonstrate that MG1 activates NK cells via direct infection and maturation of conventional dendritic cells. Using NK depletion and conventional dendritic cells ablation studies in vivo, we established that both are required for MG1 efficacy. We further explored the efficacy of attenuated MG1 (nonreplicating MG1-UV(2min) and single-cycle replicating MG1-Gless) and demonstrated that these viruses activate conventional dendritic cells, although to a lesser extent than live MG1. This translates to equivalent abilities to remove tumor metastases only at the highest viral doses of attenuated MG1. In tandem, we characterized the antitumor ability of NK cells following preoperative administration of live and attenuated MG1. Our results demonstrates that a similar level of NK activation and reduction in postoperative tumor metastases was achieved with equivalent high viral doses concluding that viral replication is important, but not necessary for NK activation. Biochemical characterization of a panel of UV-inactivated MG1 (2-120 minutes) revealed that intact viral particle and target cell recognition are essential for NK cell-mediated antitumor responses. These findings provide mechanistic insight and preclinical rationale for safe perioperative virotherapy to effectively reduce metastatic disease following cancer surgery.


Assuntos
Células Dendríticas/citologia , Células Matadoras Naturais/citologia , Melanoma/terapia , Rhabdoviridae/fisiologia , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica/métodos
3.
Mol Ther ; 20(6): 1148-57, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22273579

RESUMO

Replicating viruses for the treatment of cancer have a number of advantages over traditional therapeutic modalities. They are highly targeted, self-amplifying, and have the added potential to act as both gene-therapy delivery vehicles and oncolytic agents. Parapoxvirus ovis or Orf virus (ORFV) is the prototypic species of the Parapoxvirus genus, causing a benign disease in its natural ungulate host. ORFV possesses a number of unique properties that make it an ideal viral backbone for the development of a cancer therapeutic: it is safe in humans, has the ability to cause repeat infections even in the presence of antibody, and it induces a potent T(h)-1-dominated immune response. Here, we show that live replicating ORFV induces an antitumor immune response in multiple syngeneic mouse models of cancer that is mediated largely by the potent activation of both cytokine-secreting, and tumoricidal natural killer (NK) cells. We have also highlighted the clinical potential of the virus by demonstration of human cancer cell oncolysis including efficacy in an A549 xenograft model of cancer.


Assuntos
Vetores Genéticos/administração & dosagem , Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Vírus do Orf/imunologia , Animais , Linhagem Celular Tumoral , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Terapia Genética , Vetores Genéticos/efeitos adversos , Humanos , Imunidade Inata , Células Matadoras Naturais/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/genética , Vírus Oncolíticos/genética , Vírus do Orf/genética , Baço/imunologia , Baço/metabolismo , Carga Tumoral , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Ther ; 20(9): 1791-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22760544

RESUMO

Treatment of permissive tumors with the oncolytic virus (OV) VSV-Δ51 leads to a robust antitumor T-cell response, which contributes to efficacy; however, many tumors are not permissive to in vivo treatment with VSV-Δ51. In an attempt to channel the immune stimulatory properties of VSV-Δ51 and broaden the scope of tumors that can be treated by an OV, we have developed a potent oncolytic vaccine platform, consisting of tumor cells infected with VSV-Δ51. We demonstrate that prophylactic immunization with this infected cell vaccine (ICV) protected mice from subsequent tumor challenge, and expression of granulocyte-monocyte colony stimulating factor (GM-CSF) by the virus (VSVgm-ICV) increased efficacy. Immunization with VSVgm-ICV in the VSV-resistant B16-F10 model induced maturation of dendritic and natural killer (NK) cell populations. The challenge tumor is rapidly infiltrated by a large number of interferon γ (IFNγ)-producing T and NK cells. Finally, we demonstrate that this approach is robust enough to control the growth of established tumors. This strategy is broadly applicable because of VSV's extremely broad tropism, allowing nearly all cell types to be infected at high multiplicities of infection in vitro, where the virus replication kinetics outpace the cellular IFN response. It is also personalized to the unique tumor antigen(s) displayed by the cancer cell.


Assuntos
Vacinas Anticâncer/imunologia , Melanoma Experimental/prevenção & controle , Melanoma Experimental/terapia , Neoplasias Cutâneas/prevenção & controle , Neoplasias Cutâneas/terapia , Vesiculovirus/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunização , Interferon gama/biossíntese , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Vero , Vesiculovirus/genética , Replicação Viral
5.
Mol Ther ; 19(5): 886-94, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21364541

RESUMO

Oncolytic viruses (OVs) have been engineered or selected for cancer cell-specific infection however, we have found that following intravenous administration of vesicular stomatitis virus (VSV), tumor cell killing rapidly extends far beyond the initial sites of infection. We show here for the first time that VSV directly infects and destroys tumor vasculature in vivo but leaves normal vasculature intact. Three-dimensional (3D) reconstruction of infected tumors revealed that the majority of the tumor mass lacks significant blood flow in contrast to uninfected tumors, which exhibit relatively uniform perfusion. VSV replication in tumor neovasculature and spread within the tumor mass, initiates an inflammatory reaction including a neutrophil-dependent initiation of microclots within tumor blood vessels. Within 6 hours of intravenous administration of VSV and continuing for at least 24 hours, we observed the initiation of blood clots within the tumor vasculature whereas normal vasculature remained clot free. Blocking blood clot formation with thrombin inhibitors prevented tumor vascular collapse. Our results demonstrate that the therapeutic activity of an OV can go far beyond simple infection and lysis of malignant cells.


Assuntos
Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus da Estomatite Vesicular Indiana , Adenocarcinoma/genética , Animais , Coagulação Sanguínea , Linhagem Celular Tumoral , Proliferação de Células , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos , Trombina/antagonistas & inibidores
6.
Arch Cancer Biol Ther ; 1(1): 18-21, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32728673

RESUMO

BACKGROUND: We have recently introduced a modification of the seminal Simeoni model for tumor growth, the modification entailing the incorporation of delay differential equations into its formulation. We found that the modification was competitive with the Simeoni construct in modeling mammary tumor growth under cisplatin treatment in an animal model. METHODS: In our original study, we had two cohorts of animals: untreated, and treatment with bolus injection of cisplatin on day 0. We here explore how modifications in the cisplatin dosing scheme affect tumor growth in our model. RESULTS: We found that modest fractionation dosing schemes have little ultimate impact on tumor growth. In contrast, metronomic dosing schemes seem quite efficacious, and might yield effective control over tumor progression. CONCLUSIONS: With regard to cisplatin as single agent chemotherapy, a minimum level of drug for a prolonged period of time seems more critical than rapid achievement of a very high dose for a shorter time frame for deterring tumor growth or progression. Exploration of tumor dose schedules with mathematical models can provide valuable insights into potentially effective therapeutic regimens.

7.
Mol Ther ; 16(8): 1437-43, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18560417

RESUMO

Creation of potent oncolytic viruses (OVs) suitable for the clinic may require new strategies in virus design. Replication-competent viruses facilitate a variety of approaches to achieving tumor specificity. Altered expression of microRNAs is a common hallmark of cancer that we demonstrate can be used to alter expression of a potent wild-type viral gene to achieve tumor-specific replication of an engineered vesicular stomatitis virus (VSV). Incorporation of let-7 microRNA complementary sequences within VSV eliminates undesirable replication and associated toxicity in normal cells but permits growth in cancer cells in vitro and in vivo. This is proof of concept that viruses designed to exploit the differential microRNA expression in cancer cells is a viable approach, potentially useful in optimizing oncolytic viral gene expression for maximal antitumor activity and safety.


Assuntos
MicroRNAs/genética , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vesiculovirus/fisiologia , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Feminino , Genoma Viral/genética , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Neoplasias/virologia , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Vesiculovirus/genética , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Ther ; 15(1): 123-30, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164783

RESUMO

Oncolytic viruses capable of tumor-selective replication and cytolysis have shown early promise as cancer therapeutics. However, the host immune system remains a significant obstacle to effective systemic administration of virus in a clinical setting. Here, we demonstrate the severe negative impact of the adaptive immune response on the systemic delivery of oncolytic vesicular stomatitis virus (VSV) in an immune-competent murine tumor model, an effect mediated primarily by the neutralization of injected virions by circulating antibodies. We show that this obstacle can be overcome by administering virus within carrier cells that conceal viral antigen during delivery. Infected cells were delivered to tumor beds and released virus to infect malignant cells while sparing normal tissues. Repeated administration of VSV in carrier cells to animals bearing metastatic tumors greatly improved therapeutic efficacy when compared with naked virion injection. Whole-body molecular imaging revealed that carrier cells derived from solid tumors accumulate primarily in the lungs following intravenous injection, whereas leukemic carriers disseminate extensively throughout the body. Furthermore, xenogeneic cells were equally effective at delivering virus as syngeneic cells. These findings emphasize the importance of establishing cell-based delivery platforms in order to maximize the efficacy of oncolytic therapeutics.


Assuntos
Vírus Oncolíticos/imunologia , Transgenes/genética , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular Tumoral , Feminino , Terapia Genética , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/virologia , Taxa de Sobrevida , Vesiculovirus/imunologia
9.
Sci Rep ; 7(1): 5153, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28698590

RESUMO

Tudor domain containing protein 3 (TDRD3) is a modular protein identified based on its ability to recognize methylated arginine motifs through its Tudor domain. We have previously shown that TDRD3 localizes to cytoplasmic stress granules, a structure shown to promote survival upon treatment with chemotherapeutic drugs in cancer cells. Here, we report TDRD3 as a novel regulator of cell proliferation and invasion in breast cancer cells. Our study also demonstrates that TDRD3 depletion inhibits tumor formation and metastasis to the lung in vivo. Furthermore, we show that TDRD3 regulates the expression of a number of key genes associated with promotion of breast cancer tumorigenesis and disease progression. Strikingly, we report that TDRD3 regulates some of these key targets at the level of translation. These findings provide the first experimental demonstration of a functional role for TDRD3 in promoting breast cancer development and progression, and identify TDRD3 as a potential new therapeutic target for breast cancer.


Assuntos
Neoplasias da Mama/patologia , Neoplasias Pulmonares/secundário , Proteínas/genética , Proteínas/metabolismo , Regulação para Cima , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células MCF-7 , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Biossíntese de Proteínas
10.
Oncotarget ; 6(5): 3013-32, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25605249

RESUMO

Recent evidence points to the protein arginine methyltransferase (PRMT) family of enzymes playing critical roles in cancer. PRMT7 has been identified in several gene expression studies to be associated with increased metastasis and decreased survival in breast cancer patients. However, this has not been extensively studied. Here we report that PRMT7 expression is significantly upregulated in both primary breast tumour tissues and in breast cancer lymph node metastases. We have demonstrated that reducing PRMT7 levels in invasive breast cancer cells using RNA interference significantly decreased cell invasion in vitro and metastasis in vivo. Conversely, overexpression of PRMT7 in non-aggressive MCF7 cells enhanced their invasiveness. Furthermore, we show that PRMT7 induces the expression of matrix metalloproteinase 9 (MMP9), a well-known mediator of breast cancer metastasis. Importantly, we significantly rescued invasion of aggressive breast cancer cells depleted of PRMT7 by the exogenous expression of MMP9. Our results demonstrate that upregulation of PRMT7 in breast cancer may have a significant role in promoting cell invasion through the regulation of MMP9. This identifies PRMT7 as a novel and potentially significant biomarker and therapeutic target for breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Movimento Celular , Metaloproteinase 9 da Matriz/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Linfonodos/enzimologia , Linfonodos/patologia , Metástase Linfática , Metaloproteinase 9 da Matriz/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Proteína-Arginina N-Metiltransferases/genética , Interferência de RNA , Transdução de Sinais , Transfecção , Regulação para Cima
11.
Cancer Cell ; 28(2): 210-24, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26212250

RESUMO

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Assuntos
Neoplasias/virologia , Vírus Oncolíticos/fisiologia , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/virologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Interferência de RNA , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Vaccinia virus/fisiologia
12.
Clin Cancer Res ; 19(14): 3832-43, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23714728

RESUMO

PURPOSE: Acute lymphoblastic leukemia (ALL) remains incurable in most adults. It has been difficult to provide effective immunotherapy to improve outcomes for the majority of patients. Rhabdoviruses induce strong antiviral immune responses. We hypothesized that mice administered ex vivo rhabdovirus-infected ALL cells [immunotherapy by leukemia-oncotropic virus (iLOV)] would develop robust antileukemic immune responses capable of controlling ALL. EXPERIMENTAL DESIGN: Viral protein production, replication, and cytopathy were measured in human and murine ALL cells exposed to attenuated rhabdovirus. Survival following injection of graded amounts of ALL cells was compared between cohorts of mice administered γ-irradiated rhabdovirus-infected ALL cells (iLOV) or multiple control vaccines to determine key immunotherapeutic components and characteristics. Host immune requirements were assessed in immunodeficient and bone marrow-transplanted mice or by adoptive splenocyte transfer from immunized donors. Antileukemic immune memory was ascertained by second leukemic challenge in long-term survivors. RESULTS: Human and murine ALL cells were infected and killed by rhabdovirus; this produced a potent antileukemia vaccine. iLOV protected mice from otherwise lethal ALL by developing durable leukemia-specific immune-mediated responses (P < 0.0001), which required an intact CTL compartment. Preexisting antiviral immunity augmented iLOV potency. Splenocytes from iLOV-vaccinated donors protected 60% of naïve recipients from ALL challenge (P = 0.0001). Injecting leukemia cells activated by, or concurrent with, multiple Toll-like receptor agonists could not reproduce the protective effect of iLOV. Similarly, injecting uninfected irradiated viable, apoptotic, or necrotic leukemia cells with/without concurrent rhabdovirus administration was ineffective. CONCLUSION: Rhabdovirus-infected leukemia cells can be used to produce a vaccine that induces robust specific immunity against aggressive leukemia.


Assuntos
Imunoterapia Adotiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Rhabdoviridae/fisiologia , Transferência Adotiva , Animais , Transplante de Medula Óssea , Vacinas Anticâncer , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Transplante de Neoplasias , Medicina de Precisão , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Células Vero
13.
Cancer Res ; 73(1): 97-107, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23090117

RESUMO

Natural killer (NK) cell clearance of tumor cell emboli following surgery is thought to be vital in preventing postoperative metastases. Using a mouse model of surgical stress, we transferred surgically stressed NK cells into NK-deficient mice and observed enhanced lung metastases in tumor-bearing mice as compared with mice that received untreated NK cells. These results establish that NK cells play a crucial role in mediating tumor clearance following surgery. Surgery markedly reduced NK cell total numbers in the spleen and affected NK cell migration. Ex vivo and in vivo tumor cell killing by NK cells were significantly reduced in surgically stressed mice. Furthermore, secreted tissue signals and myeloid-derived suppressor cell populations were altered in surgically stressed mice. Significantly, perioperative administration of oncolytic parapoxvirus ovis (ORFV) and vaccinia virus can reverse NK cell suppression, which correlates with a reduction in the postoperative formation of metastases. In human studies, postoperative cancer surgery patients had reduced NK cell cytotoxicity, and we show for the first time that oncolytic vaccinia virus markedly increases NK cell activity in patients with cancer. These data provide direct in vivo evidence that surgical stress impairs global NK cell function. Perioperative therapies aimed at enhancing NK cell function will reduce metastatic recurrence and improve survival in surgical cancer patients.


Assuntos
Células Matadoras Naturais/imunologia , Metástase Neoplásica/prevenção & controle , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Células Neoplásicas Circulantes/imunologia , Terapia Viral Oncolítica/métodos , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Animais , Citometria de Fluxo , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Metástase Neoplásica/imunologia , Neoplasias Experimentais/cirurgia , Células Neoplásicas Circulantes/patologia , Vírus Oncolíticos , Estresse Fisiológico/imunologia
14.
Mol Ther ; 15(9): 1686-93, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17579581

RESUMO

Oncolytic viruses (OVs) are selected or designed to eliminate malignancies by direct infection and lysis of cancer cells. In contrast to this concept of direct tumor lysis by viral infection, we observed that a significant portion of the in vivo tumor killing activity of two OVs, vesicular stomatitis virus (VSV) and vaccinia virus is caused by indirect killing of uninfected tumor cells. Shortly after administering the oncolytic virus we observed limited virus infection, coincident with a loss of blood flow to the interior of the tumor that correlated with induction of apoptosis in tumor cells. Transcript profiling of tumors showed that virus infection resulted in a dramatic transcriptional activation of pro-inflammatory genes including the neutrophil chemoattractants CXCL1 and CXCL5. Immunohistochemical examination of infected tumors revealed infiltration by neutrophils correlating with chemokine induction. Depletion of neutrophils in animals prior to VSV administration eliminated uninfected tumor cell apoptosis and permitted more extensive replication and spreading of the virus throughout the tumor. Taken all together, these results indicate that targeted recruitment of neutrophils to infected tumor beds enhances the killing of malignant cells. We propose that activation of inflammatory cells can be used for enhancing the effectiveness of oncolytic virus therapeutics, and that this approach should influence the planning of therapeutic doses.


Assuntos
Inflamação/terapia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Apoptose/fisiologia , Velocidade do Fluxo Sanguíneo , Linhagem Celular Tumoral , Imunofluorescência , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/genética , Neoplasias/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA