Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Methods Mol Biol ; 2058: 295-306, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31486047

RESUMO

Fluorescence in situ hybridization (FISH) is a specific, sensitive, accurate, and reliable technique widely applied in both research and clinic. Here we describe the detailed protocol of a FISH method established by us to serve the scientific purposes of the first oncolytic parvovirus clinical trial (ParvOryx01). This trial was launched in Germany in 2011. After trial completion in 2015, results were published in Molecular Therapy in 2017. The primary purpose of the trial was to evaluate the safety of an oncolytic parvovirus, H-1PV (ParvOryx), in recurrent glioblastoma patients. In addition, the efficiency of H-1PV tumor targeting after intratumoral or systemic virus administration was assessed by FISH detection of viral nucleic acids (genomic single-stranded DNA, mRNA and parvovirus double-stranded replicative forms) in formalin-fixed paraffin-embedded glioblastoma tissues resected at day 10 after ParvOryx treatment. The FISH method allowed the detection-for the first time in humans-of H-1PV replication markers in brain tumors of parvovirus-treated patients. A protocol combining mRNA FISH with simultaneous immunofluorescent staining for tumor and tumor microenvironment markers was also developed and is described here, in order to better characterize H-1PV cellular targets and H-1PV treatment-associated tumor microenvironment changes.


Assuntos
Neoplasias Encefálicas/diagnóstico , DNA Viral , Vetores Genéticos , Parvovirus H-1 , Hibridização in Situ Fluorescente , Vírus Oncolíticos , Neoplasias Encefálicas/terapia , Imunofluorescência , Vetores Genéticos/genética , Parvovirus H-1/genética , Parvovirus H-1/imunologia , Humanos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Microambiente Tumoral , Replicação Viral
2.
Viruses ; 10(4)2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29584637

RESUMO

Single nucleotide changes were introduced into the non-structural (NS) coding sequence of the H-1 parvovirus (PV) infectious molecular clone and the corresponding virus stocks produced, thereby generating H1-PM-I, H1-PM-II, H1-PM-III, and H1-DM. The effects of the mutations on viral fitness were analyzed. Because of the overlapping sequences of NS1 and NS2, the mutations affected either NS2 (H1-PM-II, -III) or both NS1 and NS2 proteins (H1-PM-I, H1-DM). Our results show key benefits of PM-I, PM-II, and DM mutations with regard to the fitness of the virus stocks produced. Indeed, these mutants displayed a higher production of infectious virus in different cell cultures and better spreading capacity than the wild-type virus. This correlated with a decreased particle-to-infectivity (P/I) ratio and stimulation of an early step(s) of the viral cycle prior to viral DNA replication, namely, cell binding and internalization. These mutations also enhance the transduction efficiency of H-1PV-based vectors. In contrast, the PM-III mutation, which affects NS2 at a position downstream of the sequence deleted in Del H-1PV, impaired virus replication and spreading. We hypothesize that the NS2 protein-modified in H1-PM-I, H1-PM-II, and H1-DM-may result in the stimulation of some maturation step(s) of the capsid and facilitate virus entry into subsequently infected cells.


Assuntos
Vetores Genéticos/genética , Parvovirus H-1/fisiologia , Fases de Leitura Aberta/genética , Infecções por Parvoviridae/virologia , Transdução Genética , Proteínas não Estruturais Virais/genética , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Linhagem Celular , DNA Viral/biossíntese , DNA Viral/metabolismo , Parvovirus H-1/genética , Parvovirus H-1/crescimento & desenvolvimento , Humanos , Mutação , Processamento de Proteína Pós-Traducional , Ratos , Proteínas Virais/metabolismo , Ligação Viral , Internalização do Vírus , Liberação de Vírus , Replicação Viral
3.
Mol Ther ; 25(12): 2620-2634, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-28967558

RESUMO

Oncolytic virotherapy may be a means of improving the dismal prognosis of malignant brain tumors. The rat H-1 parvovirus (H-1PV) suppresses tumors in preclinical glioma models, through both direct oncolysis and stimulation of anticancer immune responses. This was the basis of ParvOryx01, the first phase I/IIa clinical trial of an oncolytic parvovirus in recurrent glioblastoma patients. H-1PV (escalating dose) was administered via intratumoral or intravenous injection. Tumors were resected 9 days after treatment, and virus was re-administered around the resection cavity. Primary endpoints were safety and tolerability, virus distribution, and maximum tolerated dose (MTD). Progression-free and overall survival and levels of viral and immunological markers in the tumor and peripheral blood were also investigated. H-1PV treatment was safe and well tolerated, and no MTD was reached. The virus could cross the blood-brain/tumor barrier and spread widely through the tumor. It showed favorable pharmacokinetics, induced antibody formation in a dose-dependent manner, and triggered specific T cell responses. Markers of virus replication, microglia/macrophage activation, and cytotoxic T cell infiltration were detected in infected tumors, suggesting that H-1PV may trigger an immunogenic stimulus. Median survival was extended in comparison with recent meta-analyses. Altogether, ParvOryx01 results provide an impetus for further H-1PV clinical development.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/terapia , Parvovirus H-1/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Feminino , Expressão Gênica , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Radioterapia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Transgenes , Resultado do Tratamento
4.
Cancer Gene Ther ; 22(5): 271-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25857359

RESUMO

Although H-1 parvovirus is used as an antitumor agent, not much is known about the relationship between its specific tropism and oncolytic activity. We hypothesize that VP2, a major capsid protein of H-1 virus, determines H-1-specific tropism. To assess this, we constructed chimeric H-1 viruses expressing Kilham rat virus (KRV) capsid proteins, in their complete or partial forms. Chimeric H-1 viruses (CH1, CH2 and CH3) containing the whole KRV VP2 domain could not induce cytolysis in HeLa, A549 and Panc-1 cells. However, the other chimeric H-1 viruses (CH4 and CH5) expressing a partial KRV VP2 domain induced cytolysis. Additionally, the significant cytopathic effect caused by CH4 and CH5 infection in HeLa cells resulted from preferential viral amplification via DNA replication, RNA transcription and protein synthesis. Modeling of VP2 capsid protein showed that two variable regions (VRs) (VR0 and VR2) of H-1 VP2 protein protrude outward, because of the insertion of extra amino-acid residues, as compared with those of KRV VP2 protein. This might explain the precedence of H-1 VP2 protein over KRV in determining oncolytic activity in human cancer cells. Taking these results together, we propose that the VP2 protein of oncolytic H-1 parvovirus determines its specific tropism in human cancer cells.


Assuntos
Proteínas do Capsídeo/metabolismo , Parvovirus H-1/fisiologia , Neoplasias/virologia , Infecções por Parvoviridae/virologia , Animais , Proteínas do Capsídeo/genética , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Ratos , Transfecção
5.
Curr Opin Virol ; 13: 17-24, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25841215

RESUMO

The H-1 parvovirus (H-1PV) exerts oncosuppressive action that has two components: oncotoxicity and immunostimulation. While many human tumor cells, including conventional drug-resistant ones, can be killed by H-1PV, some fail to support progeny virus production, necessary for infection propagation in neoplastic tissues. This limitation can be overcome through forced selection of H-1PV variants capable of enhanced multiplication and spreading in human tumor cells. In the context of further developing H-1PV for use in cancer therapy, arming it with immunostimulatory CpG motifs under conditions preserving replication and oncolysis enhances its action as an anticancer vaccine adjuvant. A first clinical study of H-1PV treatment in glioma patients has yielded evidence of intratumoral synthesis of the viral oncotoxic protein NS1 and immune cell infiltration.


Assuntos
Alphaherpesvirinae/fisiologia , Parvovirus H-1/fisiologia , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Alphaherpesvirinae/genética , Animais , Parvovirus H-1/genética , Humanos , Neoplasias/imunologia , Neoplasias/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética
6.
Comp Med ; 65(1): 23-35, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25730754

RESUMO

Parvovirus H1 (H1PV) is an autonomous parvovirus that is transmitted in rodent populations. Its natural host is rats. H1PV infection is nonpathogenic except in rat and hamster fetuses and newborns. H1PV infection of human cancer cells caused strong oncolytic effects in preclinical models. For a clinical trial of H1PV in patients with brain tumors, clinical-grade H1PV was produced according to Good Manufacturing Practices. This report focuses on results obtained after a single high-dose intravenous injection of highly purified H1PV in 30 rats and multiple (n = 17) intravenous injections at 3 dose levels in 223 rats. In both studies, no virus-related mortality or macroscopic organ changes related to H1PV occurred. Histopathology after multiple virus injections revealed minimal diffuse bile duct hyperplasia in livers of animals of the highest dose group and germinal center development in spleens of animals from the high-dose group. Liver changes were reversible within a 2-wk recovery period after the last injection. Hematology, blood chemistry, and coagulation analyses did not reveal significant toxicologic changes due to H1PV. Virus injection stimulated the production of IgG antibodies but did not alter mononuclear cell function or induce cytokine release. PCR analysis showed dose-dependent levels of viral genomes in all organs tested. The virus was excreted primarily through feces. These data provide important information regarding H1PV infection in its natural host. Due to the confirmation of the favorable safety profile of H1PV in a permissive animal model, a phase I/IIa clinical trial of H1PV in brain tumor patients could be initiated.


Assuntos
Genoma Viral/genética , Parvovirus H-1/patogenicidade , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/imunologia , Infecções por Parvoviridae/patologia , Animais , Análise Química do Sangue , Testes de Coagulação Sanguínea , Relação Dose-Resposta Imunológica , Avaliação Pré-Clínica de Medicamentos , Fezes/virologia , Parvovirus H-1/genética , Imunoglobulina G/imunologia , Injeções Intravenosas , Fígado/patologia , Reação em Cadeia da Polimerase , Ratos , Baço/patologia
7.
PLoS Pathog ; 9(10): e1003671, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204256

RESUMO

Disassembly of the nuclear lamina is essential in mitosis and apoptosis requiring multiple coordinated enzymatic activities in nucleus and cytoplasm. Activation and coordination of the different activities is poorly understood and moreover complicated as some factors translocate between cytoplasm and nucleus in preparatory phases. Here we used the ability of parvoviruses to induce nuclear membrane breakdown to understand the triggers of key mitotic enzymes. Nuclear envelope disintegration was shown upon infection, microinjection but also upon their application to permeabilized cells. The latter technique also showed that nuclear envelope disintegration was independent upon soluble cytoplasmic factors. Using time-lapse microscopy, we observed that nuclear disassembly exhibited mitosis-like kinetics and occurred suddenly, implying a catastrophic event irrespective of cell- or type of parvovirus used. Analyzing the order of the processes allowed us to propose a model starting with direct binding of parvoviruses to distinct proteins of the nuclear pore causing structural rearrangement of the parvoviruses. The resulting exposure of domains comprising amphipathic helices was required for nuclear envelope disintegration, which comprised disruption of inner and outer nuclear membrane as shown by electron microscopy. Consistent with Ca⁺⁺ efflux from the lumen between inner and outer nuclear membrane we found that Ca⁺⁺ was essential for nuclear disassembly by activating PKC. PKC activation then triggered activation of cdk-2, which became further activated by caspase-3. Collectively our study shows a unique interaction of a virus with the nuclear envelope, provides evidence that a nuclear pool of executing enzymes is sufficient for nuclear disassembly in quiescent cells, and demonstrates that nuclear disassembly can be uncoupled from initial phases of mitosis.


Assuntos
Sinalização do Cálcio , Caspase 3/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Parvovirus H-1/metabolismo , Mitose , Membrana Nuclear/enzimologia , Infecções por Parvoviridae/enzimologia , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Caspase 3/genética , Quinase 2 Dependente de Ciclina/genética , Parvovirus H-1/genética , Células HeLa , Humanos , Membrana Nuclear/genética , Membrana Nuclear/patologia , Membrana Nuclear/virologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Proteína Quinase C/genética , Xenopus laevis
8.
PLoS One ; 8(8): e70594, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967078

RESUMO

Peritoneal carcinomatosis is common in advanced pancreatic cancer. Despite current standard treatment, patients with this disease until recently were considered incurable. Cancer gene therapy using oncolytic viruses have generated much interest over the past few years. Here, we investigated a new gene directed enzyme prodrug therapy (GDEPT) approach for an oncosuppressive virotherapy strategy using parvovirus H1 (PV-H1) which preferentially replicates and kills malignant cells. Although, PV-H1 is not potent enough to destroy tumors, it represents an attractive vector for cancer gene therapy. We therefore sought to determine whether the suicide gene/prodrug system, yCD/5-FC could be rationally combined to PV-H1 augmenting its intrinsic oncolytic activity for pancreatic cancer prevention and treatment. We showed that the engineered recombinant parvovirus rPVH1-yCD with 5-FC treatment increased significantly the intrinsic cytotoxic effect and resulted in potent induction of apoptosis and tumor growth inhibition in chemosensitive and chemoresistant cells. Additionally, the suicide gene-expressing PV-H1 infection reduced significantly the constitutive activities of NFκB and Akt/PI3K. Combination of their pharmacological inhibitors (MG132 and LY294002) with rPVH1-yCD/5-FC resulted in substantial increase of antitumor activity. In vivo, high and sustained expression of NS1 and yCD was observed in the disseminated tumor nodules and absent in normal tissues. Treatment of mice bearing intraperitoneal pancreatic carcinomatosis with rPVH1-yCD/5-FC resulted in a drastic inhibition of tumor cell spreading and subsequent increase in long-term survival. Together, the presented data show the improved oncolytic activity of wPV-H1 by yCD/5-FC and thus provides valuable effective and promising virotherapy strategy for prevention of tumor recurrence and treatment. In the light of this study, the suicide gene parvovirotherapy approach represents a new weapon in the war against pancreatic cancer. Moreover, these preliminary accomplishments are opening new field for future development of new combined targeted therapies to have a meaningful impact on advanced cancer.


Assuntos
Citosina Desaminase/genética , Flucitosina/metabolismo , Genes Transgênicos Suicidas/genética , Parvovirus H-1/genética , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citosina Desaminase/metabolismo , DNA Recombinante/genética , Feminino , Parvovirus H-1/fisiologia , Humanos , Camundongos , NF-kappa B/metabolismo , Terapia Neoadjuvante , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/virologia , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Fármacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Leveduras/enzimologia
9.
J Virol ; 87(9): 5128-40, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23449783

RESUMO

The structure of single-stranded DNA (ssDNA) packaging H-1 parvovirus (H-1PV), which is being developed as an antitumor gene delivery vector, has been determined for wild-type (wt) virions and noninfectious (empty) capsids to 2.7- and 3.2-Å resolution, respectively, using X-ray crystallography. The capsid viral protein (VP) structure consists of an α-helix and an eight-stranded anti-parallel ß-barrel with large loop regions between the strands. The ß-barrel and loops form the capsid core and surface, respectively. In the wt structure, 600 nucleotides are ordered in an interior DNA binding pocket of the capsid. This accounts for ∼12% of the H-1PV genome. The wt structure is identical to the empty capsid structure, except for side chain conformation variations at the nucleotide binding pocket. Comparison of the H-1PV nucleotides to those observed in canine parvovirus and minute virus of mice, two members of the genus Parvovirus, showed both similarity in structure and analogous interactions. This observation suggests a functional role, such as in capsid stability and/or ssDNA genome recognition for encapsulation. The VP structure differs from those of other parvoviruses in surface loop regions that control receptor binding, tissue tropism, pathogenicity, and antibody recognition, including VP sequences reported to determine tumor cell tropism for oncotropic rodent parvoviruses. These structures of H-1PV provide insight into structural features that dictate capsid stabilization following genome packaging and three-dimensional information applicable for rational design of tumor-targeted recombinant gene delivery vectors.


Assuntos
Capsídeo/química , Parvovirus H-1/química , Vírion/química , Sequência de Aminoácidos , Capsídeo/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Cristalografia por Raios X , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Vírion/genética , Vírion/metabolismo
10.
J Virol ; 86(14): 7554-64, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22553326

RESUMO

An in-frame, 114-nucleotide-long deletion that affects the NS-coding sequence was created in the infectious molecular clone of the standard parvovirus H-1PV, thereby generating Del H-1PV. The plasmid was transfected and further propagated in permissive human cell lines in order to analyze the effects of the deletion on virus fitness. Our results show key benefits of this deletion, as Del H-1PV proved to exhibit (i) higher infectivity (lower particle-to-infectivity ratio) in vitro and (ii) enhanced tumor growth suppression in vivo compared to wild-type H-1PV. This increased infectivity correlated with an accelerated egress of Del H-1PV progeny virions in producer cells and with an overall stimulation of the viral life cycle in subsequently infected cells. Indeed, virus adsorption and internalization were significantly improved with Del H-1PV, which may account for the earlier appearance of viral DNA replicative forms that was observed with Del H-1PV than wild-type H-1PV. We hypothesize that the internal deletion within the NS2 and/or NS1 protein expressed by Del H-1PV results in the stimulation of some step(s) of the viral life cycle, in particular, a maturation step(s), leading to more efficient nuclear export of infectious viral particles and increased fitness of the virus produced.


Assuntos
Parvovirus H-1/genética , Parvovirus H-1/patogenicidade , Infecções por Parvoviridae/patologia , Deleção de Sequência , Proteínas não Estruturais Virais/genética , Linhagem Celular , Parvovirus H-1/imunologia , Humanos , Infecções por Parvoviridae/virologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ligação Viral , Internalização do Vírus , Liberação de Vírus
11.
BMC Cancer ; 12: 99, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22436661

RESUMO

BACKGROUND: The treatment of patients with malignant brain tumors remains a major oncological problem. The median survival of patients with glioblastoma multiforme (GBM), the most malignant type, is only 15 months after initial diagnosis and even less after tumor recurrence. Improvements of standard treatment including surgery and radio-chemotherapy have not lead to major improvements. Therefore, alternative therapeutics such as oncolytic viruses that specifically target and destroy cancer cells are under investigation. Preclinical data of oncolytic parvovirus H-1 (H-1PV) infection of glioma cells demonstrated strong cytotoxic and oncosuppressing effects, leading to a phase I/IIa trial of H-1PV in patients with recurrent GBM (ParvOryx01). ParvOryx01 is the first trial with a replication competent oncolytic virus in Germany. METHODS: ParvOryx01 is an open, non-controlled, two groups, intra-group dose escalation, single center, phase I/IIa trial. 18 patients with recurrent GBM will be treated in 2 groups of 9 patients each. Treatment group 1 will first receive H-1PV by intratumoral injection and second by administration into the walls of the tumor cavity during tumor resection. In treatment group 2 the virus will initially be injected intravenously and afterwards, identical to group 1, into the surrounding brain tissue during tumor removal. Main eligibility criteria are: age of 18 years, unifocal recurrent GBM, amenable to complete or subtotal resection. Dose escalation will be based on the Continual Reassessment Method. The primary objective of the trial is local and systemic safety and tolerability and to determine the maximum tolerated dose (MTD). Secondary objectives are proof of concept (PoC) and Progression-free Survival (PFS) up to 6 months. DISCUSSION: This is the first trial with H-1PV in patients with recurrent GBM. The risks for the participants appear well predictable and justified. Furthermore, ParvOryx01 will be the first assessment of combined intratumoral and intravenous application of an oncolytic virus. Due to its study design the trial will not only generate data on the local effect of H-1PV but it will also investigate the penetration of H-1PV into the tumor after systemic delivery and obtain safety data from systemic delivery possibly supporting clinical trials with H-1PV in other, non-CNS malignancies. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01301430.


Assuntos
Neoplasias Encefálicas/terapia , Vetores Genéticos/administração & dosagem , Glioblastoma/terapia , Parvovirus H-1/genética , Vírus Oncolíticos/genética , Administração Intravenosa , Neoplasias Encefálicas/patologia , Protocolos Clínicos , Progressão da Doença , Feminino , Glioblastoma/patologia , Humanos , Injeções Intralesionais , Masculino , Terapia Viral Oncolítica , Recidiva
12.
Cancer Biol Ther ; 12(10): 888-95, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22024742

RESUMO

Oncolytic viruses with their capacity to specifically replicate in and kill tumor cells emerged as a novel class of cancer therapeutics. Rat oncolytic parvovirus (H-1PV) was used to treat different types of cancer in preclinical settings and was lately successfully combined with standard gemcitabine chemotherapy in treating pancreatic ductal adenocarcinoma (PDAC) in rats. Our previous work showed that the immune system and particularly the release of interferon-gamma (IFNγ) seem to mediate the anticancer effect of H-1PV in that model. Therefore, we reasoned that the therapeutic properties of H-1PV can be boosted with IFNγ for the treatment of late incurable stages of PDAC like peritoneal carcinomatosis. Rats bearing established orthotopic pancreatic carcinomas with peritoneal metastases were treated with a single intratumoral (i.t.) or intraperitoneal (i.p.) injection of 5 x 108 plaque forming units of H-1PV with or without concomitant IFNγ application. Intratumoral injection proved to be more effective than the intraperitoneal route in controlling the growth of both the primary pancreatic tumors and peritoneal carcinomatosis, accompanied by migration of virus from primary to metastatic deposits. Concomitant i.p. treatment of H-1PV with recIFNγ resulted in improved therapeutic effect yielding an extended animal survival, compared with i.p. treatment with H-1PV alone. IFNγ application enhanced the H-1PV-induced peritoneal macrophage and splenocyte responses against tumor cells while causing a significant reduction in the titers of H1-PV-neutralising antibodies in ascitic fluid. Thus, IFNγ co-application together with H-1PV might be considered as a novel therapeutic option to improve the survival of PDAC patients with peritoneal carcinomatosis.


Assuntos
Carcinoma Ductal Pancreático/terapia , Parvovirus H-1/genética , Interferon gama/uso terapêutico , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/patologia , Neoplasias Peritoneais/terapia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Líquido Ascítico/imunologia , Carcinoma Ductal Pancreático/secundário , Linhagem Celular Tumoral , Citocinas/biossíntese , Terapia Genética , Vetores Genéticos/genética , Humanos , Imunidade Inata/imunologia , Imunomodulação , Interferon gama/genética , Macrófagos/imunologia , Neoplasias Pancreáticas/terapia , Neoplasias Peritoneais/secundário , Ratos , Ratos Endogâmicos Lew
13.
Clin Cancer Res ; 17(16): 5333-42, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21715567

RESUMO

PURPOSE: In previous studies, we have shown that the apathogenic rat parvovirus H-1 (H-1PV) is capable to induce regression of advanced symptomatic rat and human gliomas in a rat model, when the virus was injected in the tumor (intracranially) or intravenously. Infection with H-1PV did not provoke any pathology in nontumor tissue. This study addresses the question whether also intranasal application of this oncolytic virus is suitable and sufficient for treating gliomas in this animal model. EXPERIMENTAL DESIGN: Rat (RG-2) or human (U87) glioma cells were grafted stereotactically in the brain of rats (Wistar or RNU, respectively), and after development of tumors visible by MRI, H-1PV was instilled intranasally. Tumor regression was monitored by MRI, and survival was analyzed by Kaplan-Meier analysis. Brains from sacrificed animals were analyzed for histologic alterations, presence of viral DNA and proteins and infectious virions. In addition, distribution of virus to other organs was determined. RESULTS: A single intranasal instillation of H-1PV was sufficient to induce efficient regression of rat glioma, leading to significant prolongation of survival without any toxicity for other tissues. It is shown that the virus reaches brain and other tissues, and that the viral replication-associated (and oncolysis-associated) regulatory proteins are exclusively expressed in the tumor tissue. In rats with xenografts of human glioma, oncolytic activity of H-1PV was less pronounced, however, leading to significant prolongation of survival. CONCLUSION: In view of an ongoing clinical trial on the use of H-1PV for oncolytic virotherapy of glioma, the option of applying the virus intranasally may be a valuable alternative to invasive routes of infection.


Assuntos
Glioma/terapia , Parvovirus H-1/fisiologia , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/terapia , Administração Intranasal , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular Tumoral , DNA Viral/análise , Modelos Animais de Doenças , Feminino , Glioma/patologia , Glioma/virologia , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Resultado do Tratamento , Carga Tumoral , Proteínas Virais/análise , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Virol ; 84(12): 5909-22, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375165

RESUMO

The rat parvovirus H-1 (H-1PV) attracts high attention as an anticancer agent, because it is not pathogenic for humans and has oncotropic and oncosuppressive properties. The viral nonstructural NS1 protein is thought to mediate H-1PV cytotoxicity, but its exact contribution to this process remains undefined. In this study, we analyzed the effects of the H-1PV NS1 protein on human cell proliferation and cell viability. We show that NS1 expression is sufficient to induce the accumulation of cells in G(2) phase, apoptosis via caspase 9 and 3 activation, and cell lysis. Similarly, cells infected with wild-type H-1PV arrest in G(2) phase and undergo apoptosis. Furthermore, we also show that both expression of NS1 and H-1PV infection lead to higher levels of intracellular reactive oxygen species (ROS), associated with DNA double-strand breaks. Antioxidant treatment reduces ROS levels and strongly decreases NS1- and virus-induced DNA damage, cell cycle arrest, and apoptosis, indicating that NS1-induced ROS are important mediators of H-1PV cytotoxicity.


Assuntos
Apoptose , Parvovirus H-1/metabolismo , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas não Estruturais Virais/metabolismo , Ciclo Celular , Linhagem Celular , Dano ao DNA , Parvovirus H-1/genética , Humanos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/virologia , Proteínas não Estruturais Virais/genética
15.
Arch Virol ; 155(5): 771-5, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20237811

RESUMO

When grown in human cell lines, oncolytic H-1 parvovirus (H-1PV) replication preferentially occurs in transformed cells, which ultimately die upon infection. H-1PV-induced cytotoxicity is mainly due to P4 promoter-driven NS1 protein expression. Infection of untransformed cells generally does not induce deleterious effects because the P4 promoter is not activated. Here, we show that ectopic CMV-driven NS1 protein expression in normal human MRC-5 cells results in alterations of actin filaments and cell death, and both effects are prevented by a serine 473 mutation. The same substitution preserves actin filaments of transfected MRC-5 SV2 cells, that are MRC-5 transformed counterparts, but does not impair NS1-induced cytotoxicity.


Assuntos
Actinas/metabolismo , Parvovirus H-1/patogenicidade , Proteínas não Estruturais Virais/fisiologia , Morte Celular , Linhagem Celular Transformada , Células Cultivadas , Parvovirus H-1/genética , Humanos , Regiões Promotoras Genéticas , Transfecção , Proteínas não Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA