Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Appl Microbiol Biotechnol ; 107(15): 4777-4787, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37209160

RESUMO

The oncolytic rodent protoparvovirus H-1PV has been successfully used in phase I/II clinical trials to treat recurrent glioblastoma multiforme and pancreatic cancer. The present work focuses on the stability and environmental safety of the H-1PV drug product from production up to its use in patients. We identified hold-steps in manufacturing for up to 3 months and showed 7-years stability for the optimal product formulation. Stress testing via UV, temperature, and pH also determined that the drug product is stable. De- and rehydration for lyophilization simulation are possible without infectious virus loss. Furthermore, we prove in-use stability for 4 days at room temperature and show no virus adsorption to injection devices, guaranteeing the correct administration dose. Iodixanol in the formulation, resulting in high viscosity, protects H-1PV against UV and some disinfectants. Nonetheless, H-1PV is depleted with rapid heat deactivation, autoclavation, and nanofiltration. Assessment of chemical disinfectants that are currently recommended by the Robert Koch-Institute demonstrated that ethanol-based hand disinfectants are not effective; however, aldehyde-based disinfectants for surfaces and instruments demonstrate sufficient H-1PV deactivation in aqueous formulations by 4 to 6 log10. With these results, we could establish a specific hygiene plan for all involved facilities from manufacturing to patient application. Overall, using 48% Iodixanol in Visipaque/Ringer as a drug formulation stabilizes H-1PV infectivity over years and protects against virus loss from short-term UV, low pH, and temperature exposure. KEY POINTS: • Optimal formulation of drug product protects the H-1PV protoparvovirus against UV, temperatures up to 50 °C, and low pH (> 1.25), stabilizing the virus during manufacturing, storage, transport, and application. • H-1PV is stable during in-use and does not adsorb to injection devices during patient administration. • Hygiene plan for H-1PV with physicochemical methods has been established.


Assuntos
Glioblastoma , Parvovirus H-1 , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Pancreáticas , Humanos , Vírus Oncolíticos/fisiologia , Terapia Viral Oncolítica/métodos , Parvovirus H-1/fisiologia , Neoplasias Pancreáticas/terapia
2.
Viruses ; 14(5)2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35632759

RESUMO

Clinical studies in glioblastoma and pancreatic carcinoma patients strongly support the further development of H-1 protoparvovirus (H-1PV)-based anticancer therapies. The identification of cellular factors involved in the H-1PV life cycle may provide the knowledge to improve H-1PV anticancer potential. Recently, we showed that sialylated laminins mediate H-1PV attachment at the cell membrane. In this study, we revealed that H-1PV also interacts at the cell surface with galectin-1 and uses this glycoprotein to enter cancer cells. Indeed, knockdown/out of LGALS1, the gene encoding galectin-1, strongly decreases the ability of H-1PV to infect and kill cancer cells. This ability is rescued by the re-introduction of LGALS1 into cancer cells. Pre-treatment with lactose, which is able to bind to galectins and modulate their cellular functions, decreased H-1PV infectivity in a dose dependent manner. In silico analysis reveals that LGALS1 is overexpressed in various tumours including glioblastoma and pancreatic carcinoma. We show by immunohistochemistry analysis of 122 glioblastoma biopsies that galectin-1 protein levels vary between tumours, with levels in recurrent glioblastoma higher than those in primary tumours or normal tissues. We also find a direct correlation between LGALS1 transcript levels and H-1PV oncolytic activity in 53 cancer cell lines from different tumour origins. Strikingly, the addition of purified galectin-1 sensitises poorly susceptible GBM cell lines to H-1PV killing activity by rescuing cell entry. Together, these findings demonstrate that galectin-1 is a crucial determinant of the H-1PV life cycle.


Assuntos
Galectina 1 , Glioblastoma , Parvovirus H-1 , Terapia Viral Oncolítica , Vírus Oncolíticos , Linhagem Celular Tumoral , Galectina 1/genética , Galectina 1/metabolismo , Glioblastoma/terapia , Parvovirus H-1/fisiologia , Humanos , Recidiva Local de Neoplasia , Vírus Oncolíticos/fisiologia , Neoplasias Pancreáticas , Neoplasias Pancreáticas
3.
Viruses ; 10(6)2018 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-29865280

RESUMO

About 70% of all Ewing sarcoma (EWS) patients are diagnosed under the age of 20 years. Over the last decades little progress has been made towards finding effective treatment approaches for primarily metastasized or refractory Ewing sarcoma in young patients. Here, in the context of the search for novel therapeutic options, the potential of oncolytic protoparvovirus H-1 (H-1PV) to treat Ewing sarcoma was evaluated, its safety having been proven previously tested in adult cancer patients and its oncolytic efficacy demonstrated on osteosarcoma cell cultures. The effects of viral infection were tested in vitro on four human Ewing sarcoma cell lines. Notably evaluated were effects of the virus on the cell cycle and its replication efficiency. Within 24 h after infection, the synthesis of viral proteins was induced. Efficient H-1PV replication was confirmed in all four Ewing sarcoma cell lines. The cytotoxicity of the virus was determined on the basis of cytopathic effects, cell viability, and cell lysis. These in vitro experiments revealed efficient killing of Ewing sarcoma cells by H-1PV at a multiplicity of infection between 0.1 and 5 plaque forming units (PFU)/cell. In two of the four tested cell lines, significant induction of apoptosis by H-1PV was observed. H-1PV thus meets all the in vitro criteria for a virus to be oncolytic towards Ewing sarcoma. In the first xenograft experiments, however, although an antiproliferative effect of intratumoral H-1PV injection was observed, no significant improvement of animal survival was noted. Future projects aiming to validate parvovirotherapy for the treatment of pediatric Ewing sarcoma should focus on combinatorial treatments and will require the use of patient-derived xenografts and immunocompetent syngeneic animal models.


Assuntos
Apoptose , Parvovirus H-1/fisiologia , Terapia Viral Oncolítica , Sarcoma de Ewing/terapia , Sarcoma de Ewing/virologia , Animais , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos , Camundongos Nus , Vírus Oncolíticos/fisiologia , Parvovirus , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
4.
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
5.
Viruses ; 9(10)2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29039746

RESUMO

Osteosarcoma is the most frequent malignant disease of the bone. On the basis of early clinical experience in the 1960s with H-1 protoparvovirus (H-1PV) in osteosarcoma patients, this effective oncolytic virus was selected for systematic preclinical testing on various osteosarcoma cell cultures. A panel of five human osteosarcoma cell lines (CAL 72, H-OS, MG-63, SaOS-2, U-2OS) was tested. Virus oncoselectivity was confirmed by infecting non-malignant human neonatal fibroblasts and osteoblasts used as culture models of non-transformed mesenchymal cells. H-1PV was found to enter osteosarcoma cells and to induce viral DNA replication, transcription of viral genes, and translation to viral proteins. After H-1PV infection, release of infectious viral particles from osteosarcoma cells into the supernatant indicated successful viral assembly and egress. Crystal violet staining revealed progressive cytomorphological changes in all osteosarcoma cell lines. Infection of osteosarcoma cell lines with the standard H-1PV caused an arrest of the cell cycle in the G2 phase, and these lines had a limited capacity for standard H-1PV virus replication. The cytotoxicity of wild-type H-1PV virus towards osteosarcoma cells was compared in vitro with that of two variants, Del H-1PV and DM H-1PV, previously described as fitness variants displaying higher infectivity and spreading in human transformed cell lines of different origins. Surprisingly, wild-type H-1PV displayed the strongest cytostatic and cytotoxic effects in this analysis and thus seems the most promising for the next preclinical validation steps in vivo.


Assuntos
Morte Celular , Parvovirus H-1/fisiologia , Vírus Oncolíticos/fisiologia , Osteossarcoma/patologia , Osteossarcoma/virologia , Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Terapia Viral Oncolítica , Replicação Viral
6.
BMC Cancer ; 17(1): 576, 2017 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-28851316

RESUMO

BACKGROUND: Metastatic pancreatic cancer has a dismal prognosis, with a mean six-month progression-free survival of approximately 50% and a median survival of about 11 months. Despite intensive research, only slight improvements of clinical outcome could be achieved over the last decades. Hence, new and innovative therapeutic strategies are urgently required. ParvOryx is a drug product containing native parvovirus H-1 (H-1PV). Since H-1PV was shown to exert pronounced anti-neoplastic effects in pre-clinical models of pancreatic cancer, the drug appears to be a promising candidate for treatment of this malignancy. METHODS: ParvOryx02 is a non-controlled, single arm, open label, dose-escalating, single center trial. In total seven patients with pancreatic cancer showing at least one hepatic metastasis are to be treated with escalating doses of ParvOryx according to the following schedule: i) 40% of the total dose infused intravenously in equal fractions on four consecutive days, ii) 60% of the total dose injected on a single occasion directly into the hepatic metastasis at varying intervals after intravenous infusions. The main eligibility criteria are: age ≥ 18 years, disease progression despite first-line chemotherapy, and at least one hepatic metastasis. Since it is the second trial within the drug development program, the study primarily explores safety and tolerability after further dose escalation of ParvOryx. The secondary objectives are related to the evaluation of certain aspects of anti-tumor activity and clinical efficacy of the drug. DISCUSSION: This trial strongly contributes to the clinical development program of ParvOryx. The individual hazards for patients included in the current study and the environmental risks are addressed and counteracted adequately. Besides information on safety and tolerability of the treatment after further dose escalation, thorough evaluations of pharmacokinetics and intratumoral spread as well as proof-of-concept (PoC) in pancreatic cancer will be gained in the course of the trial. TRIAL REGISTRATION: ClinicalTrials.gov-ID: NCT02653313 , Registration date: Dec. 4th, 2015.


Assuntos
Parvovirus H-1/fisiologia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/tratamento farmacológico , Administração Intravenosa , Relação Dose-Resposta a Droga , Feminino , Humanos , Injeções Intralesionais , Masculino , Metástase Neoplásica , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/fisiologia , Tamanho da Amostra , Análise de Sobrevida , Resultado do Tratamento
7.
Expert Opin Biol Ther ; 16(5): 645-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26855087

RESUMO

INTRODUCTION: Toolan's H-1 parvovirus (H-1PV) exerts a cytotoxic/oncolytic effect, predominantly mediated by its non-structural protein (NS1). This rat parvovirus is harmless, unlike other parvoviruses, and its antitumor potential may be useful to clinicians as its oncolytic action appears to be true in numerous non-digestive and digestive cancers. AREAS COVERED: After a brief review of parvovirus genus and biology, we summarize the proposed mechanisms to explain the cytotoxicity of H-1PV to tumors which results in dysregulation of cell transcription, cell-cycle arrest, termination of cell replication, activation of cellular stress response and induction of cell death. Viral oncolysis induces a strong tumor-specific immune response leading to the recognition and elimination of minimal residual disease. As the action of H-1PV is not limited to the digestive tract, we initially analyse studies performed in non-digestive cancers such as glioma (as the virus is able to cross the blood brain barrier), and then focused more particularly on the results in digestive cancers. EXPERT OPINION: Based on the results of studies showing little H-1PV toxicity to living bodies, we advocate for the use of the parvovirus in cancers such as melanoma, glioma and pancreatic ductal adenocarcinoma in addition to conventional chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias do Sistema Digestório/terapia , Parvovirus H-1 , Terapia Viral Oncolítica/métodos , Animais , Morte Celular , Parvovirus H-1/fisiologia , Humanos
8.
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
9.
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
10.
Int J Cancer ; 134(3): 703-16, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23852775

RESUMO

Based on extensive pre-clinical studies, the oncolytic parvovirus H-1 (H-1PV) is currently applied to patients with recurrent glioblastoma in a phase I/IIa clinical trial (ParvOryx01, NCT01301430). Cure rates of about 40% in pediatric high-risk medulloblastoma (MB) patients also indicate the need of new therapeutic approaches. In order to prepare a future application of oncolytic parvovirotherapy to MB, the present study preclinically evaluates the cytotoxic efficacy of H-1PV on MB cells in vitro and characterizes cellular target genes involved in this effect. Six MB cell lines were analyzed by whole genome oligonucleotide microarrays after treatment and the results were matched to known molecular and cytogenetic risk factors. In contrast to non-transformed infant astrocytes and neurons, in five out of six MB cell lines lytic H-1PV infection and efficient viral replication could be demonstrated. The cytotoxic effects induced by H-1PV were observed at LD50s below 0.05 p. f. u. per cell indicating high susceptibility. Gene expression patterns in the responsive MB cell lines allowed the identification of candidate target genes mediating the cytotoxic effects of H-1PV. H-1PV induced down-regulation of key regulators of early neurogenesis shown to confer poor prognosis in MB such as ZIC1, FOXG1B, MYC, and NFIA. In MB cell lines with genomic amplification of MYC, expression of MYC was the single gene most significantly repressed after H-1PV infection. H-1PV virotherapy may be a promising treatment approach for MB since it targets genes of functional relevance and induces cell death at very low titers of input virus.


Assuntos
Parvovirus H-1/fisiologia , Meduloblastoma/terapia , Neurogênese , Terapia Viral Oncolítica , Linhagem Celular Tumoral , Regulação Viral da Expressão Gênica , Humanos , Meduloblastoma/genética , Meduloblastoma/patologia , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica , Replicação Viral
11.
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
12.
BMC Cancer ; 11: 464, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22029859

RESUMO

BACKGROUND: Parvovirus H-1 (H-1PV) infects and lyses human tumor cells including melanoma, hepatoma, gastric, colorectal, cervix and pancreatic cancers. We assessed whether the beneficial effects of chemotherapeutic agents or targeted agents could be combined with the oncolytic and immunostimmulatory properties of H-1PV. METHODS: Using human ex vivo models we evaluated the biological and immunological effects of H-1PV-induced tumor cell lysis alone or in combination with chemotherapeutic or targeted agents in human melanoma cells +/- characterized human cytotoxic T-cells (CTL) and HLA-A2-restricted dendritic cells (DC). RESULTS: H-1PV-infected MZ7-Mel cells showed a clear reduction in cell viability of >50%, which appeared to occur primarily through apoptosis. This correlated with viral NS1 expression levels and was enhanced by combination with chemotherapeutic agents or sunitinib. Tumor cell preparations were phagocytosed by DC whose maturation was measured according to the treatment administered. Immature DC incubated with H-1PV-induced MZ7-Mel lysates significantly increased DC maturation compared with non-infected or necrotic MZ7-Mel cells. Tumor necrosis factor-α and interleukin-6 release was clearly increased by DC incubated with H-1PV-induced SK29-Mel tumor cell lysates (TCL) and was also high with DC-CTL co-cultures incubated with H-1PV-induced TCL. Similarly, DC co-cultures with TCL incubated with H-1PV combined with cytotoxic agents or sunitinib enhanced DC maturation to a greater extent than cytotoxic agents or sunitinib alone. Again, these combinations increased pro-inflammatory responses in DC-CTL co-cultures compared with chemotherapy or sunitinib alone. CONCLUSIONS: In our human models, chemotherapeutic or targeted agents did not only interfere with the pronounced immunomodulatory properties of H-1PV, but also reinforced drug-induced tumor cell killing. H-1PV combined with cisplatin, vincristine or sunitinib induced effective immunostimulation via a pronounced DC maturation, better cytokine release and cytotoxic T-cell activation compared with agents alone. Thus, the clinical assessment of H-1PV oncolytic tumor therapy not only alone but also in combination strategies is warranted.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Parvovirus H-1/fisiologia , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/virologia , Neoplasias Cutâneas/terapia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada/métodos , Citocinas/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/virologia , Modelos Biológicos , Vírus Oncolíticos/fisiologia , Infecções por Parvoviridae/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/virologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/virologia , Células Tumorais Cultivadas/efeitos dos fármacos , Proteínas Virais/metabolismo
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.
Int J Cancer ; 128(4): 908-19, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20473905

RESUMO

Natural killer (NK) cells play a vital role in the rejection of tumors. Pancreatic ductal adenocarcinoma (PDAC), however, remains a poor prognosis malignancy, due to its resistance to radio- and chemotherapy, and low immunogenicity. We demonstrate here that IL-2-activated human NK cells are able to kill PDAC cells. Currently, novel strategies are being pursued to combat PDAC. In this regard, oncolytic viruses, in addition to killing tumor cells, may also have the potential to augment antitumor immune responses. We found that, besides having an intrinsic oncolytic activity, parvovirus H-1PV is able to enhance NK cell-mediated killing of PDAC cells. Our results show that H-1PV infection of Panc-1 cells increases NK cell capacity to release IFN-γ, TNF-α and MIP-1α/ß. Multiple activating receptors are involved in the NK cell-mediated killing of Panc-1 cells. Indeed, blocking of the natural cytotoxicity receptors-NKp30, 44 and 46 in combination, and NKG2D and DNAM1 alone inhibit the killing of Panc-1 cells. Interestingly, H-1PV infection of Panc-1 cells overcomes the part of inhibitory effects suggesting that parvovirus may induce additional NK cell ligands on Panc-1 cells. The enhanced sensitivity of H-1PV-infected PDAC cells to NK cell-dependent killing could be traced back to the upregulation of the DNAM-1 ligand, CD155 and to the downregulation of MHC class I expression. Our data suggests that NK cells display antitumor potential against PDAC and that H-1PV-based oncolytic immunotherapy could further boost NK cell-mediated immune responses and help to develop a combinatorial therapeutic approach against PDAC.


Assuntos
Carcinoma Ductal Pancreático/imunologia , Parvovirus H-1/fisiologia , Células Matadoras Naturais/imunologia , Vírus Oncolíticos/imunologia , Neoplasias Pancreáticas/imunologia , Infecções por Parvoviridae/imunologia , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/virologia , Citocinas/metabolismo , Citotoxicidade Imunológica/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Células Matadoras Naturais/patologia , Células Matadoras Naturais/virologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/virologia , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Células Tumorais Cultivadas
15.
Cancer Biol Ther ; 10(12): 1280-9, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21124075

RESUMO

Treatment of cancers by means of viruses, that specifically replicate in (oncotropism) and kill (oncolysis) neoplastic cells, is increasingly gaining acceptance in the clinic. Among these agents, parvoviruses have been shown to possess not only direct oncolytic but also immunomodulating properties, serving as an adjuvant to prime the immune system to react against infected tumors. Here, we aimed to establish whether immunomodulating mechanisms participate in the recently reported therapeutic potential of parvoviruses against pancreatic carcinoma. Using adoptive transfer experiments we discovered that the transfer of splenocytes of donor rats harboring H-1PV-treated orthotopic PDAC tumors could significantly prolong the survival of naïve tumor-bearing recipients, compared to those receiving cells from mock-treated donors. Closer investigation of immunological parameters in infected donor rats revealed that virus-induced interferon gamma production and cellular immune response played an important role in this effect. These data have also preclinical relevance since abortive H-1PV infection of human peripheral blood mononuclear cells or cocultivation of these cells with H-1PV-preinfected pancreatic cancer cells, resulted in enhancement of innate and adaptive immune reactivity. Taken together our data reveal that oncolytic H-1PV modulates the immune system into an anticancer state, and further support the concept of using parvoviruses in the fight against pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/terapia , Parvovirus H-1/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Terapia Viral Oncolítica/métodos , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Citometria de Fluxo , Parvovirus H-1/fisiologia , Humanos , Imunidade Celular/imunologia , Imunomodulação , Interferon gama/imunologia , Vírus Oncolíticos , Reação em Cadeia da Polimerase , Ratos , Baço/imunologia , Baço/virologia , Equilíbrio Th1-Th2
16.
J Biomed Biotechnol ; 2010: 350748, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20224643

RESUMO

PURPOSE: To elucidate the influence of ionizing radiation (IR) on the oncolytic activity of Parvovirus H-1 (H-1PV) in human high-grade glioma cells. METHODS: Short term cultures of human high-grade gliomas were irradiated at different doses and infected with H-1PV. Cell viability was assessed by determining relative numbers of surviving cells. Replication of H-1PV was measured by RT-PCR of viral RNA, fluorescence-activated cell sorter (FACS) analysis and the synthesis of infectious virus particles. To identify a possible mechanism for radiation induced change in the oncolytic activity of H-1PV we performed cell cycle analyses. RESULTS: Previous irradiation rendered glioma cells fully permissive to H-1PV infection. Irradiation 24 hours prior to H-1PV infection led to increased cell killing most notably in radioresistant glioma cells. Intracellular levels of NS-1, the main effector of H-1PV induced cytotoxicity, were elevated after irradiation. S-phase levels were increased one day after irradiation improving S-phase dependent viral replication and cytotoxicity. CONCLUSION: This study demonstrates intact susceptibility of previously irradiated glioma-cells for H-1PV induced oncolysis. The combination of ionizing radiation followed by H-1PV infection increased viral cytotoxicity, especially in radioresistant gliomas. These findings support the ongoing development of a clinical trial of H-1PV in patients with recurrent glioblastomas.


Assuntos
Sobrevivência Celular/efeitos da radiação , Glioma/fisiopatologia , Glioma/virologia , Parvovirus H-1/fisiologia , Parvovirus H-1/efeitos da radiação , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Ciclo Celular/efeitos da radiação , Linhagem Celular , Terapia Combinada , Humanos , Vírus Oncolíticos/efeitos da radiação
17.
Int J Cancer ; 127(5): 1230-9, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20087864

RESUMO

Despite multimodal therapeutic concepts, advanced localized and high-risk neuroblastoma remains a therapeutic challenge with a long-term survival rate below 50%. Consequently, new modalities for the treatment of neuroblastoma, e.g., oncolytic virotherapy are urgently required. H-1PV is a rodent parvovirus devoid of relevant pathogenic effects in infected adult animals. In contrast, the virus has oncolytic properties and is particularly cytotoxic for transformed or tumor-derived cells of various species including cells of human origin. Here, a preclinical in vitro assessment of the application of oncolytic H-1PV for the treatment of neuroblastoma cells was performed. Infection efficiency, viral replication and lytic activity of H-1PV were analyzed in 11 neuroblastoma cell lines with different MYCN status. Oncoselectivity of the virus was confirmed by the infection of short term cultures of nonmalignant infant cells of different origin. In these nontransformed cells, no effect of H-1PV on viability or morphology of the cells was observed. In contrast, a lytic infection was induced in all neuroblastoma cell lines examined at MOIs between 0.001 and 10 pfu/cell. H-1PV actively replicated with virus titres increasing up to 5,000-fold within 48-96 hr after infection. The lytic effect of H-1PV was observed independent of MYCN oncogene amplification or differentiation status. Moreover, a significant G2-arrest and induction of apoptosis could be demonstrated. Infection efficiency, rapid virus replication and exhaustive lytic effects on neuroblastoma cells together with the low toxicity of H-1PV for nontransformed cells, render this parvovirus a promising candidate for oncolytic virotherapy of neuroblastoma.


Assuntos
Apoptose , Parvovirus H-1/fisiologia , Neuroblastoma/patologia , Neuroblastoma/virologia , Terapia Viral Oncolítica , Infecções por Parvoviridae/virologia , Adulto , Astrócitos/citologia , Astrócitos/metabolismo , Western Blotting , Células Cultivadas , DNA Viral/genética , Citometria de Fluxo , Fase G1 , Fase G2 , Humanos , Lactente , Neuroblastoma/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/patologia , Replicação Viral
18.
Bioeng Bugs ; 1(6): 429-33, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21468212

RESUMO

The prognosis of malignant brain tumors remains extremely bad in spite of moderate improvements of conventional treatments. A promising alternative approach is the use of oncolytic viruses. Strategies to improve viral toxicity include the combination of oncolytic viruses with standard therapies. Parvovirus H-1 (H-1PV) is an oncolytic virus with proven toxicity in glioma cells. Recently it has been demonstrated that the combination of ionizing radiation (IR) with H-1PV showed promising results. Previously irradiated glioma cells remained fully permissive for H-1PV induced cytotoxicity supporting the use of H-1PV for recurrent gliomas, which typically arise from irradiated cell clones. When glioma cells were infected with H-1PV shortly (24 h) after IR, cell killing improved and only the combination of both treatments lead to complete long-term tumor cell killing. The latter finding raises the question whether IR in combination with H-1PV exerts an additional therapeutic effect on highly resistant glioma stem cells. A likely translation into current clinical treatment protocols is to use stereotactic radiation of non-resectable recurrent gliomas followed by intratumoral injection of H-1PV to harvest the synergistic effects of combination treatment.


Assuntos
Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/virologia , Glioma/radioterapia , Glioma/virologia , Parvovirus H-1/fisiologia , Terapia Viral Oncolítica/métodos , Neoplasias Encefálicas/patologia , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Glioma/patologia , Humanos , Vírus Oncolíticos/fisiologia , Células Tumorais Cultivadas/efeitos da radiação , Células Tumorais Cultivadas/virologia
19.
Int J Cancer ; 126(12): 2914-27, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19856310

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) represents the eighth frequent solid tumor and fourth leading cause of cancer death. Because current treatments against PDAC are still unsatisfactory, new anticancer strategies are required, including oncolytic viruses. Among these, autonomous parvoviruses (PV), like MVMp (minute virus of mice) and H-1PV are being explored as candidates for cancer gene therapy. Human PDAC cell lines were identified to display various susceptibilities to an infection with H-1PV. The correlation between the integrity of the transcription factor SMAD4, mutated in 50% of all PDAC, and H-1PV permissiveness was particularly striking. Indeed, mutation or deletion of SMAD4 dramatically reduced the activity of the P4 promoter and, consequently, the accumulation of the pivotal NS1 protein. By means of DNA affinity immunoblotting, novel binding sites for SMAD4 and c-JUN transcription factors could be identified in the P4 promoter of H-1PV. The overexpression of wild-type SMAD4 in deficient cell lines (AsPC-1, Capan-1) stimulated the activity of the P4 promoter, whereas interference of endogenous SMAD4 function with a dominant-negative mutant decreased the viral promoter activity in wild-type SMAD4-expressing cells (Panc-1, MiaPaCa-2) reducing progeny virus production. In conclusion, the importance of members of the SMAD family for H-1PV early promoter P4 activity should guide us to select SMAD4-positive PDACs, which may be possible targets for an H-1PV-based cancer therapy.


Assuntos
Adenocarcinoma/virologia , Carcinoma Ductal Pancreático/virologia , Parvovirus H-1/fisiologia , Neoplasias Pancreáticas/virologia , Infecções por Parvoviridae/virologia , Proteína Smad4/genética , Proteína Smad4/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/secundário , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundário , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Humanos , Luciferases/metabolismo , Mutação/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Prognóstico , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad4/antagonistas & inibidores , Transfecção , Células Tumorais Cultivadas , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
20.
Clin Immunol ; 132(1): 103-15, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19328037

RESUMO

We have recently demonstrated that upregulation of the innate immune system plays a key role in KRV-induced autoimmune diabetes in the BBDR rat, but the nature of this proinflammatory reaction has not yet been addressed. Using a DNA microarray approach, we identified 569 genes upregulated in pancreatic lymph nodes following virus infection. Among the most highly activated are IL-1 pathways, IFN-gamma-induced chemokines, and genes associated with interferon production and signaling. Ex vivo and in vitro studies indicate that KRV upregulates proinflammatory cytokines and chemokines in B lymphocytes and Flt-3L-induced plasmacytoid DCs (pDCs). Finally, in contrast to KRV, infection of BBDR rats with the non-diabetogenic KRV homologue H-1 parvovirus fails to induce a robust proinflammatory response in pancreatic lymph nodes. Our findings provide new insights into KRV-induced innate immune pathways that may play a role in early mechanisms leading to islet inflammation and diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Perfilação da Expressão Gênica , Imunidade Inata/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Análise de Variância , Animais , Análise por Conglomerados , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/virologia , Feminino , Parvovirus H-1/fisiologia , Interações Hospedeiro-Patógeno , Linfonodos/metabolismo , Masculino , Pâncreas/imunologia , Pâncreas/metabolismo , Parvovirus/fisiologia , Ratos , Ratos Endogâmicos BB , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...