Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Viruses ; 10(6)2018 06 03.
Article in English | MEDLINE | ID: mdl-29865280

ABSTRACT

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.


Subject(s)
Apoptosis , H-1 parvovirus/physiology , Oncolytic Virotherapy , Sarcoma, Ewing/therapy , Sarcoma, Ewing/virology , Animals , Cell Cycle , Cell Line, Tumor , Cell Survival , Female , Humans , Mice, Nude , Oncolytic Viruses/physiology , Parvovirus , Virus Replication , Xenograft Model Antitumor Assays
2.
Viruses ; 8(5)2016 05 19.
Article in English | MEDLINE | ID: mdl-27213425

ABSTRACT

Combining virus-induced cytotoxic and immunotherapeutic effects, oncolytic virotherapy represents a promising therapeutic approach for high-grade glioma (HGG). A clinical trial has recently provided evidence for the clinical safety of the oncolytic parvovirus H-1 (H-1PV) in adult glioblastoma relapse patients. The present study assesses the efficacy of H-1PV in eliminating HGG initiating cells. H-1PV was able to enter and to transduce all HGG neurosphere culture models (n = 6), including cultures derived from adult glioblastoma, pediatric glioblastoma, and diffuse intrinsic pontine glioma. Cytotoxic effects induced by the virus have been observed in all HGG neurospheres at half maximal inhibitory concentration (IC50) doses of input virus between 1 and 10 plaque forming units per cell. H-1PV infection at this dose range was able to prevent tumorigenicity of NCH421k glioblastoma multiforme (GBM) "stem-like" cells in NOD/SCID mice. Interestingly NCH421R, an isogenic subclone with equal capacity of xenograft formation, but resistant to H-1PV infection could be isolated from the parental NCH421k culture. To reveal changes in gene expression associated with H-1PV resistance we performed a comparative gene expression analysis in these subclones. Several dysregulated genes encoding receptor proteins, endocytosis factors or regulators innate antiviral responses were identified and represent intriguing candidates for to further study molecular mechanisms of H-1PV resistance.


Subject(s)
H-1 parvovirus/growth & development , Neuroglia/physiology , Neuroglia/virology , Oncolytic Viruses/growth & development , Stem Cells/physiology , Stem Cells/virology , Animals , Disease Models, Animal , Gene Expression Profiling , Glioma/therapy , Heterografts , Humans , Mice, Inbred NOD , Mice, SCID , Models, Theoretical , Oncolytic Virotherapy/methods
3.
G3 (Bethesda) ; 3(8): 1325-34, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23733888

ABSTRACT

In Drosophila melanogaster, two chromosome-specific targeting and regulatory systems have been described. The male-specific lethal (MSL) complex supports dosage compensation by stimulating gene expression from the male X-chromosome, and the protein Painting of fourth (POF) specifically targets and stimulates expression from the heterochromatic 4(th) chromosome. The targeting sites of both systems are well characterized, but the principles underlying the targeting mechanisms have remained elusive. Here we present an original observation, namely that POF specifically targets two loci on the X-chromosome, PoX1 and PoX2 (POF-on-X). PoX1 and PoX2 are located close to the roX1 and roX2 genes, which encode noncoding RNAs important for the correct targeting and spreading of the MSL-complex. We also found that the targeting of POF to PoX1 and PoX2 is largely dependent on roX expression and identified a high-affinity target region that ectopically recruits POF. The results presented support a model linking the MSL-complex to POF and dosage compensation to regulation of heterochromatin.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosomes/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Biological Evolution , Dosage Compensation, Genetic , Drosophila Proteins/metabolism , Female , Gene Expression , Genetic Loci , Heterochromatin/genetics , Heterochromatin/metabolism , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Transcription Factors/metabolism , X Chromosome
4.
PLoS One ; 7(5): e37239, 2012.
Article in English | MEDLINE | ID: mdl-22615950

ABSTRACT

Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for canine cancer therapy. In this study we describe the establishment of an in vivo model of canine soft tissue sarcoma (CSTS) using the new isolated cell line STSA-1 and the analysis of the virus-mediated oncolytic and immunological effects of two different Lister VACV LIVP1.1.1 and GLV-1h68 strains against CSTS. Cell culture data demonstrated that both tested VACV strains efficiently infected and destroyed cells of the canine soft tissue sarcoma line STSA-1. In addition, in our new canine sarcoma tumor xenograft mouse model, systemic administration of LIVP1.1.1 or GLV-1h68 viruses led to significant inhibition of tumor growth compared to control mice. Furthermore, LIVP1.1.1 mediated therapy resulted in almost complete tumor regression and resulted in long-term survival of sarcoma-bearing mice. The replication of the tested VACV strains in tumor tissues led to strong oncolytic effects accompanied by an intense intratumoral infiltration of host immune cells, mainly neutrophils. These findings suggest that the direct viral oncolysis of tumor cells and the virus-dependent activation of tumor-associated host immune cells could be crucial parts of anti-tumor mechanism in STSA-1 xenografts. In summary, the data showed that both tested vaccinia virus strains and especially LIVP1.1.1 have great potential for effective treatment of CSTS.


Subject(s)
Muscle Neoplasms/veterinary , Oncolytic Virotherapy/methods , Sarcoma/veterinary , Soft Tissue Neoplasms/veterinary , Vaccinia virus/physiology , Animals , Bone Neoplasms/secondary , Bone Neoplasms/veterinary , Cell Line, Tumor , Dogs , Mice , Muscle Neoplasms/therapy , Oncolytic Viruses/physiology , Sarcoma/pathology , Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Virus Replication/physiology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...