Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Hum Gene Ther ; 28(10): 800-819, 2017 10.
Article in English | MEDLINE | ID: mdl-28870120

ABSTRACT

Virotherapy is a unique modality for the treatment of cancer with oncolytic viruses (OVs) that selectively infect and lyse tumor cells, spread within tumors, and activate anti-tumor immunity. Various viruses are being developed as OVs preclinically and clinically, several of them engineered to encode therapeutic proteins for tumor-targeted gene therapy. Scientists and clinicians in German academia have made significant contributions to OV research and development, which are highlighted in this review paper. Innovative strategies for "shielding," entry or postentry targeting, and "arming" of OVs have been established, focusing on adenovirus, measles virus, parvovirus, and vaccinia virus platforms. Thereby, new-generation virotherapeutics have been derived. Moreover, immunotherapeutic properties of OVs and combination therapies with pharmacotherapy, radiotherapy, and especially immunotherapy have been investigated and optimized. German investigators are increasingly assessing their OV innovations in investigator-initiated and sponsored clinical trials. As a prototype, parvovirus has been tested as an OV from preclinical proof-of-concept up to first-in-human clinical studies. The approval of the first OV in the Western world, T-VEC (Imlygic), has further spurred the involvement of investigators in Germany in international multicenter studies. With the encouraging developments in funding, commercialization, and regulatory procedures, more German engineering will be translated into OV clinical trials in the near future.


Subject(s)
Genetic Vectors , Oncolytic Virotherapy , Oncolytic Viruses , Research , Animals , Clinical Trials as Topic , Combined Modality Therapy , Drug Evaluation, Preclinical , Genetic Therapy/methods , Genetic Vectors/genetics , Germany , Humans , Models, Animal , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Treatment Outcome
2.
Sci Rep ; 6: 18691, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26725847

ABSTRACT

A 24-amino acid leader peptide of a new human recombinant manganese superoxide dismutase can enter cells and carry molecules. Here, we demonstrated that six of the 24 amino acids penetrate cells through a particular gate represented by a specific amino acid sequence of the oestrogen receptor (ER). We analysed the internalization of the synthetic hexapeptide and the cytotoxic activity of the hexapeptide conjugated to cisplatin on a cell line panel. In most cell lines, the hexapeptide delivered an amount of cisplatin that was 2 to 8 times greater than that released by cisplatin when the drug was used alone. This increased delivery increases the therapeutic index of cisplatin and reduces side effects caused by a high dosage or long-term treatment times. We may consider this hexapeptide a new molecular carrier to deliver molecules with therapeutic activity into ER(+) cells for diagnostic purposes and clinical or immune therapy.


Subject(s)
Drug Carriers/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/chemistry , Cisplatin/metabolism , Cisplatin/pharmacology , Drug Carriers/metabolism , Drug Carriers/pharmacology , Drug Screening Assays, Antitumor , Endoplasmic Reticulum/metabolism , Fluorescein/chemistry , Fluorescein/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Inhibitory Concentration 50 , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Sorting Signals , Receptors, Estrogen/metabolism , Recombinant Proteins/chemistry , Superoxide Dismutase/chemistry
3.
Comp Med ; 65(1): 36-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25730755

ABSTRACT

The autonomous parvovirus H1 (H1PV) is transmitted in rodent populations. The natural host is the rat, in which H1PV infection is pathogenic only in fetuses and newborns. H1PV infection of human cancer cells leads to strong oncolytic effects in preclinical models. In preparation for a clinical trial of H1PV injection in patients with malignant brain tumors, H1PV had to be prepared to Good Manufacturing Practice standards, including extensive toxicology testing in rats. Because the trial involves direct intracerebral injection of H1PV into the tumor and around the resection cavity, possible toxicity to CNS tissue had to be investigated. In addition, quantitative blood levels and the tissue distribution of H1PV after single intracerebral or intravenous injection were measured. Direct injection of H1PV into rat brain at 3 dose levels (maximum, 7.96 × 107 pfu) did not cause any macroscopic or histologic pathology. Furthermore, H1PV infection of the brain did not alter central or autonomous nervous system function. H1PV DNA was detected in almost all organs at 6 h, 48 h, and 14 d after intravenous and intracerebral injection, with the highest levels in liver and spleen. H1PV concentrations in most organs were similar after intravenous and intracerebral injection, indicating high permeability of the blood-brain barrier for this small virus. The current results demonstrate wide organ distribution of H1PV after intravenous or intracerebral injection, confirm that H1PV is nonpathogenic in adult rats even after direct injection into the brain, and form the basis for the ongoing ParvOryx01 clinical trial.


Subject(s)
Central Nervous System/virology , H-1 parvovirus/pathogenicity , Oncolytic Virotherapy/methods , Animals , Biological Availability , Central Nervous System/pathology , DNA, Viral/metabolism , Drug Evaluation, Preclinical , Injections, Intravenous , Liver/virology , Oncolytic Virotherapy/standards , Rats , Spleen/virology , Time Factors , Viral Load
4.
Comp Med ; 65(1): 23-35, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25730754

ABSTRACT

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.


Subject(s)
Genome, Viral/genetics , H-1 parvovirus/pathogenicity , Oncolytic Virotherapy/methods , Parvoviridae Infections/immunology , Parvoviridae Infections/pathology , Animals , Blood Chemical Analysis , Blood Coagulation Tests , Dose-Response Relationship, Immunologic , Drug Evaluation, Preclinical , Feces/virology , H-1 parvovirus/genetics , Immunoglobulin G/immunology , Injections, Intravenous , Liver/pathology , Polymerase Chain Reaction , Rats , Spleen/pathology
5.
Virology ; 447(1-2): 221-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24210118

ABSTRACT

Anti-angiogenic therapy has been recognized as a powerful potential strategy for impeding the growth of various tumors. However no major therapeutic effects have been observed to date, mainly because of the emergence of several resistance mechanisms. Among novel strategies to target tumor vasculature, some oncolytic viruses open up new prospects. In this context, we addressed the question whether the rodent parvovirus H-1PV can target endothelial cells. We show that cultures of human normal (HUVEC) and immortalized (KS-IMM) endothelial cells sustain an abortive viral cycle upon infection with H-1PV and are sensitive to H-1PV cytotoxicity. H-1PV significantly inhibits infected KS-IMM tumor growth. This effect may be traced back by the virus ability to both kill proliferating endothelial cells and inhibit VEGF production Recombinant H-1PV vectors can also transduce tumor cells with chemokines endowed with anti-angiogenesis properties, and warrant further validation for the treatment of highly vascularized tumors.


Subject(s)
Chemokines/metabolism , Endothelial Cells/drug effects , Neovascularization, Pathologic/prevention & control , Parvovirus/physiology , Animals , Biological Therapy/methods , Cell Survival/drug effects , Cells, Cultured , Humans , Mice , Neoplasms/therapy , Survival Analysis , Treatment Outcome
6.
PLoS One ; 7(2): e32197, 2012.
Article in English | MEDLINE | ID: mdl-22359669

ABSTRACT

BACKGROUND: H-1 parvovirus (H-1 PV), a rodent autonomous oncolytic parvovirus, has emerged as a novel class of promising anticancer agents, because of its ability to selectively find and destroy malignant cells. However, to probe H-1 PV multimodal antitumor potential one of the major prerequisites is to decipher H-1 PV direct interplay with human immune system, and so prevent any risk of impairment. METHODOLOGY/PRINCIPAL FINDINGS: Non activated peripheral blood mononuclear cells (PBMCs) are not sensitive to H-1 PV cytotoxic effect. However, the virus impairs both activated PBMC proliferation ability and viability. This effect is related to H-1 PV infection as evidenced by Western blotting detection of H-1 PV main protein NS1. However, TCID50 experiments did not allow newly generated virions to be detected. Moreover, flow cytometry has shown that H-1 PV preferentially targets B lymphocytes. Despite seeming harmful at first sight, H-1 PV seems to affect very few NK cells and CD8+ T lymphocytes and, above all, clearly does not affect human neutrophils and one of the major CD4+ T lymphocyte subpopulation. Very interestingly, flow cytometry analysis and ELISA assays proved that it even activates human CD4+ T cells by increasing activation marker expression (CD69 and CD30) and both effective Th1 and Th2 cytokine secretion (IL-2, IFN-γ and IL-4). In addition, H-1 PV action does not come with any sign of immunosuppressive side effect. Finally, we have shown the efficiency of H-1 PV on xenotransplanted human nasopharyngeal carcinoma, in a SCID mouse model reconstituted with human PBMC. CONCLUSIONS/SIGNIFICANCE: Our results show for the first time that a wild-type oncolytic virus impairs some immune cell subpopulations while directly activating a Helper CD4+ T cell response. Thus, our data open numerous gripping perspectives of investigation and strongly argue for the use of H-1 PV as an anticancer treatment.


Subject(s)
Biological Therapy/methods , CD4-Positive T-Lymphocytes/virology , H-1 parvovirus/immunology , Oncolytic Viruses/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Carcinoma , Humans , Mice , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/therapy , T-Lymphocytes, Regulatory , Xenograft Model Antitumor Assays
7.
Int J Cancer ; 119(4): 932-43, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16550599

ABSTRACT

A cell line derived from a pleiomorphic liposarcoma, named LSA, was previously reported to secrete (a) factor(s) exhibiting oncotoxic properties. The present article describes the isolation, purification and sequence analysis of a protein released by LSA cells into conditioned culture medium. This protein proved to be a variant isoform of manganese superoxide dismutase (MnSOD), hence its designation as LSA-type-MnSOD. This LSA-type-SOD differed from conventional SODs in its secretion by producer cells, contrasting with the normal localization of SODs in the mitochondrial matrix. Interestingly, during the protein purification process, LSA-type-SOD cosegregated with a cytotoxic activity directed against a number of tumor cell lines, as determined under in vitro conditions. This cytopathic effect was most likely due to LSA-type-SOD, since it could be fully reproduced using recombinant SOD that was expressed from cDNA clones isolated from LSA cells mRNA preparations and henceforth designated L-rSOD. In addition to its manifestation in cell lines kept in tissue culture, the oncotoxicity of LSA-type-SOD was further reflected in a remarkable capacity of this protein for suppression of mammary tumors in Balb-C-FR(III) mice. Animals subcutaneously injected with L-rSOD in the tumor area showed a complete disruption of established mammary carcinomas, as monitored by nuclear magnetic resonance (NMR) scanning. Moreover, metastatic spreading, which was readily detected in the control group, was suppressed in the treated animals. Altogether these data suggest that LSA-type-SOD interferes with survival and spreading of neoplastically transformed cells and deserves to be future validated as a therapeutic agent against cancer, either alone or in combination with conventional treatments.


Subject(s)
Liposarcoma/enzymology , Liposarcoma/pathology , Superoxide Dismutase/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Culture Media/isolation & purification , Culture Media/toxicity , DNA, Complementary/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Liposarcoma/genetics , Liposarcoma/metabolism , Mice , Microscopy, Electron, Transmission , Molecular Sequence Data , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL