Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Virol Methods ; 302: 114487, 2022 04.
Article in English | MEDLINE | ID: mdl-35104497

ABSTRACT

The use of oncolytic viruses (OV) to precisely target and eliminate tumors ('virotherapy') is a rapidly evolving therapeutic approach to treating cancer. A major obstacle in virotherapy, especially for systemic administration, is the host's immune response towards the OV. In the case of measles virus (MeV), most individuals have been immunized against this agent leading to pre-existing neutralizing antibodies that can impair OV delivery to the tumor. These antibodies predominantly target the hemagglutinin (H) and fusion (F) envelope glycoproteins displayed at the particle's surface. Here, we introduce a novel and versatile pseudotyping platform for rapid envelope exchange of oncolytic MeV that allows for engineering of chimeric viruses invulnerable to pre-existing anti-MeV antibodies. Using this system, we have successfully exchanged the MeV F and H proteins with the glycoprotein G of vesicular stomatitis virus (VSV) and the surface proteins of Newcastle disease virus (NDV) or canine distemper virus (CDV), all of which are not endemic in the general human population. While the MeV-VSV and MeV-NDV pseudotypes were non-functional, the MeV-CDV pseudotype was successfully propagated to high-titer virus stocks. This study describes the successful generation of a robust envelope exchange platform for oncolytic MeV while also highlighting its intricate pseudotyping tolerance.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Animals , Antibodies, Neutralizing , Measles virus/genetics , Oncolytic Viruses/genetics , Vesicular stomatitis Indiana virus
2.
Eur J Nucl Med Mol Imaging ; 48(12): 3918-3924, 2021 11.
Article in English | MEDLINE | ID: mdl-34018010

ABSTRACT

PURPOSE: A high expression of fibroblast activation protein (FAP) was observed in multiple sarcomas, indicating an enormous potential for PET/CT using 68Ga-radiolabeled inhibitors of FAP (FAPI). Therefore, this retrospective study aimed to evaluate the role of the novel hybrid imaging probe for sarcomas as a first clinical evaluation. METHODS: A cohort of 15 patients underwent 68Ga-FAPI-PET/CT for staging or restaging. The acquisition of PET scans was performed 60 min after administration of 127 to 308 MBq of the tracer. The uptake of 68Ga-FAPI in malignant tissue as well as in healthy organs was quantified by standardized uptake values SUVmean and SUVmax. RESULTS: Excellent tumor-to-background ratios (> 7) could be achieved due to low background activity and high SUVmax in primary tumors (median 7.16), local relapses (median 11.47), and metastases (median 6.29). The highest uptake was found for liposarcomas and high-grade disease (range 18.86-33.61). A high SUVmax (> 10) was observed for clinically more aggressive disease. CONCLUSION: These preliminary findings suggest a high potential for the clinical use of 68Ga-FAPI-PET/CT for patients diagnosed with sarcoma.


Subject(s)
Positron Emission Tomography Computed Tomography , Sarcoma , Humans , Ligands , Neoplasm Recurrence, Local , Retrospective Studies , Sarcoma/diagnostic imaging
3.
Oncoimmunology ; 8(8): 1615817, 2019.
Article in English | MEDLINE | ID: mdl-31413923

ABSTRACT

Pexastimogene devacirepvec (Pexa-Vec) is a vaccinia virus-based oncolytic immunotherapy designed to preferentially replicate in and destroy tumor cells while stimulating anti-tumor immunity by expressing GM-CSF. An earlier randomized Phase IIa trial in predominantly sorafenib-naïve hepatocellular carcinoma (HCC) demonstrated an overall survival (OS) benefit. This randomized, open-label Phase IIb trial investigated whether Pexa-Vec plus Best Supportive Care (BSC) improved OS over BSC alone in HCC patients who failed sorafenib therapy (TRAVERSE). 129 patients were randomly assigned 2:1 to Pexa-Vec plus BSC vs. BSC alone. Pexa-Vec was given as a single intravenous (IV) infusion followed by up to 5 IT injections. The primary endpoint was OS. Secondary endpoints included overall response rate (RR), time to progression (TTP) and safety. A high drop-out rate in the control arm (63%) confounded assessment of response-based endpoints. Median OS (ITT) for Pexa-Vec plus BSC vs. BSC alone was 4.2 and 4.4 months, respectively (HR, 1.19, 95% CI: 0.78-1.80; p = .428). There was no difference between the two treatment arms in RR or TTP. Pexa-Vec was generally well-tolerated. The most frequent Grade 3 included pyrexia (8%) and hypotension (8%). Induction of immune responses to vaccinia antigens and HCC associated antigens were observed. Despite a tolerable safety profile and induction of T cell responses, Pexa-Vec did not improve OS as second-line therapy after sorafenib failure. The true potential of oncolytic viruses may lie in the treatment of patients with earlier disease stages which should be addressed in future studies. ClinicalTrials.gov: NCT01387555.

4.
Cancer Gene Ther ; 21(9): 373-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25145311

ABSTRACT

Precise oncotropism is required for successful systemic administration of next-generation oncolytic measles viruses (MVs). We have previously established a system for efficient post-entry targeting by insertion of synthetic microRNA target sites (miRTS) into the MV genome, thereby repressing replication in the presence of cognate microRNAs. Thus, differential expression of microRNAs, as frequently observed in normal compared with malignant tissues, can be exploited to increase vector specificity and safety. Here we report the combination of miRTS for different microRNAs in a single vector to detarget pivotal organs at risk during systemic administration (liver, brain, gastrointestinal tract). Accordingly, miRTS for miR-122, miR-7 and miR-148a that are enriched in these tissues were inserted to create multi-tissue-detargeted MV (MV-EGFP(mtd)). Replication of MV-EGFP(mtd) is repressed in cell lines as well as in non-transformed primary human hepatocytes and liver slices expressing cognate microRNAs. Oncolytic potency of MV-EGFP(mtd) is retained in a model of pancreatic cancer in vitro and in vivo. This work is a proof-of-concept that favorable expression profiles of multiple microRNAs can be exploited concomitantly to reshape the tropism of MV without compromising oncolytic efficacy. This strategy can be adapted to different vectors and cancer entities for safe and efficient high-dose systemic administration in clinical trials.


Subject(s)
Genetic Vectors/genetics , Measles virus/genetics , MicroRNAs/genetics , Oncolytic Viruses/genetics , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Cell Survival , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Disease Models, Animal , Female , Gene Expression , Gene Knockdown Techniques , Gene Order , Genes, Reporter , Genetic Vectors/administration & dosage , Humans , Mice , MicroRNAs/chemistry , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , RNA Interference , Transduction, Genetic , Vero Cells , Virus Replication/genetics , Xenograft Model Antitumor Assays
5.
Gene Ther ; 20(1): 16-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22218301

ABSTRACT

Lentiviral vectors are vectors of choice for many gene therapy applications. Recently, efficient targeting of lentiviral vectors pseudotyped with the Measles virus (MV) glycoproteins has been reported. However, MV antibodies in patients might limit the clinical use of these vectors. We demonstrate here that lentiviral vectors can also be pseudotyped with the glycoproteins of Tupaia paramyxovirus (TPMV), the hemagglutinin (H) and fusion (F) protein. As this animal paramyxovirus has no known close relatives in humans, we do not expect TPMV antibodies in patients. Because TPMV normally does not infect human cells, 'detargeting' from natural receptors is unnecessary. Similar to the MV system, TPMV glycoproteins can mediate targeted cell entry by displaying different single-chain antibodies (scAb) directed against surface molecules on target cells on the viral hemagglutinin. We generated a panel of H and F proteins with truncated cytoplasmic tails and determined the variants that efficiently pseudotyped lentiviral vectors. The B-cell marker CD20 was used as a model antigen, and CD20-targeted TPMV vectors selectively transduced CD20-positive cells, including quiescent primary human B-cells. Lentiviral vectors pseudotyped with targeted TPMV envelope proteins might be a valuable vector choice when systemic application of targeted lentiviral vectors in humans is required.


Subject(s)
Genetic Vectors/genetics , Lentivirus/genetics , Paramyxoviridae/genetics , Viral Fusion Proteins/genetics , Amino Acid Sequence , Animals , Antigens, CD20/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Transformation, Genetic , Tupaia/virology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology
6.
Cancer Gene Ther ; 19(3): 181-91, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22076043

ABSTRACT

First-line treatment of recurrent and/or refractory head and neck squamous cell carcinoma (HNSCC) is based on platinum, 5-fluorouracil (5-FU) and the monoclonal antiEGFR antibody cetuximab. However, in most cases this chemoimmunotherapy does not cure the disease, and more than 50% of HNSCC patients are dying because of local recurrence of the tumors. In the majority of cases, HNSCC overexpress the epidermal growth factor receptor (EGFR), and its presence is associated with a poor outcome. In this study, we engineered an EGFR-targeted oncolytic measles virus (MV), armed with the bifunctional enzyme cytosine deaminase/uracil phosphoribosyltransferase (CD/UPRT). CD/UPRT converts 5-fluorocytosine (5-FC) into the chemotherapeutic 5-FU, a mainstay of HNSCC chemotherapy. This virus efficiently replicates in and lyses primary HNSCC cells in vitro. Arming with CD/UPRT mediates efficient prodrug activation with high bystander killing of non-infected tumor cells. In mice bearing primary HNSCC xenografts, intratumoral administration of MV-antiEGFR resulted in statistically significant tumor growth delay and prolongation of survival. Importantly, combination with 5-FC is superior to virus-only treatment leading to significant tumor growth inhibition. Thus, chemovirotherapy with EGFR-targeted and CD/UPRT-armed MV is highly efficacious in preclinical settings with direct translational implications for a planned Phase I clinical trial of MV for locoregional treatment of HNSCC.


Subject(s)
Carcinoma, Squamous Cell/therapy , Cytosine Deaminase/genetics , ErbB Receptors/metabolism , Head and Neck Neoplasms/therapy , Measles virus/physiology , Oncolytic Virotherapy/methods , Pentosyltransferases/genetics , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Chlorocebus aethiops , Cytosine Deaminase/biosynthesis , Cytosine Deaminase/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Flucytosine/pharmacokinetics , Flucytosine/pharmacology , Fluorouracil/pharmacokinetics , Fluorouracil/pharmacology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/virology , Humans , Measles virus/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Pentosyltransferases/biosynthesis , Pentosyltransferases/metabolism , Prodrugs/pharmacokinetics , Squamous Cell Carcinoma of Head and Neck , Vero Cells , Xenograft Model Antitumor Assays
7.
Cancer Gene Ther ; 18(8): 598-608, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21701532

ABSTRACT

No curative therapy is currently available for locally advanced or metastatic pancreatic cancer. Therefore, new therapeutic approaches must be considered. Measles virus (MV) vaccine strains have shown promising oncolytic activity against a variety of tumor entities. For specific therapy of pancreatic cancer, we generated a fully retargeted MV that enters cells exclusively through the prostate stem cell antigen (PSCA). Besides a high-membrane frequency on prostate cancer cells, this antigen is expressed on pancreatic adenocarcinoma, but not on non-neoplastic tissue. PSCA expression levels differ within heterogeneous tumor bulks and between human pancreatic cell lines, and we could show specific infection of pancreatic adenocarcinoma cell lines with both high- and low-level PSCA expression. Furthermore, we generated a fully retargeted and armed MV-PNP-anti-PSCA to express the prodrug convertase purine nucleoside phosphorylase (PNP). PNP, which activates the prodrug fludarabine effectively, enhanced the oncolytic efficacy of the virus on infected and bystander cells. Beneficial therapeutic effects were shown in a pancreatic cancer xenograft model. Moreover, in the treatment of gemcitabine-resistant pancreatic adenocarcinoma cells, no cross-resistance to both MV oncolysis and activated prodrug was detected.


Subject(s)
Adenocarcinoma/therapy , Measles virus/physiology , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/virology , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chlorocebus aethiops , Combined Modality Therapy , Female , GPI-Linked Proteins/metabolism , Humans , Male , Measles virus/immunology , Measles virus/metabolism , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/virology , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Vero Cells , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Vidarabine/pharmacokinetics , Xenograft Model Antitumor Assays
8.
Gene Ther ; 17(12): 1506-16, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20686506

ABSTRACT

Measles virus (MV)-PNP H(blind)antiCD20 is a CD20-targeted and prodrug convertase-armed MV that temporarily controls growth of lymphoma xenografts in severe combined immunodeficiency (SCID) mice in combination with fludarabine phosphate (fludarabine). Herein, we examine the replication of this targeted virus and of a vaccine-lineage MV in disease bulks and circulating cells from mantle cell lymphoma (MCL) patients, and show that only the targeted virus is specific for CD20-expressing cells. We then assessed the efficacy of different regimens of administration of this virus in combination with fludarabine and cyclophosphamide (CPA) in an MCL xenograft model. We show that CPA administration before the beginning of virus treatment enhances oncolytic efficacy, likely through temporary immunosuppression. An interval of 1 week between intravenous virus administration and fludarabine treatment further enhanced oncolysis, by synchronizing maximum prodrug convertase expression with fludarabine availability. Finally, three 23-day courses of triple sequential treatment with CPA, virus and fludarabine treatment resulted in complete regression of the xenografts. Secondary disease symptoms interfered with survival, but average survival times increased from 22 to 77 days. These studies document a reprogrammed oncolytic virus, consolidating the effects of two chemotherapeutics, a concept well suited for a phase I clinical trial for MCL patients for whom conventional therapies have failed.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, Mantle-Cell/therapy , Oncolytic Viruses/genetics , Salvage Therapy , Animals , Antigens, CD20/metabolism , Cells, Cultured , Chlorocebus aethiops , Cyclophosphamide/therapeutic use , Humans , Kaplan-Meier Estimate , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/mortality , Lymphoma, Mantle-Cell/pathology , Measles virus/genetics , Mice , Mice, SCID , Molecular Targeted Therapy , Tumor Burden/drug effects , Vero Cells , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Xenograft Model Antitumor Assays
9.
Gene Ther ; 11(7): 599-608, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14724676

ABSTRACT

Current concerns over insertional mutagenesis by retroviral vectors mitigate investigations into alternative, potentially persistent gene therapy vector systems not dependent on genomic integration, such as Sendai virus vectors (SeVV). Prenatal gene therapy requires efficient gene delivery to several tissues, which may not be achievable by somatic gene transfer to the adult. Initially, to test the potential and tropism of the SeVV for gene delivery to fetal tissues, first-generation (replication- and propagation-competent) recombinant SeVV, expressing beta-galactosidase was introduced into late gestation immunocompetent mice via the amniotic and peritoneal cavities and the yolk sac vessels. At 2 days, this resulted in very high levels of expression particularly in the airway epithelium, mesothelium and vascular endothelium, respectively. However, as expected, substantial vector toxicity was observed. The efficiency of gene transfer and the level of gene expression were then examined using a second-generation SeVV. The second generation was developed to be still capable of cytoplasmic RNA replication and therefore high-level gene expression, but incapable of vector spread due to lack of the gene for viral F-protein. Vector was introduced into the fetal amniotic and peritoneal cavities, intravascularly, intramuscularly and intraspinally; at 2 days, expression was observed in the airway epithelia, peritoneal mesothelia, unidentified cells in the gut wall, locally at the site of muscle injection and in the dorsal root ganglia, respectively. Mortality was dramatically diminished compared with the first-generation vector.


Subject(s)
Fetal Diseases/therapy , Fetus/metabolism , Genetic Therapy/methods , Genetic Vectors/toxicity , Lung Diseases/therapy , Viral Fusion Proteins/genetics , Adenoviridae/genetics , Animals , Animals, Newborn , Defective Viruses , Injections, Intravenous , Lung/enzymology , Lung Diseases/embryology , Mice , Mice, Inbred Strains , Models, Animal , Sendai virus/genetics , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL