Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Cancer ; 14: 30, 2014 Jan 18.
Article in English | MEDLINE | ID: mdl-24438073

ABSTRACT

BACKGROUND: Adoptive transfer of T cells genetically engineered with a chimeric antigen receptor (CAR) has successfully been used to treat both chronic and acute lymphocytic leukemia as well as other hematological cancers. Experimental therapy with CAR-engineered T cells has also shown promising results on solid tumors. The prostate stem cell antigen (PSCA) is a protein expressed on the surface of prostate epithelial cells as well as in primary and metastatic prostate cancer cells and therefore a promising target for immunotherapy of prostate cancer. METHODS: We developed a third-generation CAR against PSCA including the CD28, OX-40 and CD3 ζ signaling domains. T cells were transduced with a lentivirus encoding the PSCA-CAR and evaluated for cytokine production (paired Student's t-test), proliferation (paired Student's t-test), CD107a expression (paired Student's t-test) and target cell killing in vitro and tumor growth and survival in vivo (Log-rank test comparing Kaplan-Meier survival curves). RESULTS: PSCA-CAR T cells exhibit specific interferon (IFN)-γ and interleukin (IL)-2 secretion and specific proliferation in response to PSCA-expressing target cells. Furthermore, the PSCA-CAR-engineered T cells efficiently kill PSCA-expressing tumor cells in vitro and systemic treatment with PSCA-CAR-engineered T cells significantly delays subcutaneous tumor growth and prolongs survival of mice. CONCLUSIONS: Our data confirms that PSCA-CAR T cells may be developed for treatment of prostate cancer.


Subject(s)
Antigens, Neoplasm/immunology , Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Neoplasm Proteins/immunology , Prostatic Neoplasms/therapy , Receptors, Antigen, T-Cell/biosynthesis , T-Lymphocytes/transplantation , Animals , Cell Degranulation , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic , GPI-Linked Proteins/immunology , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Male , Mice , Mice, Nude , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/biosynthesis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transduction, Genetic , Transfection , Tumor Burden
2.
Hum Gene Ther ; 24(8): 766-75, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23889332

ABSTRACT

Secretogranin III (SGC3) belongs to the granin family and is highly expressed in endocrine and neural tissues. The human SCG3 promoter has not yet been characterized. We identified that a 0.5-kb DNA fragment upstream of the SCG3 gene can selectively drive transgene expression in neuroblastoma cell lines. The strength of transgene expression was further increased, with specificity maintained, by addition of the human achaete-scute complex homolog 1 (ASH1) enhancer. We developed an oncolytic serotype 5-based adenovirus, in which the SCG3 promoter and ASH1 enhancer drive E1A gene expression. The virus was further modified with a cell-penetrating peptide (Tat-PTD) in the viral capsid, which we have previously shown results in increased adenovirus transduction efficiency of many neuroblastoma cell lines. The virus, Ad5PTD(ASH1-SCG3-E1A), shows selective and efficient killing of neuroblastoma cell lines in vitro, including cisplatin-, etoposide-, and doxorubicin-insensitive neuroblastoma cells. Furthermore, it delays tumor growth and thereby prolonged survival for nude mice harboring subcutaneous human neuroblastoma xenograft. In conclusion, we report a novel oncolytic adenovirus with potential use for neuroblastoma therapy.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell-Penetrating Peptides/therapeutic use , Enhancer Elements, Genetic , Genetic Therapy , Neuroblastoma/therapy , Oncolytic Viruses/genetics , Promoter Regions, Genetic , Adenovirus E1A Proteins/genetics , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/genetics , Female , Humans , Mice , Mice, Nude
3.
J Virol ; 85(24): 13114-23, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21957304

ABSTRACT

Adenovirus serotype 5 (Ad5) is widely used as an oncolytic agent for cancer therapy. However, its infectivity is highly dependent on the expression level of coxsackievirus-adenovirus receptor (CAR) on the surfaces of tumor cells. Furthermore, infected cells overproduce adenovirus fiber proteins, which are released prior to cell lysis. The released fibers block CAR on noninfected neighboring cells, thereby preventing progeny virus entry. Our aim was to add a CAR-independent infection route to Ad5 to increase the infectivity of tumor cells with low CAR expression and prevent the fiber-masking problem. We constructed Ad5 viruses that encode the protein transduction domain (PTD) of the HIV-1 Tat protein (Tat-PTD) in hypervariable region 5 (HVR5) of the hexon protein. Tat-PTD functions as a cell-penetrating peptide, and Tat-PTD-modified Ad5 showed a dramatic increased transduction of CAR-negative cell lines compared to unmodified vector. Moreover, while tumor cell infectivity was severely reduced for Ad5 in the presence of fiber proteins, it was only marginally reduced for Tat-PTD-modified Ad5. Furthermore, because of the sequence alteration in the hexon HVR, coagulation factor X-mediated virus uptake was significantly reduced. Mice harboring human neuroblastoma and neuroendocrine tumors show suppressed tumor growths and prolonged survival when treated with Tat-PTD-modified oncolytic viruses. Our data suggest that modification of Ad5 with Tat-PTD in HVR5 expands its utility as an oncolytic agent.


Subject(s)
Adenoviruses, Human/growth & development , Capsid Proteins/metabolism , Neuroblastoma/therapy , Neuroendocrine Tumors/therapy , Oncolytic Viruses/growth & development , Virus Internalization , tat Gene Products, Human Immunodeficiency Virus/metabolism , Adenoviruses, Human/genetics , Animals , Capsid Proteins/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Nude , Neuroblastoma/mortality , Neuroblastoma/pathology , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/pathology , Oncolytic Viruses/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Survival Analysis , Transduction, Genetic , Treatment Outcome , tat Gene Products, Human Immunodeficiency Virus/genetics
4.
Genet Vaccines Ther ; 8: 6, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20804557

ABSTRACT

BACKGROUND: Impaired wound healing in diabetes is related to decreased production of growth factors. Hence, gene therapy is considered as promising treatment modality. So far, efforts concentrated on single gene therapy with particular emphasis on vascular endothelial growth factor-A (VEGF-A). However, as multiple proteins are involved in this process it is rational to test new approaches. Therefore, the aim of this study was to investigate whether single AAV vector-mediated simultaneous transfer of VEGF-A and fibroblast growth factor 4 (FGF4) coding sequences will improve the wound healing over the effect of VEGF-A in diabetic (db/db) mice. METHODS: Leptin receptor-deficient db/db mice were randomized to receive intradermal injections of PBS or AAVs carrying ß-galactosidase gene (AAV-LacZ), VEGF-A (AAV-VEGF-A), FGF-4 (AAV-FGF4-IRES-GFP) or both therapeutic genes (AAV-FGF4-IRES-VEGF-A). Wound healing kinetics was analyzed until day 21 when all animals were sacrificed for biochemical and histological examination. RESULTS: Complete wound closure in animals treated with AAV-VEGF-A was achieved earlier (day 19) than in control mice or animals injected with AAV harboring FGF4 (both on day 21). However, the fastest healing was observed in mice injected with bicistronic AAV-FGF4-IRES-VEGF-A vector (day 17). This was paralleled by significantly increased granulation tissue formation, vascularity and dermal matrix deposition. Mechanistically, as shown in vitro, FGF4 stimulated matrix metalloproteinase-9 (MMP-9) and VEGF receptor-1 expression in mouse dermal fibroblasts and when delivered in combination with VEGF-A, enhanced their migration. CONCLUSION: Combined gene transfer of VEGF-A and FGF4 can improve reparative processes in the wounded skin of diabetic mice better than single agent treatment.

5.
PLoS One ; 5(1): e8916, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20111709

ABSTRACT

BACKGROUND: We have previously developed an oncolytic serotype 5 adenovirus (Ad5) with chromogranin-A (CgA) promoter-controlled E1A expression, Ad[CgA-E1A], with the intention to treat neuroendocrine tumors, including carcinoids. Since carcinoids tend to metastasize to the liver it is important to fully repress viral replication in hepatocytes to avoid adenovirus-related liver toxicity. Herein, we explore miRNA-based regulation of E1A expression as a complementary mechanism to promoter-based transcriptional control. METHODOLOGY/PRINCIPAL FINDINGS: Ad[CgA-E1A-miR122], where E1A expression is further controlled by six tandem repeats of the target sequence for the liver-specific miR122, was constructed and compared to Ad[CgA-E1A]. We observed E1A suppression and replication arrest of the miR122-detargeted adenovirus in normal hepatocytes, while the two viruses killed carcinoid cells to the same degree. Repeated intravenous injections of Ad[CgA-E1A] induced liver toxicity in mice while Ad[CgA-E1A-miR122] injections did not. Furthermore, a miR122-detargeted adenovirus with the wild-type E1A promoter showed reduced replication in hepatic cells compared to wild-type Ad5 but not to the same extent as the miR122-detargeted adenovirus with the neuroendocrine-selective CgA promoter. CONCLUSIONS/SIGNIFICANCE: A combination of transcriptional (promoter) and post-transcriptional (miRNA target) regulation to control virus replication may allow for the use of higher doses of adenovirus for efficient tumors treatment without liver toxicity.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor , Models, Theoretical , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Neoplasms/pathology , Stochastic Processes
6.
Mod Pathol ; 22(2): 261-72, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18953328

ABSTRACT

The gene expression profile of metastasizing serotonin-producing neuroendocrine carcinomas, which arise from enterochromaffin cells in the jejunum and ileum, is still largely unknown. The aim of this study was to identify genes and proteins, which are preferentially expressed by neuroendocrine carcinoma and enterochromaffin cells and therefore potential novel biomarkers and/or therapeutic targets. Six carcinoma specimens and six normal ileal mucosas were profiled by Affymetrix microarrays. Advanced bioinformatics identified differentially and specifically expressed genes, which were validated by quantitative real-time-PCR on tumor cells extracted by laser capture microdissection and normal enterochromaffin cells extracted by immunolaser capture microdissection. We identified six novel marker genes for neuroendocrine carcinoma cells: paraneoplastic antigen Ma2 (PNMA2), testican-1 precursor (SPOCK1), serpin A10 (SERPINA10), glutamate receptor ionotropic AMPA 2 (GRIA2), G protein-coupled receptor 112 (GPR112) and olfactory receptor family 51 subfamily E member 1 (OR51E1). GRIA2 is specifically expressed by neuroendocrine carcinoma cells whereas the others are also expressed by normal enterochromaffin cells. GPR112 and OR51E1 encode proteins associated with the plasma membrane and may therefore become targets for antibody-based diagnosis and therapy. Hierarchical clustering shows high similarity between primary lesions and liver metastases. However, chemokine C-X-C motif ligand 14 (CXCL14) and NK2 transcription factor related locus 3 Drosophila (NKX2-3) are expressed to a lower level in liver metastases than in primary tumors and normal enterochromaffin cells, which implies a role in neuroendocrine carcinoma differentiation. In conclusion, this study provides a list of genes, which possess relatively specific expression to enterochromaffin and neuroendocrine carcinoma cells and genes with differential expression between primary tumors and metastases. We verified six novel marker genes that may be developed as biomarkers and/or therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , Enterochromaffin Cells/chemistry , Gene Expression Regulation, Neoplastic , Ileal Neoplasms/genetics , Ileum/chemistry , RNA, Messenger/analysis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/pathology , Cluster Analysis , Databases, Genetic , Female , Gene Expression Profiling/methods , Humans , Ileal Neoplasms/chemistry , Ileal Neoplasms/pathology , Immunohistochemistry , Intestinal Mucosa/chemistry , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Male , Microdissection , Middle Aged , Neoplasms, Mesothelial/genetics , Neoplasms, Mesothelial/secondary , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Reproducibility of Results
7.
Clin Cancer Res ; 13(8): 2455-62, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17438105

ABSTRACT

PURPOSE: The use of replication-selective oncolytic adenoviruses is an emerging therapeutic approach for cancer, which thus far has not been employed for carcinoids. We therefore constructed Ad[CgA-E1A], a novel replication-selective oncolytic adenovirus, where the chromogranin A (CgA) promoter controls expression of the adenoviral E1A gene. EXPERIMENTAL DESIGN: The Ad[CgA-E1A] virus was evaluated for E1A protein expression, replication ability, and cytolytic activity in various cell lines. It was also evaluated for treatment of xenografted human carcinoid tumors in nude mice. To use Ad[CgA-E1A] for the treatment of carcinoid liver metastases, it is important that normal hepatocytes do not support virus replication to minimize hepatotoxicity. We therefore evaluated CgA protein expression in normal hepatocytes. We also evaluated CgA gene expression in normal hepatocytes and microdissected tumor cells from carcinoid metastases. RESULTS: We found that Ad[CgA-E1A] replicates similarly to wild-type virus in tumor cells with neuroendocrine features, including the BON carcinoid cell line and the SH-SY-5Y neuroblastoma cell lines, whereas it is attenuated in other cell types. Thus, cells where the CgA promoter is active are selectively killed. We also found that Ad[CgA-E1A] is able to suppress fast-growing human BON carcinoid tumors in nude mice. Furthermore, CgA is highly expressed in microdissected cells from carcinoid metastases, whereas it is not expressed in normal hepatocytes. CONCLUSION: Ad[CgA-E1A] is an interesting agent for the treatment of carcinoid liver metastases in conjunction with standard therapy for these malignancies.


Subject(s)
Adenoviridae/genetics , Carcinoid Tumor/therapy , Chromogranin A/genetics , Pancreatic Neoplasms/therapy , Promoter Regions, Genetic , Animals , Carcinoid Tumor/pathology , Cell Line , Cell Line, Tumor , DNA, Viral/genetics , Gene Amplification , Humans , Kidney , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Recombination, Genetic , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...