Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 8(7): 702-11, 2016 07.
Article in English | MEDLINE | ID: mdl-27189167

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a new successful treatment for refractory B-cell leukemia. Successful therapeutic outcome depends on long-term expression of CAR transgene in T cells, which is achieved by delivering transgene using integrating gamma retrovirus (RV) or lentivirus (LV). However, uncontrolled RV/LV integration in host cell genomes has the potential risk of causing insertional mutagenesis. Herein, we describe a novel episomal long-term cell engineering method using non-integrating lentiviral (NILV) vector containing a scaffold/matrix attachment region (S/MAR) element, for either expression of transgenes or silencing of target genes. The insertional events of this vector into the genome of host cells are below detection level. CD19 CAR T cells engineered with a NILV-S/MAR vector have similar levels of CAR expression as T cells engineered with an integrating LV vector, even after numerous rounds of cell division. NILV-S/MAR-engineered CD19 CAR T cells exhibited similar cytotoxic capacity upon CD19(+) target cell recognition as LV-engineered T cells and are as effective in controlling tumor growth in vivo We propose that NILV-S/MAR vectors are superior to current options as they enable long-term transgene expression without the risk of insertional mutagenesis and genotoxicity.


Subject(s)
Adoptive Transfer/methods , Cell Engineering/methods , Cell- and Tissue-Based Therapy/methods , Neoplasms/therapy , Plasmids , T-Lymphocytes/immunology , Transgenes , Animals , Antigens, CD19/immunology , Cells, Cultured , Disease Models, Animal , Gene Expression , Genetic Vectors , Genomic Instability , Humans , Lentivirus/genetics , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen/biosynthesis , Receptors, Antigen/genetics , Treatment Outcome
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.
PLoS One ; 8(1): e54952, 2013.
Article in English | MEDLINE | ID: mdl-23372800

ABSTRACT

Recombinant adenovirus serotype 5 (Ad5) vectors represent one of the most efficient gene delivery vectors in life sciences. However, Ad5 is dependent on expression of the coxsackievirus-adenovirus-receptor (CAR) on the surface of target cell for efficient transduction, which limits it's utility for certain cell types. Herein we present a new vector, Ad5PTDf35, which is an Ad5 vector having serotype 35 fiber-specificity and Tat-PTD hexon-modification. This vector shows dramatically increased transduction capacity of primary human cell cultures including T cells, monocytes, macrophages, dendritic cells, pancreatic islets and exocrine cells, mesenchymal stem cells and tumor initiating cells. Biodistribution in mice following systemic administration (tail-vein injection) show significantly reduced uptake in the liver and spleen of Ad5PTDf35 compared to unmodified Ad5. Therefore, replication-competent viruses with these modifications may be further developed as oncolytic agents for cancer therapy. User-friendly backbone plasmids containing these modifications were developed for compatibility to the AdEasy-system to facilitate the development of surface-modified adenoviruses for gene delivery to difficult-to-transduce cells in basic, pre-clinical and clinical research.


Subject(s)
Adenoviridae/genetics , Capsid Proteins/genetics , Genetic Vectors/genetics , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/genetics , Adenoviridae/immunology , Animals , Cell Line , Gene Order , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacokinetics , Humans , Mice , Primary Cell Culture , T-Lymphocytes/immunology , Tissue Distribution
4.
Proc Natl Acad Sci U S A ; 109(39): 15877-81, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23019373

ABSTRACT

To produce genetically engineered T cells directed against prostate and breast cancer cells, we have cloned the T-cell receptor recognizing the HLA-A2-restricted T-cell receptor γ-chain alternate reading-frame protein (TARP)(4-13) epitope. TARP is a protein exclusively expressed in normal prostate epithelium and in adenocarcinomas of the prostate and breast. Peripheral blood T cells transduced with a lentiviral vector encoding the TARP-TCR proliferated well when exposed to peptide-specific stimuli. These cells exerted peptide-specific IFN-γ production and cytotoxic activity. Importantly, HLA-A2(+) prostate and breast cancer cells expressing TARP were also killed, demonstrating that the TARP(4-13) epitope is a physiologically relevant target for T-cell therapy of prostate and breast cancer. In conclusion, we present the cloning of a T cell receptor (TCR) directed against a physiologically relevant HLA-A2 epitope of TARP. To our knowledge this report on engineering of T cells with a TCR directed against an antigen specifically expressed by prostate cells is unique.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/therapy , Cell Engineering , HLA-A2 Antigen/immunology , Immunity, Cellular , Nuclear Proteins/immunology , Prostatic Neoplasms/therapy , T-Lymphocytes/immunology , Adoptive Transfer , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Gene Expression Regulation/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Male , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
5.
J Neuroinflammation ; 9: 112, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22647574

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS). In the murine experimental autoimmune encephalomyelitis (EAE) model of MS, T regulatory (Treg) cell therapy has proved to be beneficial, but generation of stable CNS-targeting Tregs needs further development. Here, we propose gene engineering to achieve CNS-targeting Tregs from naïve CD4 cells and demonstrate their efficacy in the EAE model. METHODS: CD4+ T cells were modified utilizing a lentiviral vector system to express a chimeric antigen receptor (CAR) targeting myelin oligodendrocyte glycoprotein (MOG) in trans with the murine FoxP3 gene that drives Treg differentiation. The cells were evaluated in vitro for suppressive capacity and in C57BL/6 mice to treat EAE. Cells were administered by intranasal (i.n.) cell delivery. RESULTS: The engineered Tregs demonstrated suppressive capacity in vitro and could efficiently access various regions in the brain via i.n cell delivery. Clinical score 3 EAE mice were treated and the engineered Tregs suppressed ongoing encephalomyelitis as demonstrated by reduced disease symptoms as well as decreased IL-12 and IFNgamma mRNAs in brain tissue. Immunohistochemical markers for myelination (MBP) and reactive astrogliosis (GFAP) confirmed recovery in mice treated with engineered Tregs compared to controls. Symptom-free mice were rechallenged with a second EAE-inducing inoculum but remained healthy, demonstrating the sustained effect of engineered Tregs. CONCLUSION: CNS-targeting Tregs delivered i.n. localized to the CNS and efficiently suppressed ongoing inflammation leading to diminished disease symptoms.


Subject(s)
Cell Engineering/methods , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Forkhead Transcription Factors/administration & dosage , Genetic Therapy/methods , Receptors, Antigen, T-Cell/administration & dosage , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Administration, Intranasal , Animals , Cell Line , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Forkhead Transcription Factors/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Lentivirus/genetics , Lentivirus/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/transplantation
6.
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
7.
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
8.
Hum Gene Ther ; 21(7): 807-13, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20001452

ABSTRACT

Prostate cancer is at present the most common malignancy in men in the Western world. When localized to the prostate, this disease can be treated by curative therapy such as surgery and radiotherapy. However, a substantial number of patients experience a recurrence, resulting in spreading of tumor cells to other parts of the body. In this advanced stage of the disease only palliative treatment is available. Therefore, there is a clear clinical need for new treatment modalities that can, on the one hand, enhance the cure rate of primary therapy for localized prostate cancer and, on the other hand, improve the treatment of metastasized disease. Gene therapy is now being explored in the clinic as a treatment option for the various stages of prostate cancer. Current clinical experiences are based predominantly on trials with adenoviral vectors. As the first of a trilogy of reviews on the state of the art and future prospects of gene therapy in prostate cancer, this review focuses on the clinical experiences and progress of adenovirus-mediated gene therapy for this disease.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Prostatic Neoplasms/therapy , Genetic Therapy/trends , Humans , Male , Neoplasm Staging , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery , Treatment Outcome
9.
J Infect Dis ; 192(11): 1912-20, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16267762

ABSTRACT

Cytomegalovirus (CMV) disease remains a severe complication in patients who have undergone transplantation. Viremia can be prevented and treated by the adoptive transfer of donor-derived CMV-directed T cells. To ensure long-term protection against CMV disease, it is important to transfer CMV antigen-specific T cells that represent both the CD8+ and the CD4+ subsets. In the present study, we used as stimulators dendritic cells (DCs) that were electroporated with in vitro-transcribed 5'-capped polyadenylated messenger RNA (mRNA) that encoded the CMV pp65 protein (i.e., pp65 mRNA). These DCs could efficiently activate CMV-directed CD8+ T cells, as assayed by tetramer staining, interferon- gamma production, and cytolytic activity. We also used DCs that were pulsed with a recombinant pp65 protein to activate CMV-directed CD4+ T cells. When DCs were comodified with pp65 mRNA and pp65 protein, large numbers of CMV-directed CD8+ and CD4+ T cells were generated simultaneously. The approach outlined in the present study can be adapted for a clinical protocol that circumvents potential virus-related biohazards and is available to all patients independently of their human leukocyte antigen haplotype.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Dendritic Cells/immunology , Phosphoproteins , RNA, Messenger , Viral Matrix Proteins , Dendritic Cells/cytology , Dendritic Cells/metabolism , Electroporation , Humans , Lymphocyte Activation , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Species Specificity , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/metabolism
10.
Mol Ther ; 10(2): 355-64, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15294182

ABSTRACT

TARP (T cell receptor gamma-chain alternate reading frame protein) is a protein that in males is uniquely expressed in prostate epithelial cells and prostate cancer cells. We have previously shown that the transcriptional activity of a chimeric sequence comprising the TARP promoter (TARPp) and the PSA enhancer (PSAe) is strictly controlled by testosterone and highly restricted to cells of prostate origin. Here we report that a chimeric sequence comprising TARPp and the PSMA enhancer (PSMAe) is highly active in testosterone-deprived prostate cancer cells, while a regulatory sequence comprising PSAe, PSMAe, and TARPp (PPT) has high prostate-specific activity both in the presence and in the absence of testosterone. Therefore, the PPT sequence may, in a gene therapy setting, be beneficial to prostate cancer patients that have been treated with androgen withdrawal. A recombinant adenovirus vector with the PPT sequence, shielded from interfering adenoviral sequences by the mouse H19 insulator, yields high and prostate-specific transgene expression both in cell cultures and when prostate cancer, PC-346C, tumors were grown orthotopically in nude mice. Intravenous virus administration reveals both higher activity and higher selectivity for the insulator-shielded PPT sequence than for the immediate-early CMV promoter. Therefore, we believe that an adenovirus with therapeutic gene expression controlled by an insulator-shielded PPT sequence is a promising candidate for gene therapy of prostate cancer.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Neoplasms, Hormone-Dependent/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic , Genes, Reporter/genetics , Genetic Vectors/genetics , Humans , Insulator Elements/genetics , Luciferases/analysis , Luciferases/genetics , Male , Mice , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Testosterone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...