Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hum Gene Ther ; 33(15-16): 789-800, 2022 08.
Article in English | MEDLINE | ID: mdl-35297680

ABSTRACT

Diabetes mellitus, caused by loss or dysfunction of the insulin-producing beta cells of the pancreas, is a promising target for recombinant adeno-associated virus (rAAV)-mediated gene therapy. To target potential therapeutic payloads specifically to beta cells, a cell type-specific expression control element is needed. In this study, we tested a series of rAAV vectors designed to express transgenes specifically in human beta cells using the islet-tropic rAAV-KP1 capsid. A small promoter, consisting of only 84 bp of the insulin core promoter was not beta cell-specific in AAV, but highly active in multiple cell types, including tissues outside the pancreas. A larger 363 bp fragment of the insulin promoter (INS) also lacked beta cell specificity. However, beta cell-specific expression was achieved by combining two regulatory elements, a promoter consisting of two copies of INS (INS × 2) and microRNA (miRNA) recognition elements (MREs). The INS × 2 promoter alone showed some beta cell preference, but not tight specificity. To reduce unspecific transgene expression in alpha cells, negative regulation by miRNAs was applied. MREs that are recognized by miRNAs abundant in alpha cells effectively downregulated the transgene expression in these cells. The INS2 × -MRE expression vector was highly specific to human beta cells and stem cell-derived beta cells.


Subject(s)
Dependovirus , MicroRNAs , Dependovirus/genetics , Dependovirus/metabolism , Genetic Vectors/genetics , Humans , Insulin/metabolism , MicroRNAs/metabolism , Transgenes
2.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31723052

ABSTRACT

While gene transfer using recombinant adeno-associated viral (rAAV) vectors has shown success in some clinical trials, there remain many tissues that are not well transduced. Because of the recent success in reprogramming islet-derived cells into functional ß cells in animal models, we constructed 2 highly complex barcoded replication competent capsid shuffled libraries and selected for high-transducing variants on primary human islets. We describe the generation of a chimeric AAV capsid (AAV-KP1) that facilitates transduction of primary human islet cells and human embryonic stem cell-derived ß cells with up to 10-fold higher efficiency compared with previously studied best-in-class AAV vectors. Remarkably, this chimeric capsid also enabled transduction of both mouse and human hepatocytes at very high levels in a humanized chimeric mouse model, thus providing a versatile vector that has the potential to be used in both preclinical testing and human clinical trials for liver-based diseases and diabetes.


Subject(s)
Capsid/chemistry , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Animals , Cells, Cultured , Diabetes Mellitus , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Library , Gene Transfer Techniques , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice
3.
Mol Ther ; 26(5): 1327-1342, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29550076

ABSTRACT

Direct lineage reprogramming can convert readily available cells in the body into desired cell types for cell replacement therapy. This is usually achieved through forced activation or repression of lineage-defining factors or pathways. In particular, reprogramming toward the pancreatic ß cell fate has been of great interest in the search for new diabetes therapies. It has been suggested that cells from various endodermal lineages can be converted to ß-like cells. However, it is unclear how closely induced cells resemble endogenous pancreatic ß cells and whether different cell types have the same reprogramming potential. Here, we report in vivo reprogramming of pancreatic ductal cells through intra-ductal delivery of an adenoviral vector expressing the transcription factors Pdx1, Neurog3, and Mafa. Induced ß-like cells are mono-hormonal, express genes essential for ß cell function, and correct hyperglycemia in both chemically and genetically induced diabetes models. Compared with intrahepatic ducts and hepatocytes treated with the same vector, pancreatic ducts demonstrated more rapid activation of ß cell transcripts and repression of donor cell markers. This approach could be readily adapted to humans through a commonly performed procedure, endoscopic retrograde cholangiopancreatography (ERCP), and provides potential for cell replacement therapy in type 1 diabetes patients.


Subject(s)
Cellular Reprogramming , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreatic Ducts/cytology , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenoviridae/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Cellular Reprogramming/genetics , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/genetics , Hepatocytes/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
4.
PLoS One ; 12(8): e0181812, 2017.
Article in English | MEDLINE | ID: mdl-28813430

ABSTRACT

The gallbladder and cystic duct (GBCs) are parts of the extrahepatic biliary tree and share a common developmental origin with the ventral pancreas. Here, we report on the very first genetic reprogramming of patient-derived human GBCs to ß-like cells for potential autologous cell replacement therapy for type 1 diabetes. We developed a robust method for large-scale expansion of human GBCs ex vivo. GBCs were reprogrammed into insulin-producing pancreatic ß-like cells by a combined adenoviral-mediated expression of hallmark pancreatic endocrine transcription factors PDX1, MAFA, NEUROG3, and PAX6 and differentiation culture in vitro. The reprogrammed GBCs (rGBCs) strongly induced the production of insulin and pancreatic endocrine genes and these responded to glucose stimulation in vitro. rGBCs also expressed an islet-specific surface marker, which was used to enrich for the most highly reprogrammed cells. More importantly, global mRNA and microRNA expression profiles and protein immunostaining indicated that rGBCs adopted an overall ß-like state and these rGBCs engrafted in immunodeficient mice. Furthermore, comparative global expression analyses identified putative regulators of human biliary to ß cell fate conversion. In summary, we have developed, for the first time, a reliable and robust genetic reprogramming and culture expansion of primary human GBCs-derived from multiple unrelated donors-into pancreatic ß-like cells ex vivo, thus showing that human gallbladder is a potentially rich source of reprogrammable cells for autologous cell therapy in diabetes.


Subject(s)
Cellular Reprogramming , Gallbladder/cytology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Animals , Cell Transdifferentiation , Cell Transplantation , Cells, Cultured , Cellular Reprogramming Techniques , Cluster Analysis , Gene Expression , Gene Expression Profiling , Genetic Vectors/genetics , High-Throughput Nucleotide Sequencing , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , MicroRNAs/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic , Transgenes
5.
Curr Opin Endocrinol Diabetes Obes ; 24(2): 92-97, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28099206

ABSTRACT

PURPOSE OF REVIEW: This report examines recent publications identifying phenotypic and functional heterogeneity among pancreatic ß cells and investigating their potential roles in normal and abnormal islet function. The development of new methods and tools for the study of individual islet cells has produced a surge of interest in this topic. RECENT FINDINGS: Studies of ß cell maturation and pregnancy-induced proliferation have identified changes in serotonin and transcription factors SIX2/3 expression as markers of temporal heterogeneity. Structural and functional heterogeneity in the form of functionally distinct 'hub' and 'follower' ß cells was found in mouse islets. Heterogeneous expression of Fltp (in mouse ß cells) and ST8SIA1 and CD9 (in human ß cells) were associated with distinct functional potential. Several impressive reports describing the transcriptomes of individual ß cells were also published in recent months. Some of these reveal previously unknown ß cell subpopulations. SUMMARY: A wealth of information on functional and phenotypic heterogeneity has been collected recently, including the transcriptomes of individual ß cells and the identities of functionally distinct ß cell subpopulations. Several studies suggest the existence of two broad categories: a more proliferative but less functional and a less proliferative but more functional ß cell type. The identification of functionally distinct subpopulations and their association with type 2 diabetes underlines the potential clinical importance of these investigations.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/physiology , Islets of Langerhans/cytology , Animals , Biomarkers/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology
6.
Stem Cell Res ; 17(3): 587-596, 2016 11.
Article in English | MEDLINE | ID: mdl-27833043

ABSTRACT

Direct reprogramming is a promising approach for the replacement of ß cells in diabetes. Reprogramming of cells originating from the endodermal lineage, such as acinar cells in the pancreas, liver cells and gallbladder cells has been of particular interest because of their developmental proximity to ß cells. Our previous work showed that mouse gallbladder epithelium can be partially reprogrammed in vitro to generate islet-like cells (rGBC1). Here, the reprogramming protocol was substantially improved, yielding cells (rGBC2) closer to functional ß cells than the 1st generation method with higher conversion efficiency and insulin expression. In addition to insulin synthesis and processing, rGBC2 presented many hallmark features of ß cells, including insulin secretion in response to high glucose stimulation. Gene expression analysis indicated that rGBC2 clustered closer with ß cells and had a metabolic gene expression profile resembling neonatal ß cells. When transplanted into immune-deficient animals, rGBC2 were stable for at least 5months and further matured in vivo. Taken together, this approach provides further understanding of endodermal lineage conversion and potential for development of cell replacement therapy for type 1 diabetes patients.


Subject(s)
Cellular Reprogramming/physiology , Gallbladder/cytology , Insulin-Secreting Cells/cytology , Animals , Disease Models, Animal , Female , Gallbladder/metabolism , Gene Expression Profiling , Humans , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred NOD
7.
Stem Cell Res ; 15(1): 172-81, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26079872

ABSTRACT

Novel cell surface-reactive monoclonal antibodies generated against extrahepatic biliary cells were developed for the isolation and characterization of different cell subsets from normal adult human gallbladder. Eleven antigenically distinct gallbladder subpopulations were isolated by fluorescence-activated cell sorting. They were classified into epithelial, mesenchymal, and pancreatobiliary (PDX1(+)SOX9(+)) subsets based on gene expression profiling. These antigenically distinct human gallbladder cell subsets could potentially also reflect different functional properties in regards to bile physiology, cell renewal and plasticity. Three of the novel monoclonal antibodies differentially labeled archival sections of primary carcinoma of human gallbladder relative to normal tissue. The novel monoclonal antibodies described herein enable the identification and characterization of antigenically diverse cell subsets within adult human gallbladder and are putative tumor biomarkers.


Subject(s)
Biomarkers/metabolism , Gallbladder/metabolism , Adenocarcinoma/pathology , Adult , Animals , Antibodies/immunology , Bile Ducts, Extrahepatic/metabolism , Cell Line, Tumor , Cell Separation , Cystic Duct/metabolism , Epithelial Cells/cytology , Female , Flow Cytometry , Fluorescent Antibody Technique , Gallbladder/pathology , Gene Expression Regulation , Humans , Mesoderm/cytology , Mice, Inbred BALB C , Pancreas/metabolism , Staining and Labeling
8.
Stem Cell Res ; 11(1): 503-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23562832

ABSTRACT

Cell replacement is an emerging therapy for type 1 diabetes. Pluripotent stem cells have received a lot of attention as a potential source of transplantable ß-cells, but their ability to form teratomas poses significant risks. Here, we evaluated the potential of primary mouse gall bladder epithelial cells (GBCs) as targets for ex vivo genetic reprogramming to the ß-cell fate. Conditions for robust expansion and genetic transduction of primary GBCs by adenoviral vectors were developed. Using a GFP reporter for insulin, conditions for reprogramming were then optimized. Global expression analysis by RNA-sequencing was used to quantitatively compare reprogrammed GBCs (rGBCs) to true ß-cells, revealing both similarities and differences. Adenoviral-mediated expression of NEUROG3, Pdx1, and MafA in GBCs resulted in robust induction of pancreatic endocrine genes, including Ins1, Ins2, Neurod1, Nkx2-2 and Isl1. Furthermore, expression of GBC-specific genes was repressed, including Sox17 and Hes1. Reprogramming was also enhanced by addition of retinoic acid and inhibition of Notch signaling. Importantly, rGBCs were able to engraft long term in vivo and remained insulin-positive for 15weeks. We conclude that GBCs are a viable source for autologous cell replacement in diabetes, but that complete reprogramming will require further manipulations.


Subject(s)
Gallbladder/cytology , Islets of Langerhans/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins , Pluripotent Stem Cells/cytology , Rats , Signal Transduction , Transcription Factors
9.
Nat Biotechnol ; 30(4): 337-43, 2012 Mar 18.
Article in English | MEDLINE | ID: mdl-22426030

ABSTRACT

Multiple intravenous injections of a cDNA library, derived from human melanoma cell lines and expressed using the highly immunogenic vector vesicular stomatitis virus (VSV), cured mice with established melanoma tumors. Successful tumor eradication was associated with the ability of mouse lymphoid cells to mount a tumor-specific CD4(+) interleukin (IL)-17 recall response in vitro. We used this characteristic IL-17 response to screen the VSV-cDNA library and identified three different VSV-cDNA virus clones that, when used in combination but not alone, achieved the same efficacy against tumors as the complete parental virus library. VSV-expressed cDNA libraries can therefore be used to identify tumor rejection antigens that can cooperate to induce anti-tumor responses. This technology should be applicable to antigen discovery for other cancers, as well as for other diseases in which immune reactivity against more than one target antigen contributes to disease pathology.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/isolation & purification , Interleukin-17/immunology , Melanoma/immunology , Melanoma/therapy , Neoplasms, Experimental/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Gene Library , Genetic Vectors , HSP70 Heat-Shock Proteins/metabolism , Humans , Lymphocytes/immunology , Melanoma/genetics , Melanoma, Experimental , Mice , Neoplasms, Experimental/immunology , Vesiculovirus
10.
Nat Med ; 17(7): 854-9, 2011 Jun 19.
Article in English | MEDLINE | ID: mdl-21685898

ABSTRACT

Effective cancer immunotherapy requires the release of a broad spectrum of tumor antigens in the context of potent immune activation. We show here that a cDNA library of normal tissue, expressed from a highly immunogenic viral platform, cures established tumors of the same histological type from which the cDNA library was derived. Immune escape occurred with suboptimal vaccination, but tumor cells that escaped the immune pressure were readily treated by second-line virus-based immunotherapy. This approach has several major advantages. Use of the cDNA library leads to presentation of a broad repertoire of (undefined) tumor-associated antigens, which reduces emergence of treatment-resistant variants and also permits rational, combined-modality approaches in the clinic. Finally, the viral vectors can be delivered systemically, without the need for tumor targeting, and are amenable to clinical-grade production. Therefore, virus-expressed cDNA libraries represent a novel paradigm for cancer treatment addressing many of the key issues that have undermined the efficacy of immuno- and virotherapy to date.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Gene Library , Neoplasms/therapy , Animals , Cancer Vaccines/immunology , DNA, Complementary/genetics , Genetic Vectors/genetics , Male , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Polymerase Chain Reaction , Prostatic Neoplasms/therapy , Vesiculovirus/genetics
11.
Hum Gene Ther ; 22(11): 1343-53, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21366404

ABSTRACT

We have shown that the antitumor activity of vesicular stomatitis virus (VSV) against B16ova tumors in C57BL/6 mice is predominantly due to innate antiviral immune effectors. We have also shown that the innate immune-activating properties of VSV can be harnessed to prime adaptive T-cell responses against a tumor-associated antigen (TAA) if the virus is engineered to express the cDNA of the antigen. Here, we show that the combination of VSV expressing OVA as a model tumor antigen, along with adoptive T-cell therapy targeted against the same antigen, is superior to either treatment alone and induces systemic antitumor activity. In addition, we extend our findings with the OVA model to the therapeutic use of VSV expressing hgp100, a self TAA against which tolerance is well established in C57BL/6 mice. In contrast to VSV-ova, T-cell responses raised by VSV-hgp100 were insufficient to improve therapy against B16ova tumors compared with VSV-GFP alone. However, in combination with adoptive transfer of gp100-specific pmel T cells, intratumoral VSV-hgp100 cured significantly more mice than either virus or T cells alone. Even in an aggressive model of metastatic disease, antitumor therapy was generated at levels similar to those observed in the VSV-ova/OT-I model in which a potently immunogenic, nonself TAA was targeted. Therefore, individual poorly effective virotherapies and T-cell therapies that target self TAA of low immunogenicity, which reflects the situation in patients, can be combined to generate very effective antitumor therapy.


Subject(s)
Melanoma, Experimental/therapy , T-Lymphocytes/immunology , Vesicular stomatitis Indiana virus/physiology , gp100 Melanoma Antigen/immunology , Adoptive Transfer , Animals , Combined Modality Therapy , Immunity, Innate , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncolytic Virotherapy , T-Lymphocytes/transplantation , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/immunology , Virus Replication , gp100 Melanoma Antigen/genetics
12.
Mol Ther ; 19(1): 140-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20978474

ABSTRACT

Despite having potent oncolytic activity, in vitro, direct intratumoral injection of oncolytic vesicular stomatitis virus (VSV) into established AE17ova mesothelioma tumors in C57Bl/6 mice had no therapeutic effect. During studies to combine systemic cyclophosphamide (CPA) with VSV to suppress the innate immune reaction against VSV, we observed that CPA alone had highly significant antitumor effects in this model. However, against our expectations, the combination of CPA and VSV consistently reduced therapeutic efficacy compared to CPA alone, despite the fact that the combination increased intratumoral VSV titers. We show here that CPA-mediated therapy against AE17ova tumors was immune-mediated and dependent upon both CD4 T cells and natural killer (NK) cells. However, intratumoral VSV induced a transforming growth factor-ß (TGF-ß)-dependent suppressive activity, mediated by CD11b(+)GR-1(+) cells that significantly inhibited both antigen-specific T-cell activation, and CPA-activated, NK-dependent killing of AE17ova tumor cells. Overall, our results show that treatment with oncolytic viruses can induce a variety of immune-mediated consequences in vivo with both positive, or negative, effects on antitumor therapy. These underexplored immune consequences of treatment with oncolytic viruses may have significant, and possibly unexpected, impacts on how virotherapy interacts in combination with other agents which modulate antitumor immune effectors.


Subject(s)
Cyclophosphamide/pharmacology , Genetic Therapy/methods , Mesothelioma/immunology , Mesothelioma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Vesicular stomatitis Indiana virus/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Combined Modality Therapy , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/virology , Mesothelioma/drug therapy , Mesothelioma/virology , Mice , Mice, Inbred C57BL , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Transforming Growth Factor beta/metabolism , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/metabolism , Virus Replication
13.
Mol Ther ; 19(1): 150-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20959810

ABSTRACT

We show here, for the first time to our knowledge, that the antitumor therapy of oncolytic vesicular stomatitis virus (VSV) in the B16ova model depends upon signaling through myeloid differentiation primary response gene 88 (MyD88) in host cells. VSV-mediated therapy of B16ova tumors was abolished in MyD88(-/-) mice despite generation of antigen-specific T cell responses similar to those in immune-competent mice. Mice defective in only toll-like receptor 4 (TLR4), TLR7, or interleukin 1 (IL-1) signaling retained VSV-induced therapy, suggesting that multiple, redundant pathways of innate immune activation by the virus contribute to antitumor immune reactivity. Lack of MyD88 signaling was associated with decreased expression of proinflammatory cytokines and neutrophil infiltration in response to intratumoral virus, as well as decreased infiltration of draining lymph nodes (LN) with plasmacytoid dendritic cells (pDCs) (CD11b(-)GR1(+)B220(+)) and myeloid-derived suppressor cells (CD11b(+)GR1(+)F4/80(+)). MyD88 signaling in response to VSV was also closely associated with a type I interferon (IFN) response. This inhibited virus replication within the tumor but also protected the host from viral dissemination from the tumor. Therefore, the innate immune response to oncolytic viruses can be, simultaneously, protherapeutic, antioncolytic, and systemically protective. These paradoxically conflicting roles need to be carefully considered in future strategies designed to improve the efficacy of oncolytic virotherapy.


Subject(s)
Melanoma, Experimental/therapy , Myeloid Differentiation Factor 88/metabolism , Oncolytic Virotherapy/methods , Vesicular stomatitis Indiana virus/physiology , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Immunity, Innate/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-1/deficiency , Interleukin-1/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/virology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Signal Transduction , T-Lymphocytes/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/metabolism , Virus Replication/genetics
14.
Cancer Res ; 70(11): 4539-49, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20484025

ABSTRACT

Innate immune effector mechanisms triggered by oncolytic viruses may contribute to the clearance of both infected and uninfected tumor cells in immunocompetent murine hosts. Here, we developed an in vitro tumor cell/bone marrow coculture assay and used it to dissect innate immune sensor and effector responses to intratumoral vesicular stomatitis virus (VSV). We found that the type III IFN interleukin-28 (IL-28) was induced by viral activation of innate immune-sensing cells, acting as a key mediator of VSV-mediated virotherapy of B16ova melanomas. Using tumor variants which differentially express the IL-28 receptor, we showed that IL-28 induced by VSV within the tumor microenvironment sensitizes tumor cells to natural killer cell recognition and activation. These results revealed new insights into the immunovirological mechanisms associated with oncolytic virotherapy in immune-competent hosts. Moreover, they defined a new class of tumor-associated mutation, such as acquired loss of responsiveness to IL-28 signaling, which confers insensitivity to oncolytic virotherapy through a mechanism independent of viral replication in vitro. Lastly, the findings suggested new strategies to manipulate immune signals that may enhance viral replication, along with antitumor immune activation, and improve the efficacy of oncolytic virotherapies.


Subject(s)
Cytokines/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Oncolytic Virotherapy/methods , Vesicular stomatitis Indiana virus/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/virology , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , Immunity, Innate/immunology , Immunocompromised Host , Injections, Intralesional , Interferon Type I/immunology , Killer Cells, Natural/immunology , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytokine/biosynthesis , Receptors, Cytokine/immunology , Virus Replication/immunology
15.
Hum Gene Ther ; 21(4): 439-50, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19922169

ABSTRACT

Oncolytic virotherapy can be achieved in two ways: (1) by exploiting an innate ability of certain viruses to selectively replicate in tumor tissues, and (2) by using viruses to deliver toxic or immunostimulatory genes to tumors. Vesicular stomatitis virus (VSV) selectively replicates in tumors lacking adequate type I interferon response. The efficacy of oncolytic virotherapy using VSV against B16 melanomas in C57BL/6 mice is dependent on CD8(+) T and natural killer cells. Because immunotherapies that prime specific CD8(+) T cells against melanocyte/melanoma antigens can generate significant therapeutic responses, we hypothesized that engineering VSV to express the potent T cell costimulatory molecule CD40 ligand (VSV-CD40L) would enhance virotherapy with concomitant priming of melanoma-specific T cells. However, we observed no difference in antitumor efficacy between the parental VSV-GFP and VSV-CD40L. In contrast, intratumoral injection of a replication-defective adenovirus expressing CD40L (Ad-CD40L) consistently produced significantly greater therapy than either replication-competent VSV-GFP or VSV-CD40L. The Ad-CD40L-mediated tumor regressions were associated with specific T cell responses against tumor-associated antigens (TAAs), which took several days to develop, whereas VSV-CD40L rapidly induced high levels of T cell activation without specificity for TAAs. These data suggest that the high levels of VSV-associated immunogenicity distracted immune responses away from priming of tumor-specific T cells, even in the presence of potent costimulatory signals. In contrast, a replication-defective Ad-CD40L allowed significant priming of T cells directed against TAAs. These observations suggest that an efficiently primed antitumor T cell response can produce similar, if not better, therapy against an established melanoma compared with intratumoral injection of a replication-competent oncolytic virus.


Subject(s)
CD40 Ligand , Genetic Therapy/methods , Immunotherapy/methods , Melanoma, Experimental/therapy , Oncolytic Viruses , Vesicular stomatitis Indiana virus , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae/metabolism , Adenoviridae/physiology , Animals , CD40 Ligand/genetics , CD40 Ligand/immunology , CD40 Ligand/metabolism , CD8-Positive T-Lymphocytes/immunology , Cricetinae , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Oncolytic Viruses/metabolism , Oncolytic Viruses/physiology , Transgenes , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/metabolism , Vesicular stomatitis Indiana virus/physiology
16.
Clin Cancer Res ; 15(13): 4374-4381, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19509134

ABSTRACT

PURPOSE: Reovirus is a naturally occurring oncolytic virus in clinical trials. Although tumor infection by reovirus can generate adaptive antitumor immunity, its therapeutic importance versus direct viral oncolysis is undefined. This study addresses the requirement for viral oncolysis and replication, and the relative importance of antitumor immunity and direct oncolysis in therapy. EXPERIMENTAL DESIGN: Nonantigen specific T cells loaded with reovirus were delivered i.v. to C57BL/6 and severe combined immunodeficient mice bearing lymph node and splenic metastases from the murine melanoma, B16ova, with assessment of viral replication, metastatic clearance by tumor colony outgrowth, and immune priming. Human cytotoxic lymphocyte priming assays were done with dendritic cells loaded with Mel888 cells before the addition of reovirus. RESULTS: B16ova was resistant to direct oncolysis in vitro, and failed to support reovirus replication in vitro or in vivo. Nevertheless, reovirus purged lymph node and splenic metastases in C57BL/6 mice and generated antitumor immunity. In contrast, reovirus failed to reduce tumor burden in severe combined immunodeficient mice bearing either B16ova or reovirus-sensitive B16tk metastases. In the human system, reovirus acted solely as an adjuvant when added to dendritic cells already loaded with Mel888, supporting priming of specific antitumor cytotoxic lymphocyte, in the absence of significant direct tumor oncolysis; UV-treated nonreplicating reovirus was similarly immunogenic. CONCLUSION: The immune response is critical in mediating the efficacy of reovirus, and does not depend upon direct viral oncolysis or replication. The findings are of direct relevance to fulfilling the potential of this novel anticancer agent.


Subject(s)
Cytotoxicity, Immunologic/physiology , Mammalian orthoreovirus 3/physiology , Melanoma, Experimental/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Virus Replication/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Immunotherapy, Adoptive/methods , Mammalian orthoreovirus 3/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Oncolytic Viruses/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , T-Lymphocytes, Cytotoxic/virology , Treatment Outcome , Virus Replication/immunology
17.
Mol Ther ; 16(12): 1910-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18827807

ABSTRACT

To protect viral particles from neutralization, sequestration, nonspecific adhesion, and mislocalization following systemic delivery, we have previously exploited the natural tumor-homing properties of antigen-specific CD8+ T cells. Thus, OT-I T cells, preloaded in vitro with the oncolytic vesicular stomatitis virus (VSV), can deliver virus to established B16ova tumors to generate significantly better therapy than that achievable with OT-I T cells, or systemically delivered VSV, alone. Here, we demonstrate that preconditioning immune-competent mice with Treg depletion and interleukin-2 (IL-2), before adoptive T-cell therapy with OT-I T cells loaded with VSV, leads to further highly significant increases in antitumor therapy. Therapy was associated with antitumor immune memory, but with no detectable toxicities associated with IL-2, Treg depletion, or systemic dissemination of the oncolytic virus. Efficacy was contributed by multiple factors, including improved persistence of T cells; enhanced delivery of VSV to tumors; increased persistence of OT-I cells in vivo resulting from tumor oncolysis; and activation of NK cells, which acquire potent antitumor and proviral activities. By controlling the levels of virus loaded onto the OT-I cells, adoptive therapy was still effective in mice preimmune to the virus, indicating that therapy with virus-loaded T cells may be useful even in virus-immune patients. Taken together, our data show that it is possible to combine adoptive T-cell therapy, with biological therapy (Treg depletion+IL-2), and VSV virotherapy, to treat established tumors under conditions where none of the individual modalities alone is successful.


Subject(s)
Adaptation, Biological/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy , T-Lymphocytes/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigens/immunology , Cell Line, Tumor , Immunologic Memory/immunology , Interleukin-2/immunology , Killer Cells, Natural/immunology , Mice , Neoplasms/genetics , T-Lymphocytes/drug effects , Vesiculovirus/genetics
18.
Mol Ther ; 16(7): 1217-26, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18431359

ABSTRACT

There are several roadblocks that hinder systemic delivery of oncolytic viruses to the sites of metastatic disease. These include the tumor vasculature, which provides a physical barrier to tumor-specific virus extravasation. Although interleukin-2 (IL-2) has been used in antitumor therapy, it is associated with endothelial cell injury, leading to vascular leak syndrome (VLS). Here, we demonstrate that IL-2-mediated VLS, accentuated by depletion of regulatory T cells (Treg), facilitates localization of intravenously (i.v.) delivered oncolytic virus into established tumors in immune-competent mice. IL-2, in association with Treg depletion, generates "hyperactivated" natural killer (NK) cells, possessing antitumor activity and secreting factors that facilitate virus spread/replication throughout the tumor by disrupting the tumor architecture. As a result, the combination of Treg depletion/IL-2 and systemic oncolytic virotherapy was found to be significantly more therapeutic against established disease than either treatment alone. These data demonstrate that it is possible to combine biological therapy with oncolytic virotherapy to generate systemic therapy against established tumors.


Subject(s)
Interleukin-2/therapeutic use , Lymphocyte Depletion , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Combined Modality Therapy , Endothelium, Vascular/drug effects , Interleukin-2/toxicity , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Oncolytic Viruses/drug effects , Oncolytic Viruses/physiology , T-Lymphocytes, Regulatory/immunology , Virus Internalization , Virus Replication/drug effects
19.
Mol Ther ; 16(7): 1217-1226, 2008 Jul.
Article in English | MEDLINE | ID: mdl-28178481

ABSTRACT

There are several roadblocks that hinder systemic delivery of oncolytic viruses to the sites of metastatic disease. These include the tumor vasculature, which provides a physical barrier to tumor-specific virus extravasation. Although interleukin-2 (IL-2) has been used in antitumor therapy, it is associated with endothelial cell injury, leading to vascular leak syndrome (VLS). Here, we demonstrate that IL-2-mediated VLS, accentuated by depletion of regulatory T cells (Treg), facilitates localization of intravenously (IV) delivered oncolytic virus into established tumors in immune-competent mice. IL-2, in association with Treg depletion, generates "hyperactivated" natural killer (NK) cells, possessing antitumor activity and secreting factors that facilitate virus spread/replication throughout the tumor by disrupting the tumor architecture. As a result, the combination of Treg depletion/IL-2 and systemic oncolytic virotherapy was found to be significantly more therapeutic against established disease than either treatment alone. These data demonstrate that it is possible to combine biological therapy with oncolytic virotherapy to generate systemic therapy against established tumors.

20.
Nat Med ; 14(1): 37-44, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18066076

ABSTRACT

In many common cancers, dissemination of secondary tumors via the lymph nodes poses the most significant threat to the affected individual. Metastatic cells often reach the lymph nodes by mimicking the molecular mechanisms used by hematopoietic cells to traffic to peripheral lymphoid organs. Therefore, we exploited naive T cell trafficking in order to chaperone an oncolytic virus to lymphoid organs harboring metastatic cells. Metastatic burden was initially reduced by viral oncolysis and was then eradicated, as tumor cell killing in the lymph node and spleen generated protective antitumor immunity. Lymph node purging of tumor cells was possible even in virus-immune mice. Adoptive transfer of normal T cells loaded with oncolytic virus into individuals with cancer would be technically easy to implement both to reduce the distribution of metastases and to vaccinate the affected individual in situ against micrometastatic disease. As such, this adoptive transfer could have a great therapeutic impact, in the adjuvant setting, on many different cancer types.


Subject(s)
Immunotherapy/methods , Lymph Nodes/pathology , Lymphatic Metastasis , Oncolytic Viruses/metabolism , T-Lymphocytes/immunology , Animals , Antigens/chemistry , Colorectal Neoplasms/pathology , Immune System , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Neoplasms/pathology , Spleen/metabolism , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...