Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Methods Mol Biol ; 1937: 177-188, 2019.
Article in English | MEDLINE | ID: mdl-30706396

ABSTRACT

Herpes simplex virus (HSV) is one of the most extensively studied oncolytic virus platforms. The recent FDA approval of talimogene laherparepvec (T-VEC) has been accelerating translational research of oncolytic HSV (oHSV) as a promising therapeutic for refractory cancers such as glioblastoma, the deadliest primary malignancy in the brain. The large genome size of HSV readily allows arming of oHSV by incorporating therapeutic transgenes within the virus, as exemplified by T-VEC carrying GM-CSF, thereby enhancing the anticancer activity of oHSV. Here we describe a bacterial artificial chromosome-based method for construction of an oHSV expressing a transgene, which we routinely use in the laboratory to create a number of different recombinant oHSV bearing either therapeutic or reporter genes.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Herpesvirus 1, Human/genetics , Oncolytic Viruses/genetics , Animals , Chlorocebus aethiops , Genetic Vectors/genetics , Humans , Oncolytic Virotherapy , Transfection , Transgenes , Vero Cells
2.
Hepatology ; 59(4): 1577-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24677197

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is the most rapidly increasing cause of cancer-related mortality in the United States. Because of the lack of viable treatment options for HCC, prevention in high-risk patients has been proposed as an alternative strategy. The main risk factor for HCC is cirrhosis and several lines of evidence implicate epidermal growth factor (EGF) in the progression of cirrhosis and development of HCC. We therefore examined the effects of the EGF receptor (EGFR) inhibitor erlotinib on liver fibrogenesis and hepatocellular transformation in three different animal models of progressive cirrhosis: a rat model induced by repeated, low-dose injections of diethylnitrosamine (DEN), a mouse model induced by carbon tetrachloride (CCl4 ), and a rat model induced by bile duct ligation (BDL). Erlotinib reduced EGFR phosphorylation in hepatic stellate cells (HSC) and reduced the total number of activated HSC. Erlotinib also decreased hepatocyte proliferation and liver injury. Consistent with all these findings, pharmacological inhibition of EGFR signaling effectively prevented the progression of cirrhosis and regressed fibrosis in some animals. Moreover, by alleviating the underlying liver disease, erlotinib blocked the development of HCC and its therapeutic efficacy could be monitored with a previously reported gene expression signature predictive of HCC risk in human cirrhosis patients. CONCLUSION: These data suggest that EGFR inhibition using Food and Drug Administration-approved inhibitors provides a promising therapeutic approach for reduction of fibrogenesis and prevention of HCC in high-risk cirrhosis patients who can be identified and monitored by gene expression signatures.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Disease Progression , ErbB Receptors/antagonists & inhibitors , Liver Cirrhosis/prevention & control , Liver Neoplasms/prevention & control , Quinazolines/therapeutic use , Animals , Bile Ducts/physiopathology , Carbon Tetrachloride/adverse effects , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cells, Cultured , Diethylnitrosamine/adverse effects , Disease Models, Animal , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Ligation/adverse effects , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred Strains , Phosphorylation/drug effects , Prognosis , Quinazolines/pharmacology , Rats , Rats, Wistar , Transcriptome
3.
Proc Natl Acad Sci U S A ; 110(29): 12006-11, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23754388

ABSTRACT

Glioblastoma (World Health Organization grade IV) is an aggressive adult brain tumor that is inevitably fatal despite surgery, radiation, and chemotherapy. Treatment failures are attributed to combinations of cellular heterogeneity, including a subpopulation of often-resistant cancer stem cells, aberrant vasculature, and noteworthy immune suppression. Current preclinical models and treatment strategies do not incorporate or address all these features satisfactorily. Herein, we describe a murine glioblastoma stem cell (GSC) model that recapitulates tumor heterogeneity, invasiveness, vascularity, and immunosuppressive microenvironment in syngeneic immunocompetent mice and should prove useful for a range of therapeutic studies. Using this model, we tested a genetically engineered oncolytic herpes simplex virus that is armed with an immunomodulatory cytokine, interleukin 12 (G47-mIL12). G47Δ-mIL12 infects and replicates similarly to its unarmed oncolytic herpes simplex virus counterpart in mouse 005 GSCs in vitro, whereas in vivo, it significantly enhances survival in syngeneic mice bearing intracerebral 005 tumors. Mechanistically, G47-mIL12 targets not only GSCs but also increases IFN-γ release, inhibits angiogenesis, and reduces the number of regulatory T cells in the tumor. The increased efficacy is dependent upon T cells, but not natural killer cells. Taken together, our findings demonstrate that G47Δ-mIL12 provides a multifaceted approach to targeting GSCs, tumor microenvironment, and the immune system, with resultant therapeutic benefit in a stringent glioblastoma model.


Subject(s)
Disease Models, Animal , Glioblastoma/therapy , Immunotherapy/methods , Interleukin-12/metabolism , Oncolytic Virotherapy/methods , Simplexvirus/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioblastoma/virology , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells
4.
Mol Ther ; 21(1): 68-77, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22929661

ABSTRACT

Only a subset of cancer patients inoculated with oncolytic herpes simplex virus (oHSV) type-1 has shown objective response in phase 1 and 2 clinical trials. This has raised speculations whether resistance of tumor cells to oHSV therapy may be a limiting factor. In this study, we have identified established and patient derived primary glioblastoma multiforme (GBM) stem cell lines (GSC) resistant to oHSV and also to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) that has recently shown promise in preclinical and initial clinical studies. We created a recombinant oHSV bearing a secretable TRAIL (oHSV-TRAIL) and hypothesized that oHSV-TRAIL could be used as a cancer therapeutic to target a broad spectrum of resistant tumors in a mechanism-based manner. Using the identified resistant GBM lines, we show that oHSV-TRAIL downregulates extracellular signal-regulated protein kinase (ERK)-mitogen-activated protein kinase (MAPK) and upregulates c-Jun N-terminal kinase (JNK) and p38-MAPK signaling, which primes resistant GBM cells to apoptosis via activation of caspase-8, -9, and -3. We further show that oHSV-TRAIL inhibits tumor growth and invasiveness and increases survival of mice bearing resistant intracerebral tumors without affecting the normal tissues. This study sheds new light on the mechanism by which oHSV and TRAIL function in concert to overcome therapeutic-resistance, and provides an oncolytic virus based platform to target a broad spectrum of different cancer types.


Subject(s)
Brain Neoplasms/therapy , Drug Resistance, Neoplasm , Glioblastoma/therapy , Neoplastic Stem Cells/pathology , Oncolytic Virotherapy , Animals , Apoptosis , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , MAP Kinase Signaling System , Mice , Neoplasm Invasiveness , TNF-Related Apoptosis-Inducing Ligand/metabolism
5.
Ann Surg Oncol ; 19(5): 1517-28, 2012 May.
Article in English | MEDLINE | ID: mdl-22012028

ABSTRACT

PURPOSE: We retrospectively investigated the impact of race/ethnicity on prognosis in patients who underwent surgery for colon cancer. METHODS: Surveillance, Epidemiology, and End Results population-based data on 39,210 colon cancer patients without distant metastasis who underwent radical surgery were analyzed. Prognostic impact of race/ethnicity for non-Hispanic white, Hispanic white, African American, and East Asian (Japanese, Chinese, Korean) American patients, and confounding factors of age, sex, registry region, year of diagnosis, tumor, node, metastasis system stage, tumor grade, tumor site, and the number of lymph nodes examined were analyzed by the Cox proportional hazard model. The lymph node count was analyzed and adjusted means were calculated by a generalized multiple regression model with respect to race and other factors. RESULTS: Significant differences due to race/ethnicity were observed in crude hazard ratios with respect to overall and colon cancer-specific mortality, which persisted even after adjusting for confounding factors. Adjusted hazard ratios of colon cancer-specific mortality for non-Hispanic white, Hispanic white, African American, and East Asian American patients were 1 (reference), 1.01 (95% confidence interval 0.91-1.12), 1.40 (95% confidence interval 1.31-1.50), and 0.83 (95% confidence interval 0.74-0.94), respectively. There were significant differences in crude number of lymph nodes examined among races, which were no longer significant after adjusting for covariates. CONCLUSIONS: East Asian American patients had significantly better prognosis, while African American patients had worse prognosis than non-Hispanic white patients, despite the identical adjusted number of lymph nodes examined after surgery for colon cancer. This disparity in prognosis among races/ethnicities should be taken into consideration when deciding adjuvant chemotherapy for nonwhite patients.


Subject(s)
Asian/statistics & numerical data , Black or African American/statistics & numerical data , Colonic Neoplasms/ethnology , Colonic Neoplasms/surgery , White People/statistics & numerical data , Adolescent , Adult , Aged , Colonic Neoplasms/pathology , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Analysis , United States/epidemiology , Young Adult
6.
Genes Chromosomes Cancer ; 50(3): 137-45, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21213367

ABSTRACT

MicroRNAs (miRNAs) are noncoding small RNAs that function as an endogenous regulator of gene expression. Their dysregulation has been implicated in the development of several cancers. However, the status of miRNA in soft tissue sarcomas has not yet been thoroughly investigated. This study examined the global miRNA expression in synovial sarcoma and compared the results to those in another translocation-associated sarcoma, the Ewing family of tumors, and in normal skeletal muscle. The 3D-Gene miRNA microarray platform (Toray, Kamakura, Japan) and unsupervised hierarchical clustering revealed a distinct expression pattern of miRNAs in synovial sarcoma from Ewing tumors and skeletal muscle. Thirty-five of the more than 700 miRNAs analyzed were differentially expressed in synovial sarcomas in comparison to other tissue types. There were 21 significantly up-regulated miRNAs, including some miRNAs, such as let-7e, miR-99b, and miR-125a-3p, clustered within the same chromosomal loci. Quantitative reverse transcription-polymerase chain reaction also demonstrated that these miRNAs were over-expressed in synovial sarcomas. The down-regulation of let-7e and miR-99b by anti-miR miRNA inhibitors resulted in the suppression of the proliferation of synovial sarcoma cells, and modulated the expression of their putative targets, HMGA2 and SMARCA5, suggesting that these molecules have a potential oncogenic role. The unique miRNA expression pattern including the over-expressed miRNA clusters in synovial sarcoma warrants further investigation to develop a better understanding of the oncogenic mechanisms and future therapeutic strategies for synovial sarcoma.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Sarcoma, Synovial/metabolism , Cell Line, Tumor , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Sarcoma, Synovial/genetics , Up-Regulation
7.
Cancer Res ; 69(8): 3472-81, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19351838

ABSTRACT

Glioblastoma, the most malignant type of primary brain tumor, is one of the solid cancers where cancer stem cells have been isolated, and studies have suggested resistance of those cells to chemotherapy and radiotherapy. Here, we report the establishment of CSC-enriched cultures derived from human glioblastoma specimens. They grew as neurospheres in serum-free medium with epidermal growth factor and fibroblast growth factor 2, varied in the level of CD133 expression and very efficiently formed highly invasive and/or vascular tumors upon intracerebral implantation into immunodeficient mice. As a novel therapeutic strategy for glioblastoma-derived cancer stem-like cells (GBM-SC), we have tested oncolytic herpes simplex virus (oHSV) vectors. We show that although ICP6 (UL39)-deleted mutants kill GBM-SCs as efficiently as wild-type HSV, the deletion of gamma34.5 significantly attenuated the vectors due to poor replication. However, this was significantly reversed by the additional deletion of alpha47. Infection with oHSV G47Delta (ICP6(-), gamma34.5(-), alpha47(-)) not only killed GBM-SCs but also inhibited their self-renewal as evidenced by the inability of viable cells to form secondary tumor spheres. Importantly, despite the highly invasive nature of the intracerebral tumors generated by GBM-SCs, intratumoral injection of G47Delta significantly prolonged survival. These results for the first time show the efficacy of oHSV against human GBM-SCs, and correlate this cytotoxic property with specific oHSV mutations. This is important for designing new oHSV vectors and clinical trials. Moreover, the new glioma models described in this study provide powerful tools for testing experimental therapeutics and studying invasion and angiogenesis.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/therapy , Embryonic Stem Cells/pathology , Glioblastoma/pathology , Glioblastoma/therapy , Neoplastic Stem Cells/pathology , Oncolytic Virotherapy/methods , Simplexvirus/physiology , Animals , Brain Neoplasms/virology , Embryonic Stem Cells/virology , Glioblastoma/virology , Humans , Mice , Mutation , Neoplastic Stem Cells/virology , Simplexvirus/genetics , Simplexvirus/growth & development , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
8.
Curr Opin Mol Ther ; 9(5): 447-66, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17932809

ABSTRACT

HSV-1 was one of the first oncolytic viruses to have been investigated for its therapeutic potential against cancer. Among the dozens of oncolytic HSV-1 vectors that have so far been reported, some carry transgenes, including interleukins, anti-angiogenic peptides, and prodrug-converting enzymes. 'Arming' of HSV-1 with therapeutic transgenes such as these is expected to enhance its efficacy. HSV-1 is a 152-kb double-stranded DNA virus, and the generation of armed HSV usually takes several months to achieve by conventional homologous recombination methods. Recently, bacterial artificial chromosome (BAC)-based systems termed 'HSVQuik' and 'Flip-Flop HSV-BAC' were developed to enable the fast generation of recombinant HSV-1 vectors within weeks by using site-specific recombinases. These systems provide powerful tools for screening potential transgenes that might greatly enhance the efficacy of HSV-1 vectors. This review discusses the current state of research into the development of oncolytic HSV-1 vectors, and highlights the promise that armed oncolytic HSV-1 vectors might hold for the future.


Subject(s)
Chromosomes, Artificial, Bacterial , Genetic Vectors/chemical synthesis , Herpesvirus 1, Human/genetics , Oncolytic Viruses/genetics , Animals , Genetic Engineering , Humans
9.
Clin Cancer Res ; 12(22): 6791-9, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121900

ABSTRACT

PURPOSE: Oncolytic herpes simplex viruses (HSV) appear to be a promising platform for cancer therapy. However, efficacy as single agents has thus far been unsatisfactory. Fibroblast growth factor (FGF) signaling is important for the growth and migration of endothelial and tumor cells. Here, we examine the strategy of arming oncolytic HSV with a dominant-negative FGF receptor (dnFGFR) that targets the FGF signaling pathway. EXPERIMENTAL DESIGN: A mouse Nf1:p53 malignant peripheral nerve sheath tumor (MPNST) cell line expressing dnFGFR was generated by transfection. The effects of dnFGFR expression on cell growth and migration in vitro and tumor formation in vivo were determined. The dnFGFR transgene was then inserted into oncolytic HSV G47Delta using a bacterial artificial chromosome construction system. Antitumoral and antiangiogenic activities of bG47Delta-dnFGFR were examined. RESULTS: MPNST 61E4 cells expressing dnFGFR grew less well than parental control cells. bG47Delta-dnFGFR showed enhanced killing of both tumor (human U87 glioma and F5 malignant meningioma cells and murine MPNST 61E4 and 37-3-18-4 cells) and proliferating endothelial cells (human umbilical vascular endothelial cell and Py-4-1) in vitro compared with the control vector bG47Delta-empty without inhibiting viral replication. In vivo, bG47Delta-dnFGFR was more efficacious than its nonexpressing parent bG47Delta-empty at inhibiting tumor growth and angiogenesis in both human U87 glioma and mouse 37-3-18-4 MPNST tumors in nude mice. CONCLUSIONS: By using multiple therapeutic mechanisms, including destruction of both tumor cells and tumor endothelial cells, an oncolytic HSV encoding dnFGFR enhances antitumor efficacy. This strategy can be applied to other oncolytic viruses and for clinical translation.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Oncolytic Virotherapy/methods , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics , Recombinant Proteins/therapeutic use , Simplexvirus , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Movement , Cell Proliferation , Chick Embryo , Endothelium, Vascular/metabolism , Genes, Dominant , Genetic Vectors/physiology , Genetic Vectors/therapeutic use , Humans , Mice , Mice, Nude , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Simplexvirus/genetics , Simplexvirus/physiology , Treatment Outcome , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
10.
Cancer Res ; 66(20): 10127-35, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047077

ABSTRACT

The Wnt/beta-catenin/T-cell factor (Tcf) pathway is aberrantly up-regulated in the majority of colorectal cancers (CRC) and hepatoblastomas due to either an APC or beta-catenin gene mutation. We constructed synthetic promoters, T and TE, which contain tandem repeats of a Tcf responsive element without and with the human 4F2 gene intronic enhancer, respectively. Although the T and TE promoters showed higher transcriptional activity than a control promoter in all CRC and hepatoblastoma cell lines tested, with low activities in most other tumor cell lines, the level of transcription varied considerably among the CRC and hepatoblastoma cell lines. In some CRC cell lines, the TE promoter displayed higher levels of transcription than even the human CMV(IE) promoter. In those CRC cells, the APC gene mutations were located within a small segment between the first and second 20-amino-acid repeats in the mutation cluster region of the APC protein. We created a transcriptionally targeted oncolytic herpes simplex virus vector (bM24-TE) in which replication is driven by the TE promoter. This vector efficiently and specifically replicated in and killed tumor cells with strong beta-catenin/Tcf signaling. Intratumoral injection of bM24-TE significantly reduced the growth of highly beta-catenin active SW480 CRC tumors and induced a complete response in half of them, whereas it had no effect on the growth of beta-catenin-inactive A549 tumors. Our results suggest that a transcriptionally regulated oncolytic herpes vector targeting beta-catenin/Tcf signal is very efficacious against CRC tumors carrying an APC gene mutation between the first and second 20-amino-acid repeats.


Subject(s)
Carcinoma, Hepatocellular/therapy , Colorectal Neoplasms/therapy , Liver Neoplasms/therapy , Oncolytic Virotherapy/methods , Simplexvirus/genetics , TCF Transcription Factors/genetics , beta Catenin/genetics , Animals , Base Sequence , Caco-2 Cells , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Chlorocebus aethiops , Colorectal Neoplasms/genetics , Colorectal Neoplasms/virology , Female , Genetic Vectors/genetics , HCT116 Cells , HeLa Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Promoter Regions, Genetic , Tandem Repeat Sequences , Transcription, Genetic , Vero Cells , Xenograft Model Antitumor Assays
11.
Mol Ther ; 14(6): 789-97, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17045531

ABSTRACT

Oncolytic herpes simplex viruses (HSV) have emerged as a promising platform for cancer therapy. However, efficacy as single agents has thus far been unsatisfactory. Tumor vasculature is critical in supporting tumor growth, but successful antiangiogenic approaches often require maintaining constant levels of antiangiogenic products. We hypothesized that oncolytic HSV has the potential to destroy tumor vasculature and that this effect can be enhanced by combination with antiangiogenic gene transfer. We examined the strategy of arming oncolytic HSV with an antiangiogenic transgene, platelet factor 4 (PF4). The PF4 transgene was inserted into oncolytic HSV G47Delta utilizing a bacterial artificial chromosome construction system. Whereas bG47Delta-empty showed robust cell killing and migration inhibition of proliferating endothelial cells (HUVEC and Py-4-1), the effect was further enhanced by PF4 expression. Importantly, enhanced potency did not impede viral replication. In vivo, bG47Delta-PF4 was more efficacious than its nonexpressing parent bG47Delta-empty at inhibiting tumor growth and angiogenesis in both human U87 glioma and mouse 37-3-18-4 malignant peripheral nerve sheath tumor models. Enhancing the antiangiogenic properties of oncolytic HSV through the expression of antiangiogenic factors such as PF4 is a powerful new strategy that targets both the tumor cells and tumor vasculature.


Subject(s)
Glioma/therapy , Oncolytic Virotherapy/methods , Platelet Factor 4/genetics , Simplexvirus/genetics , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/physiology , Animals , Cell Line , Cell Line, Tumor , Cell Survival , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/genetics , Glioma/genetics , Glioma/pathology , Humans , Mice , Neoplasms/genetics , Neoplasms/therapy , Oncolytic Viruses/genetics , Platelet Factor 4/physiology , Time Factors , Transfection , Treatment Outcome , Virus Replication , Xenograft Model Antitumor Assays
12.
BMC Biotechnol ; 6: 40, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16995942

ABSTRACT

BACKGROUND: Oncolytic herpes simplex virus (HSV) vectors that specifically replicate in and kill tumor cells sparing normal cells are a promising cancer therapy. Traditionally, recombinant HSV vectors have been generated through homologous recombination between the HSV genome and a recombination plasmid, which usually requires laborious screening or selection and can take several months. Recent advances in bacterial artificial chromosome (BAC) technology have enabled cloning of the whole HSV genome as a BAC plasmid and subsequent manipulation in E. coli. Thus, we sought a method to generate recombinant oncolytic HSV vectors more easily and quickly using BAC technology. RESULTS: We have developed an HSV-BAC system, termed the Flip-Flop HSV-BAC system, for the rapid generation of oncolytic HSV vectors. This system has the following features: (i) two site-specific recombinases, Cre and FLPe, are used sequentially to integrate desired sequences and to excise the BAC sequences, respectively; and (ii) the size of the HSV-BAC-insert genome exceeds the packaging limit of HSV so only correctly recombined virus grows efficiently. We applied this to the construction of an HSV-BAC plasmid that can be used for the generation of transcriptionally-targeted HSV vectors. BAC sequences were recombined into the UL39 gene of HSV ICP4-deletion mutant d120 to generate M24-BAC virus, from which HSV-BAC plasmid pM24-BAC was isolated. An ICP4 expression cassette driven by an exogenous promoter was re-introduced to pM24-BAC by Cre-mediated recombination and nearly pure preparations of recombinant virus were obtained typically in two weeks. Insertion of the ICP4 coding sequence alone did not restore viral replication and was only minimally better than an ICP4-null construct, whereas insertion of a CMVIE promoter-ICP4 transgene (bM24-CMV) efficiently drove viral replication. The levels of bM24-CMV replication in tumor cells varied considerably compared to hrR3 (UL39 mutant). CONCLUSION: Our Flip-Flop HSV-BAC system enables rapid generation of HSV vectors carrying transgene inserts. By introducing a tumor-specific-promoter-driven ICP4 cassette into pM24-BAC using this system, one should be able to generate transcriptionally-targeted oncolytic HSV vectors. We believe this system will greatly facilitate the screening of a plethora of clinically useful tumor-specific promoters in the context of oncolytic HSV vectors.


Subject(s)
Genetic Vectors , Oncolytic Viruses/genetics , Recombinases/genetics , Simplexvirus/genetics , Chromosomes, Artificial, Bacterial/genetics , Cloning, Molecular
13.
Proteomics ; 6(17): 4845-55, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16878292

ABSTRACT

The proteomic analysis of serum (plasma) has been a major approach to determining biomarkers essential for early disease diagnoses and drug discoveries. The determination of these biomarkers, however, is analytically challenging since the dynamic concentration range of serum proteins/peptides is extremely wide (more than 10 orders of magnitude). Thus, the reduction in sample complexity prior to proteomic analyses is essential, particularly in analyzing low-abundance protein biomarkers. Here, we demonstrate a novel approach to the proteomic analyses of human serum that uses an originally developed serum protein separation device and a sequentially linked 3-D-LC-MS/MS system. Our hollow-fiber-membrane-based serum pretreatment device can efficiently deplete high-molecular weight proteins and concentrate low-molecular weight proteins/peptides automatically within 1 h. Four independent analyses of healthy human sera pretreated using this unique device, followed by the 3-D-LC-MS/MS successfully produced 12 000-13 000 MS/MS spectra and hit around 1800 proteins (>95% reliability) and 2300 proteins (>80% reliability). We believe that the unique serum pretreatment device and proteomic analysis protocol reported here could be a powerful tool for searching physiological biomarkers by its high throughput (3.7 days per one sample analysis) and high performance of finding low abundant proteins from serum or plasma samples.


Subject(s)
Blood Proteins/analysis , Blood Proteins/chemistry , Blood Proteins/isolation & purification , Blood Proteins/metabolism , Chromatography, Liquid , Humans , Mass Spectrometry , Molecular Weight , Sensitivity and Specificity
14.
Cancer Res ; 65(23): 10663-8, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322208

ABSTRACT

Conditionally replicating herpes simplex virus type 1 (HSV-1) vectors are promising therapeutic agents for cancer. Certain antitumor functions may be added to oncolytic activities of recombinant HSV-1 vectors by inserting transgenes into the viral genome. Because conventional homologous recombination techniques had required time-consuming processes to create "armed" oncolytic HSV-1 vectors, we established an innovative construction system using bacterial artificial chromosome and two recombinase systems (Cre/loxP and FLPe/FRT). Using G47Delta, a safe and efficacious oncolytic HSV-1 with triple gene mutations, as the backbone, this system allowed a rapid generation of multiple vectors with desired transgenes inserted in the deleted ICP6 locus. Four oncolytic HSV-1 vectors, expressing murine interleukin 18 (mIL-18), soluble murine B7-1 [B7-1-immunoglobulin (B7-1-Ig)], both, or none, were created simultaneously within 3 months. In vitro, all newly created recombinant vectors exhibited virus yields and cytopathic effects similar to the parental G47Delta. In two immunocompetent mouse tumor models, TRAMP-C2 prostate cancer and Neuro2a neuroblastoma, the vector expressing both mIL-18 and B7-1-Ig showed a significant enhancement of antitumor efficacy via T-cell-mediated immune responses. The results show that "arming" with multiple transgenes can improve the efficacy of oncolytic HSV-1 vectors. The use of our system may facilitate the development and testing of various armed oncolytic HSV-1 vectors.


Subject(s)
B7-1 Antigen/genetics , Herpesvirus 1, Human/genetics , Interleukin-18/genetics , Oncolytic Virotherapy/methods , Animals , Chlorocebus aethiops , Chromosomes, Artificial, Bacterial , Gene Deletion , Genetic Vectors/genetics , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Neuroblastoma/genetics , Neuroblastoma/therapy , Neuroblastoma/virology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Prostatic Neoplasms/virology , Recombinases/genetics , Transfection , Vero Cells
15.
Med Electron Microsc ; 32(1): 20-24, 1999 May.
Article in English | MEDLINE | ID: mdl-11810421

ABSTRACT

Spa-1-like protein (SPAL) is a novel GTPase-activating protein (GAP) family protein. We generated anti-SPAL antibody, and examined localization of SPAL in the rat esophagus and heart by immunofluorescence microscopy. In the esophagus, SPAL was highly expressed in the spinous and granular layers and localized at the cell-cell border of the epithelial cells. SPAL in the spinous layer was almost colocalized with beta-catenin, an intracellular anchor protein of E-cadherin, while colocalization of SPAL with the tight junction protein ZO-1 was shown in the granular layer. In the heart, SPAL was localized at the intercalated disks, and there colocalized with both beta-catenin and ZO-1. In addition, much punctate localization of SPAL was seen along lateral borders of the cardiomyocytes, where beta-catenin and ZO-1 were absent. From these results, it is suggested that SPAL may regulate cell-cell adhesion in the rat esophagus and heart.

SELECTION OF CITATIONS
SEARCH DETAIL
...