Try a new search

Format these results:

Searched for:

person:merued01

in-biosketch:yes

Total Results:

151


Restricted tissue tropism and acquired resistance to Sindbis viral vector expression in the absence of innate and adaptive immunity

Tseng, J-C; Zheng, Y; Yee, H; Levy, D E; Meruelo, D
Our previous studies suggest that replication-defective Sindbis vectors might be promising agents for specific tumor targeting and detection. However, the effects of innate and/or adaptive anti-viral immunity, in particular, the IFN-I/STAT1 signaling pathway, may impact their therapeutic potential. Using a bioluminescent imaging system, we demonstrate that although most normal cells are not permissively transduced by replication-defective Sindbis vector, transduction of liver non-sinusoidal endothelial occurs the first time IFN-I/STAT1 signaling deficient mice are inoculated with the vector. Transduction of some cells is not surprising since STAT1 knockout animals show significant delay in IFN responses such as the production of IFN-alpha/beta and transcriptional activation of several anti-viral genes (IRF7, RIG-I, PKR, TLR3, USP18, ISG15). However, beyond the initial vector transduction, which resolves rapidly, secondary inoculums of Sindbis vectors do not transduce any liver cells, suggesting that an alternative antiviral pathway may protect against further transduction. Other known signaling pathways were examined using mice lacking functional TLR3, tumor necrosis factoralphaR or nuclear factor-kappa B (p50). Surprisingly, none of those pathways seem to play a significant role in anti-Sindbis responses. Thus it appears that in vivo, in contrast to the ready transduction of tumor cells, transduction of normal cells by replication-defective Sindbis vector is limited, possibly by a novel mechanism
PMID: 17508007
ISSN: 0969-7128
CID: 74661

Tumor-specific in vivo transfection with HSV-1 thymidine kinase gene using a Sindbis viral vector as a basis for prodrug ganciclovir activation and PET

Tseng, Jen-Chieh; Zanzonico, Pat B; Levin, Brandi; Finn, Ronald; Larson, Steven M; Meruelo, Daniel
One type of gene therapy of tumors, gene-directed enzyme-prodrug therapy (GDEPT), holds considerable promise, although practical considerations limit its clinical applicability. These include the lack of acceptable noninvasive methods that are adaptable to humans for selective tumor targeting of the therapeutic genetic material. Sindbis virus is an oncolytic, alpha-virus that selectively targets tumors through the 67-kDa laminin receptor (LAMR). In this report we describe a novel approach that permits tumor-selective tumor targeting and quantitative in vivo monitoring using PET of a commonly applied GDEPT, based on herpes simplex virus thymidine kinase type 1 (HSVtk) and ganciclovir (GCV). METHODS: Sindbis/tk vectors were harvested from the supernatant of in vitro cultures of a packaging cell produced by electroporation of both replicon RNA (SinRep5/tk) and helper RNA (DH-BB) into baby hamster kidney (BHK) cells. The therapeutic effect of GCV was determined by incubation of transfected tumor cells with increasing concentrations of GCV. BHK tumors growing as xenografts in severe combined immunodeficiency disease (SCID) mice were transfected by parenteral administration of the vector. Imaging was performed using small-animal PET at 2 h after injection of 18F fluoro-ethyl-arabinosyluridine (18F-FEAU) and 24 h after the final parenteral injection of Sindbis/tk viral vector. RESULTS: The vector efficiently expresses the HSVtk enzyme in infected tumor cells, both in vitro and in vivo. High levels of HSVtk expression ensure sufficient prodrug GCV conversion and activation for bystander effects that kill the surrounding untransduced tumor cells. Tumor localization of intravenously administered 18F-FEAU after 2 and 3 parenteral vector treatments of Sindbis/tk demonstrated uptake of 1.7 and 3.1 %ID/g (percentage injected dose per gram), respectively. CONCLUSION: The vector efficiently targets the HSVtk enzyme gene into Sindbis-infected tumor cells. High levels of HSVtk expression ensure sufficient prodrug GCV conversion and activation for bystander effects that killed many surrounding untransduced tumor cells. In addition, the HSVtk activities in tumors can be noninvasively monitored using PET after systemic Sindbis/tk treatments as a basis for determining the levels and tissue distribution of vector, noninvasively in living animals, and for optimizing in vivo transfection rates of tumor
PMID: 16818948
ISSN: 0161-5505
CID: 67390

Gene therapy that safely targets and kills tumor cells throughout the body

Hurtado, Alicia; Tseng, Jen-Chieh; Meruelo, Daniel
The authors studied the therapeutic value of Sindbis vectors for advanced metastatic cancer by using a variety of clinically accurate mouse models and demonstrated through imaging, histological, and molecular data that Sindbis vectors systemically and specifically infect/detect and kill metastasized tumors in vivo, leading to significant suppression of tumor growth and enhanced survival. Use of two different bioluminescent genetic markers for the IVIS Imaging System (Xenogen Corp., Alameda, CA) permitted demonstration of an excellent correlation between vector delivery and metastatic locations in vivo. Sindbis tumor specificity is not attributable to a species difference between human tumor and mouse normal cells. Sindbis virus is known to infect mammalian cells using the Mr 67,000 laminin receptor, which is elevated in tumor versus normal cells, and downregulated expression of laminin receptor with small interfering RNA significantly reduces the infectivity of Sindbis vectors. Tumor overexpression of the laminin receptor may explain the specificity and efficacy that Sindbis vectors demonstrate for tumor cells in vivo. Laser capture microdissection of mouse tumor implants showed equivalent laminin receptor expression levels in the different tumor metastases in the peritoneal cavity. Incorporation of antitumor cytokine genes such as interleukin-12 and interleukin-15 genes enhances the efficacy of the vector. These results suggest that Sindbis viral vectors may be promising agents for both specific detection and growth suppression of metastatic ovarian cancer
PMID: 16608394
ISSN: 1549-1684
CID: 64391

Identification of amino acids of Sindbis virus E2 protein involved in targeting tumor metastases in vivo

Hurtado, Alicia; Tseng, Jen-Chieh; Boivin, Christopher; Levin, Brandi; Yee, Herman; Pampeno, Christine; Meruelo, Daniel
Previous studies conducted in our laboratory with Sindbis viral vectors in animal models demonstrated excellent in vivo targeting of tumor cells and significant reduction of metastatic implant size. To explore the influence of Sindbis strain on these factors, we constructed new plasmids from the wild-type Ar-339 Sindbis virus strain and compared their sequences. We found differences in the replicase and envelope proteins between JT, HRSP, and Ar-339 sequences. We made chimeras combining both strains and studied their efficiency in SCID mice bearing tumor xenograft using IVIS in vivo imaging techniques. We found that JT envelope proteins targeted tumors more efficiently than those of Ar-339, while the Ar-339 replicase showed increased efficacy in tumor reduction. To determine which residues are responsible for tumor targeting, we made mutants of Ar-339 E2 envelope protein and tested them by IVIS imaging in ES-2 tumor-bearing and tumor-free mice. The change of only one amino acid from E70 to K70 in Ar-339 E2 suppressed the ability to target metastatic tumor implants in mice. A K70 and V251 double E2 mutant did not reverse the loss of targeting capability. Only the mutant with JT E2 and Ar-339 helper targeted tumor, though with less intensity
PMID: 16109508
ISSN: 1525-0016
CID: 61365

The effect of Sindbis viral vectors on the N-nitroso-bis(2-oxopropyl)amine(BOP) induced hamster pancreatic ductal adenocarcinoma and cholangiocarcinoma [Meeting Abstract]

Liu, Q; West, AB; Merali, S; Levin, B; Meruelo, D; Pellicer, A
ISI:000226117901463
ISSN: 0893-3952
CID: 50445

The effect of sindbis viral vectors on the N-nitroso-bis(2-oxopropyl)amine(BOP) induced hamster pancreatic ductal adenocarcinoma and cholangiocarcinoma [Meeting Abstract]

Lin, Q; West, AB; Merali, S; Levin, B; Meruelo, D; Pellicer, A
ISI:000226238601483
ISSN: 0023-6837
CID: 50475

Anti-angiogenic activities of hypericin in vivo: potential for ophthalmologic applications

Lavie, Gad; Mandel, Mathilda; Hazan, Sadick; Barliya, Tilda; Blank, Michael; Grunbaum, Aaron; Meruelo, Daniel; Solomon, Arieh
Hypericin, a perihydroxylated dianthraquinone is shown here to be a highly potent inhibitor of angiogenesis in several ocular models examined in rat eyes. Extensive angiogenesis induced in the cornea and iris by intra-ocular administration of FGF-2 was effectively inhibited by a minimum of four dose regimens of hypericin (2 mg/kg) administered via the intraperitoneal route at 48 h intervals. Maximal inhibition was achieved when animal treatment with hypericin was initiated 48 h prior to inoculation of FGF-2. The molecular basis for the hypericin-mediated inhibition of angiogenesis in the anterior eye compartment appears to involve several sites in the cascade leading to angiogenesis. We show that the activating phosphorylation of extracellular signal-regulated MAP kinases (ERK1/2) is inhibited by hypericin in human retinal pigment epithelial cells and in EA.hy926 cells, an endothelial hybridoma expressing endothelial cell properties. ERK1/2 activity is required for the transactivation of hypoxia-inducible factor 1 alpha (HIF-1alpha) and in VEGF-induced blood vessel sprouting. MT1-MMP activity in human microvascular endothelial cells was also inhibited. The findings identify hypericin as a potentially useful agent in the treatment of ophthalmic neovascularization pathogeneses
PMID: 16132616
ISSN: 0969-6970
CID: 64515

Using sindbis viral vectors for specific detection and suppression of advanced ovarian cancer in animal models

Tseng, Jen-Chieh; Hurtado, Alicia; Yee, Herman; Levin, Brandi; Boivin, Christopher; Benet, Marta; Blank, Stephanie V; Pellicer, Angel; Meruelo, Daniel
We studied the therapeutic value of Sindbis vectors for advanced metastatic ovarian cancer by using two highly reproducible and clinically accurate mouse models: a SCID xenograft model, established by i.p. inoculation of human ES-2 ovarian cancer cells, and a syngenic C57BL/6 model, established by i.p. inoculation of mouse MOSEC ovarian cancer cells. We demonstrate through imaging, histologic, and molecular data that Sindbis vectors systemically and specifically infect/detect and kill metastasized tumors in the peritoneal cavity, leading to significant suppression of the carcinomatosis in both animal models. Use of two different bioluminescent genetic markers for the IVIS Imaging System permitted demonstration, for the first time, of an excellent correlation between vector delivery and metastatic locations in vivo. Sindbis vector infection and growth suppression of murine MOSEC tumor cells indicate that Sindbis tumor specificity is not attributable to a species difference between human tumor and mouse normal cells. Sindbis virus is known to infect mammalian cells using the M(r) 67,000 laminin receptor. Immunohistochemical staining of tumor cells indicates that laminin receptor is elevated in tumor versus normal cells. Down-regulated expression of laminin receptor with small interfering RNA significantly reduces the infectivity of Sindbis vectors. Tumor overexpression of the laminin receptor may explain the specificity and efficacy that Sindbis vectors demonstrate for tumor cells in vivo. We show that incorporation of antitumor cytokine genes such as interleukin-12 and interleukin-15 genes enhances the efficacy of the vector. These results suggest that Sindbis viral vectors may be promising agents for both specific detection and growth suppression of metastatic ovarian cancer
PMID: 15374985
ISSN: 0008-5472
CID: 44950

Antimetastatic activity of the photodynamic agent hypericin in the dark

Blank, Michael; Lavie, Gad; Mandel, Mathilda; Hazan, Sadick; Orenstein, Arie; Meruelo, Daniel; Keisari, Yona
A unique property of the photodynamic signal transduction inhibitor hypericin (HY) is high functionality in the dark, which has been shown to result in portfolio of anticancer activities both in vitro and in vivo. Here we show that treatment with HY significantly reduces growth rate of metastases in 2 murine models: breast adenocarcinoma (DA3) and squamous cell carcinoma (SQ2). Focus on metastases was achieved by resection of primary tumors at stages in which micrometastases exist in lungs. Long-term animal survival in DA3 tumor-excised groups increased from 15.6% in controls to 34.5% following supplementary treatment with HY. In mice bearing SQ2 tumor metastases, therapy with HY increased animal survival from 17.7% in controls to 46.1%. Using Laser-induced fluorescence and multipixel spectral image analyses, we demonstrate that HY has a high tendency to accumulate in primary and metastatic tumors; HY content in lungs bearing metastases was approximately 2-fold higher than in the lungs of healthy animals. The tendency of HY to preferentially concentrate in lung metastases, combined with its potent antiproliferative activities, may render HY as a useful supplementary modality in the treatment of metastatic cancer irrespective of photoactivation
PMID: 15239139
ISSN: 0020-7136
CID: 44810

Systemic gene therapy by Sindbis vectors: A potentially safe and effective targeted therapy for identifying and killing tumor cells in vivo

Meruelo, Daniel
Extract: A major obstacle to the development of gene therapy for cancer has been the inability to specifically and systemically deliver gene therapy vectors throughout the body to primary and/or metastasized tumor cells. Although intratumor injections of gene therapy vectors have sometimes been possible, no viral vector has been available that could be administered systemically and would selectively and efficiently target tumors without infection of normal tissues. Furthermore, even when locally injected, many viral vectors end up at high concentrations in the liver, because many cells of the body have low receptor numbers for some of the vectors in current use, whereas the liver has high numbers of such receptors. A number of ingenious approaches have been tried but none so far have fully resolved the problem. For example, tumor-specific promoters have been incorporated into the vectors so that gene expression and/or replication can occur in tumor cells but not normal cells. Unfortunately, only a small fraction of these vectors are typically taken up by the target tumor and expressed. In such cases, tumor cell death is generally insufficient to eradicate or significantly slow tumor growth
PMID: 20705022
ISSN: 1944-7930
CID: 111829