Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Shanthi Ganesh is active.

Publication


Featured researches published by Shanthi Ganesh.


Cancer Research | 2007

Relaxin-expressing, fiber chimeric oncolytic adenovirus prolongs survival of tumor-bearing mice.

Shanthi Ganesh; Melissa Gonzalez Edick; Neeraja Idamakanti; Marina Abramova; Melinda VanRoey; Michael L. Robinson; Chae-Ok Yun; Karin Jooss

Selective replication of oncolytic viruses in tumor cells provides a promising approach for the treatment of human cancers. One of the limitations observed with oncolytic viruses currently used in the treatment of solid tumors is the inefficient spread of virus throughout the tumor mass following intratumoral injection. Data are presented showing that oncolytic adenoviruses expressing the relaxin gene and containing an Ad5/Ad35 chimeric fiber showed significantly enhanced transduction and increased virus spread throughout the tumor when compared with non-relaxin-expressing, Ad5-based viruses. The increased spread of such viruses throughout tumors correlated well with improved antitumor efficacy and overall survival in two highly metastatic tumor models. Furthermore, nonreplicating viruses expressing relaxin did not increase metastases, suggesting that high level expression of relaxin will not enhance metastatic spread of tumors. In summary, the data show that relaxin may play a role in rearranging matrix components within tumors, which helps recombinant oncolytic adenoviruses to spread effectively throughout the tumor mass and thereby increase the extent of viral replication within the tumor. Expressing relaxin from Ad5/Ad35 fiber chimeric adenoviruses may prove a potent and novel approach to treating patients with cancer.


Clinical Cancer Research | 2008

Intratumoral Coadministration of Hyaluronidase Enzyme and Oncolytic Adenoviruses Enhances Virus Potency in Metastatic Tumor Models

Shanthi Ganesh; Melissa Gonzalez-Edick; Douglas Gibbons; Melinda Van Roey; Karin Jooss

Purpose: Evaluate the codelivery of hyaluronidase enzyme with oncolytic adenoviruses to determine whether it improves the spread of the virus throughout tumors, thereby leading to a greater overall antitumor efficacy in tumor models. Experimental Design: The optimal dose of hyaluronidase that provided best transduction efficiency and spread of a green fluorescent protein (GFP)-expressing adenovirus within tumors was combined with oncolytic viruses in tumor models to determine whether the combination treatment results in an improvement of antitumor efficacy. Results: In mice injected with the adenovirus Ad5/35GFP and an optimal dose of hyaluronidase (50 U), a significant increase in the number of GFP-expressing cells was observed when compared with animals injected with virus only (P < 0.0001). When the oncolytic adenoviruses Ad5OV or Ad5/35 OV (OV-5 or OV5T35H) were codelivered with 50 U of hyaluronidase, a significant delay in tumor progression was observed, which translated into a significant increase in the mean survival time of tumor-bearing mice compared with either of the monotherapy-treated groups (P < 0.0001). Furthermore, the mice that received the combination of Ad5/35 OV and hyaluronidase showed the best antitumor efficacy. Importantly, the combination treatment did not increase the metastatic potential of the tumors. Lastly, the increase in virus potency observed in animals injected with both enzyme and virus correlated with enhanced virus spread throughout tumors. Conclusion: Antitumor activity and overall survival of mice bearing highly aggressive tumors are significantly improved by codelivery of oncolytic adenoviruses and hyaluronidase when compared with either of the monotherapy-treated groups, and it may prove to be a potent and novel approach to treating patients with cancer.


Hepatology | 2017

Targeting β‐catenin in hepatocellular cancers induced by coexpression of mutant β‐catenin and K‐Ras in mice

Junyan Tao; Rong Zhang; Sucha Singh; Minakshi Poddar; Emily Xu; Michael Oertel; Xin Chen; Shanthi Ganesh; Marc Abrams; Satdarshan P.S. Monga

Recently, we have shown that coexpression of hMet and mutant‐β‐catenin using sleeping beauty transposon/transposase leads to hepatocellular carcinoma (HCC) in mice that corresponds to around 10% of human HCC. In the current study, we investigate whether Ras activation, which can occur downstream of Met signaling, is sufficient to cause HCC in association with mutant‐β‐catenin. We also tested therapeutic efficacy of targeting β‐catenin in an HCC model. We show that mutant‐K‐Ras (G12D), which leads to Ras activation, cooperates with β‐catenin mutants (S33Y, S45Y) to yield HCC in mice. Affymetrix microarray showedu2009>u200990% similarity in gene expression in mutant‐K‐Ras‐β‐catenin and Met‐β‐catenin HCC. K‐Ras‐β‐catenin tumors showed up‐regulation of β‐catenin targets like glutamine synthetase (GS), leukocyte cell‐derived chemotaxin 2, Regucalcin, and Cyclin‐D1 and of K‐Ras effectors, including phosphorylated extracellular signal‐regulated kinase, phosphorylated protein kinase B, phosphorylated mammalian target of rapamycin, phosphorylated eukaryotic translation initiation factor 4E, phosphorylated 4E‐binding protein 1, and p‐S6 ribosomal protein. Inclusion of dominant‐negative transcription factor 4 at the time of K‐Ras‐β‐catenin injection prevented HCC and downstream β‐catenin and Ras signaling. To address whether targeting β‐catenin has any benefit postestablishment of HCC, we administered K‐Ras‐β‐catenin mice with EnCore lipid nanoparticles (LNP) loaded with a Dicer substrate small interfering RNA targeting catenin beta 1 (CTNNB1; CTNNB1‐LNP), scrambled sequence (Scr‐LNP), or phosphate‐buffered saline for multiple cycles. A significant decrease in tumor burden was evident in the CTNNB1‐LNP group versus all controls, which was associated with dramatic decreases in β‐catenin targets and some K‐Ras effectors, leading to reduced tumor cell proliferation and viability. Intriguingly, in relatively few mice, non‐GS‐positive tumors, which were evident as a small subset of overall tumor burden, were not affected by β‐catenin suppression. Conclusion: Ras activation downstream of c‐Met is sufficient to induce clinically relevant HCC in cooperation with mutant β‐catenin. β‐catenin suppression by a clinically relevant modality is effective in treatment of β‐catenin‐positive, GS‐positive HCCs. (Hepatology 2017;65:1581‐1599)


Molecular Cancer Therapeutics | 2016

Direct Pharmacological Inhibition of β-Catenin by RNA Interference in Tumors of Diverse Origin.

Shanthi Ganesh; Martin Koser; Wendy Cyr; Girish Chopda; Junyan Tao; Xue Shui; Bo Ying; Dongyu Chen; Purva Pandya; Edmond Chipumuro; Zakir Siddiquee; Kevin Craig; Cheng-Jung Lai; Henryk T. Dudek; Satdarshan P.S. Monga; Weimin Wang; Bob D. Brown; Marc Abrams

The Wnt/β-catenin pathway is among the most frequently altered signaling networks in human cancers. Despite decades of preclinical and clinical research, efficient therapeutic targeting of Wnt/β-catenin has been elusive. RNA interference (RNAi) technology silences genes at the mRNA level and therefore can be applied to previously undruggable targets. Lipid nanoparticles (LNP) represent an elegant solution for the delivery of RNAi-triggering oligonucleotides to disease-relevant tissues, but have been mostly restricted to applications in the liver. In this study, we systematically tuned the composition of a prototype LNP to enable tumor-selective delivery of a Dicer-substrate siRNA (DsiRNA) targeting CTNNB1, the gene encoding β-catenin. This formulation, termed EnCore-R, demonstrated pharmacodynamic activity in subcutaneous human tumor xenografts, orthotopic patient-derived xenograft (PDX) tumors, disseminated hematopoietic tumors, genetically induced primary liver tumors, metastatic colorectal tumors, and murine metastatic melanoma. DsiRNA delivery was homogeneous in tumor sections, selective over normal liver and independent of apolipoprotein-E binding. Significant tumor growth inhibition was achieved in Wnt-dependent colorectal and hepatocellular carcinoma models, but not in Wnt-independent tumors. Finally, no evidence of accelerated blood clearance or sustained liver transaminase elevation was observed after repeated dosing in nonhuman primates. These data support further investigation to gain mechanistic insight, optimize dose regimens, and identify efficacious combinations with standard-of-care therapeutics. Mol Cancer Ther; 15(9); 2143–54. ©2016 AACR.


Human Gene Therapy | 2009

Evaluation of Biodistribution of a Fiber-Chimeric, Conditionally Replication-Competent (Oncolytic) Adenovirus in CD46 Receptor Transgenic Mice

Shanthi Ganesh; Melissa Gonzalez-Edick; Douglas Gibbons; Jeff Waugh; Melinda Van Roey; Karin Jooss

The limited efficacy of adenovirus type 5 (Ad5)-based oncolytic viruses seen in the clinic thus far may be attributable in part to variable expression of its receptor on tumor cells. Replacement of the Ad5 fiber knob with the Ad35 fiber knob generated the Ad5/35 chimeric virus, which has previously been demonstrated to have significant antitumor activity in murine tumor models, presumably by virtue of its recognition of the CD46 receptor, which is abundant on many types of tumor cells. In the current study, a CD46 receptor transgenic mouse strain (hCD46Ge) that expresses the CD46 receptor in a pattern closely mirroring that in humans was used to study the in vivo properties of Ad5/Ad35 chimeric viruses. Vector distribution was evaluated after intravenous administration to hCD46Ge mice of an Ad5-based oncolytic adenovirus or an Ad5/35 chimeric oncolytic adenovirus (designated OV-5 and OV-5T35H, respectively), a wild-type Ad5 virus (Ad5wt), or an Ad5-based, E1-deleted adenovirus (Addl312) at 1.25 x 10(12) viral particles/kg. The amount of OV-5T35H vector genomes in the liver was at least two orders of magnitude lower than that of Ad5-based viruses. Moreover, animals injected with OV-5T35H virus had significantly lower elevations of serum proinflammatory cytokines and liver enzyme levels. Mice injected with Ad5wt lost more than 20% of their body weight and died or required euthanasia because of poor clinical condition within 4 days of virus administration. Mice treated with OV-5 lost as much as 15% of their body weight over 8-9 days, but recovered within 14 days. Mice that were treated with Addl312 or OV-5T35H exhibited no body weight loss during the study period. These studies suggest that the Ad5/35-based chimeric viruses may have a better safety profile after intravenous injection compared with Ad5-based viruses.


Methods in molecular medicine | 2007

Generation of Recombinant Adenovirus Using the Escherichia coli BJ5183 Recombination System

P. Seshidhar Reddy; Shanthi Ganesh; Lynda K. Hawkins; Neeraja Idamakanti

One of the most time-consuming steps in the generation of adenoviral vectors is the construction of recombinant plasmids. This chapter describes a detailed method for the rapid construction of adenoviral vectors. The method described here uses homologous recombination machinery of Escherichia coli BJ5183 to construct plasmids used in generation of adenoviral vectors. With this method, no ligation steps are involved in generating the plasmids, and any region of the adenoviral genome can be easily modified. Briefly, the full-length adenoviral genome flanked by unique restriction enzyme sites is first cloned into a bacterial plasmid. Next, the region of the viral genome to be modified is subcloned into a bacterial shuttle plasmid, and the desired changes are introduced by molecular biology techniques. The modified viral DNA fragment is gel-purified and cotransformed with the full-length plasmid, linearized in the targeted region, into BJ5183 cells. Homologous recombination in E. coli generates plasmids containing the modified adenoviral genome. Recombinant virus is generated following release of the viral DNA sequences from the plasmid backbone and transfection into a producer cell line. With this method, homogeneous recombinant adenoviruses can be obtained without plaque purification.


Molecular Cancer Therapeutics | 2017

β-catenin mRNA silencing and MEK inhibition display synergistic efficacy in preclinical tumor models

Shanthi Ganesh; Xue Shui; Kevin Craig; Martin Koser; Girish Chopda; Wendy Cyr; Cheng-Jung Lai; Henryk T. Dudek; Weimin Wang; Bob D. Brown; Marc Abrams

Colorectal carcinomas harbor well-defined genetic abnormalities, including aberrant activation of Wnt/β-catenin and MAPK pathways, often simultaneously. Although the MAPK pathway can be targeted using potent small-molecule drugs, including BRAF and MEK inhibitors, β-catenin inhibition has been historically challenging. RNAi approaches have advanced to the stage of clinical viability and are especially well suited for transcriptional modulators, such as β-catenin. In this study, we report therapeutic effects of combined targeting of these pathways with pharmacologic agents. Using a recently described tumor-selective nanoparticle containing a β-catenin–targeting RNAi trigger, in combination with the FDA-approved MEK inhibitor (MEKi) trametinib, we demonstrate synergistic tumor growth inhibition in in vivo models of colorectal cancer, melanoma, and hepatocellular carcinoma. At dose levels that were insufficient to significantly impact tumor growth as monotherapies, combination regimens resulted in synergistic efficacy and complete tumor growth inhibition. Importantly, dual MEKi/RNAi therapy dramatically improved survival of mice bearing colorectal cancer liver metastases. In addition, pharmacologic silencing of β-catenin mRNA was effective against tumors that are inherently resistant or that acquire drug-induced resistance to trametinib. These results provide a strong rationale for clinical evaluation of this dual-targeting approach for cancers harboring Wnt/β-catenin and MAPK pathway mutations. Mol Cancer Ther; 17(2); 544–53. ©2017 AACR.


Molecular Cancer Research | 2015

Abstract B20: EnCore-LNP mediated tumor delivery of MYC and CTNNB1 Dicer Substrate RNAs (DsiRNAs)

Marc Abrams; Shanthi Ganesh; Bo Ying; Girish Chopda; Utsav Saxena; Anee Shah; Martin Koser; Dongyu Chen; Serena Shui; Rohan Diwanji; Wei Zhou; Benjamin Holmes; Boyoung Kim; Hailin Yang; Mihir Patel; Wendy Cyr; Natalie Pursell; Nicole Avitahl-Curtis; Hank Dudek; Cheng Lai; Weimin Wang; Bob D. Brown

MYC and CTNNB1 are well-characterized drivers of numerous tumor types. Human and preclinical genetic evidence suggest that pharmacological intervention to reduce transactivation of MYC and CTNNB1-regulated genes would yield therapeutic benefit to many cancer patients. Since the proteins encoded by these genes are challenging to target via conventional modalities, progress in new therapeutic agents has been slow despite decades of research. RNA interference technology has enabled the inhibition of previously-undruggable genetic targets at the mRNA level, and has advanced to clinical development for several indications. DCR-MYC is a Phase I-stage lipid nanoparticle (LNP)-formulated Dicer substrate siRNA (DsiRNA), representing a potent class of RNAi triggers being developed by Dicerna Pharmaceuticals. Here we describe new preclinical data that increase our understanding of the parameters that impact tumor delivery and activity of DsiRNA. We demonstrate that the cationic lipid and PEG-lipid components of Dicerna9s unique EnCore LNP platform can be modulated to improve delivery of DsiRNA to both orthotopic and spontaneous liver tumors, as well as xenograft tumors of diverse non-hepatic tissue origin. Characterization of LNP formulations with respect to plasma PK, tissue exposure and target mRNA knockdown was employed towards understanding the pharmacology of LNP-mediated tumor delivery. Citation Format: Marc Abrams, Shanthi Ganesh, Bo Ying, Girish Chopda, Utsav Saxena, Anee Shah, Martin Koser, Rokhand Arvan, Dongyu Chen, Serena Shui, Rohan Diwanji, Wei Zhou, Benjamin Holmes, Boyoung Kim, Hailin Yang, Mihir Patel, Wendy Cyr, Wendy Cyr, Natalie Pursell, Nicole Avitahl-Curtis, Hank Dudek, Cheng Lai, Weimin Wang, Bob D. Brown. EnCore-LNP mediated tumor delivery of MYC and CTNNB1 Dicer Substrate RNAs (DsiRNAs). [abstract]. In: Proceedings of the AACR Special Conference on Myc: From Biology to Therapy; Jan 7-10, 2015; La Jolla, CA. Philadelphia (PA): AACR; Mol Cancer Res 2015;13(10 Suppl):Abstract nr B20.


Molecular Therapy | 2018

RNAi-Mediated β-Catenin Inhibition Promotes T Cell Infiltration and Antitumor Activity in Combination with Immune Checkpoint Blockade

Shanthi Ganesh; Xue Shui; Kevin Craig; Jihye Park; Weimin Wang; Bob D. Brown; Marc Abrams

Wnt/β-catenin signaling mediates cancer immune evasion and resistance to immune checkpoint therapy, in part by blocking cytokines that trigger immune cell recruitment. Inhibition of β-catenin may be an effective strategy for increasing the low response rate to these effective medicines in numerous cancer populations. DCR-BCAT is a nanoparticle drug product containing a chemically optimized RNAi trigger targeting CTNNB1, the gene that encodes β-catenin. In syngeneic mouse tumor models, β-catenin inhibition with DCR-BCAT significantly increased T cell infiltration and potentiated the sensitivity of the tumors to checkpoint inhibition. The combination of DCR-BCAT and immunotherapy yielded significantly greater tumor growth inhibition (TGI) compared to monotherapy in B16F10 melanoma, 4T1 mammary carcinoma, Neuro2A neuroblastoma, and Renca renal adenocarcinoma. Response to the RNAi-containing combination therapy was not dependent on Wnt activation status of the tumor. Importantly, this drug combination was associated with elevated levels of biomarkers of T cell-mediated cytotoxicity. Finally, when CTLA-4 and PD-1 antibodies were combined with DCR-BCAT in MMTV-Wnt1 transgenic mice, a genetic model of spontaneous Wnt-driven tumors, complete regressions were achieved in the majority of treated subjects. These data support RNAi-mediated β-catenin inhibition as an effective strategy to increase response rates to cancer immunotherapy.


Cancer Research | 2018

Abstract 2759: Rational combinations for immune checkpoint blockade using β-catenin RNAi therapy

Shanthi Ganesh; Serena Shui; Kevin Craig; Weimin Wang; Bob D. Brown; Marc Abrams

Recent research has shown that Wnt/β-catenin signaling drives resistance to cancer immunotherapy by promoting the exclusion of T-cells from the tumor microenvironment. DCR-BCAT is an RNAi-based experimental medicine targeting β-catenin, formulated in a tumor-selective nanoparticle. We had previously reported that systemic administration of DCR-BCAT increased tumor-associated cytotoxic T-cells and dramatically improved responses to immunotherapy agents, in murine syngeneic models and GEM models. In this new work, we explore the mechanisms by which Wnt/β-catenin signaling promotes resistance to immune checkpoint blockade, and propose context-specific therapeutic combinations using RNAi therapy. We will present novel in vivo experimentation demonstrating that DCR-BCAT significantly enhances sensitivity to immunotherapy using different drug regimens in multiple tumor types. Citation Format: Shanthi Ganesh, Serena Shui, Kevin Craig, Weimin Wang, Bob D. Brown, Marc Abrams. Rational combinations for immune checkpoint blockade using β-catenin RNAi therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2759.

Collaboration


Dive into the Shanthi Ganesh's collaboration.

Top Co-Authors

Avatar

Marc Abrams

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Bob D. Brown

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin Craig

Northeastern University

View shared research outputs
Top Co-Authors

Avatar

Dongyu Chen

Northeastern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junyan Tao

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge