Network


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

Hotspot


Dive into the research topics where Marc Abrams is active.

Publication


Featured researches published by Marc Abrams.


Journal of Biological Chemistry | 2004

Inhibition of Glucocorticoid-induced Apoptosis by Targeting the Major Splice Variants of BIM mRNA with Small Interfering RNA and Short Hairpin RNA

Marc Abrams; Noreen M. Robertson; Kyonggeun Yoon; Eric Wickstrom

Glucocorticoids (GCs) induce apoptosis in lymphocytes and are effective agents for the treatment of leukemia. The activated glucocorticoid receptor initiates a transcriptional program leading to caspase activation and cell death, but the critical signaling intermediates in GC-induced apoptosis remain largely undefined. We have observed that GC induction of the three major protein products of the Bcl-2 relative Bim (BimEL, BimS, and BimL) correlates with GC sensitivity in a panel of human precursor B-cell (pre-B) acute lymphoblastic leukemia (ALL) cell lines. To test the hypothesis that Bim facilitates GC-induced apoptosis, we reduced BIM mRNA levels and Bim protein levels by RNA interference in highly GC-sensitive pre-B ALL cells. Reducing Bim proteins by either electroporation of synthetic small interfering RNA (siRNA) duplexes or lentivirus-mediated stable expression of short hairpin RNA inhibited the activation of caspase-3 and increased cell viability following GC exposure. We also observed that the extent of GC resistance correlated with siRNA silencing potency. siRNA duplexes that reduced only BimEL or BimEL and BimL (but not BimS) exhibited less GC resistance than a potent siRNA that silenced all three major isoforms, implying that induction of all three Bim proteins contributes to cell death. Finally, the modulation of GC-induced apoptosis caused by Bim silencing was independent of Bcl-2 expression levels, negating the hypothesis that the ratio of Bim to Bcl-2 regulates apoptosis. These results offer evidence that the induction of Bim by GC is a required event for the complete apoptotic response in pre-B ALL cells.


Journal of Medicinal Chemistry | 2008

Kinesin spindle protein (KSP) inhibitors. 9. Discovery of (2S)-4-(2,5-difluorophenyl)-n-[(3R,4S)-3-fluoro-1-methylpiperidin-4-yl]-2-(hydroxymethyl)-N-methyl-2-phenyl-2,5-dihydro-1H-pyrrole-1-carboxamide (MK-0731) for the treatment of taxane-refractory cancer.

Christopher D. Cox; Paul J. Coleman; Michael J. Breslin; David B. Whitman; Robert M. Garbaccio; Mark E. Fraley; Carolyn A. Buser; Eileen S. Walsh; Kelly Hamilton; Michael D. Schaber; Robert B. Lobell; Weikang Tao; Joseph P. Davide; Ronald E. Diehl; Marc Abrams; Vicki J. South; Hans E. Huber; Maricel Torrent; Thomayant Prueksaritanont; Chunze Li; Donald E. Slaughter; Elizabeth Mahan; Carmen Fernandez-Metzler; Youwei Yan; Lawrence C. Kuo; Nancy E. Kohl; George D. Hartman

Inhibition of kinesin spindle protein (KSP) is a novel mechanism for treatment of cancer with the potential to overcome limitations associated with currently employed cytotoxic agents. Herein, we describe a C2-hydroxymethyl dihydropyrrole KSP inhibitor ( 11) that circumvents hERG channel binding and poor in vivo potency, issues that limited earlier compounds from our program. However, introduction of the C2-hydroxymethyl group caused 11 to be a substrate for cellular efflux by P-glycoprotein (Pgp). Utilizing knowledge garnered from previous KSP inhibitors, we found that beta-fluorination modulated the p K a of the piperidine nitrogen and reduced Pgp efflux, but the resulting compound ( 14) generated a toxic metabolite in vivo. Incorporation of fluorine in a strategic, metabolically benign position by synthesis of an N-methyl-3-fluoro-4-(aminomethyl)piperidine urea led to compound 30 that has an optimal in vitro and metabolic profile. Compound 30 (MK-0731) was recently studied in a phase I clinical trial in patients with taxane-refractory solid tumors.


Journal of Cellular Biochemistry | 2006

RNAi-mediated silencing of insulin receptor substrate 1 (IRS-1) enhances tamoxifen-induced cell death in MCF-7 breast cancer cells

Gregory Cesarone; Cecilia Garofalo; Marc Abrams; Olga Igoucheva; Vitali Alexeev; Kyonggeun Yoon; Eva Surmacz; Eric Wickstrom

Insulin receptor substrate 1 (IRS‐1) is a major downstream signaling protein for insulin and insulin‐like growth factor I (IGF‐I) receptors, conveying signals to PI‐3K/Akt and ERK1/2 pathways. In breast cancer, IRS‐1 overexpression has been associated with tumor development, hormone‐independence and antiestrogen‐resistance. In part, these effects are related to potentiation of IRS‐1/PI‐3K/Akt signaling. In estrogen sensitive breast cancer cell lines, tamoxifen treatment reduces IRS‐1 expression and function; consequently, inhibiting IRS‐1/PI‐3K signaling. We tested whether anti‐IRS1 siRNA could inhibit growth and survival of estrogen‐sensitive MCF‐7 breast cancer cells, when used alone or in combination with TAM. Our results indicated: (a) out of four tested anti‐IRS1 siRNAs, two siRNAs reduced IRS‐1 protein by approximately three‐fold in both growing and IGF‐I‐stimulated cells without affecting a closely related protein, IRS‐2; (b) these effects paralleled IRS1 mRNA downregulation by approximately three‐fold, measured by quantitative real time‐polymerase chain reaction; (c) action of anti‐IRS1 siRNAs induced the apoptotic response, observed by altered mitochondrial membrane potential coupled with downregulation of NF‐κB target Bcl‐xL and reduced cell viability; (d) anti‐IRS1 siRNA treatment enhanced the cytotoxic effects of TAM by ∼20%. In summary, anti‐IRS1 RNAi strategy could become a potent tool to induce breast cancer cell death, especially if combined with standard TAM therapy. J. Cell. Biochem. 98: 440–450, 2006.


Molecular Therapy | 2016

Inhibition of Glycolate Oxidase With Dicer-substrate siRNA Reduces Calcium Oxalate Deposition in a Mouse Model of Primary Hyperoxaluria Type 1

Chaitali Dutta; Nicole Avitahl-Curtis; Natalie Pursell; Marita Larsson Cohen; Benjamin Holmes; Rohan Diwanji; Wei Zhou; Luciano Apponi; Martin Koser; Bo Ying; Dongyu Chen; Xue Shui; Utsav Saxena; Wendy Cyr; Anee Shah; Naim Nazef; Weimin Wang; Marc Abrams; Henryk T. Dudek; Eduardo Salido; Bob D. Brown; Cheng-Jung Lai

Primary hyperoxaluria type 1 (PH1) is an autosomal recessive, metabolic disorder caused by mutations of alanine-glyoxylate aminotransferase (AGT), a key hepatic enzyme in the detoxification of glyoxylate arising from multiple normal metabolic pathways to glycine. Accumulation of glyoxylate, a precursor of oxalate, leads to the overproduction of oxalate in the liver, which accumulates to high levels in kidneys and urine. Crystalization of calcium oxalate (CaOx) in the kidney ultimately results in renal failure. Currently, the only treatment effective in reduction of oxalate production in patients who do not respond to high-dose vitamin B6 therapy is a combined liver/kidney transplant. We explored an alternative approach to prevent glyoxylate production using Dicer-substrate small interfering RNAs (DsiRNAs) targeting hydroxyacid oxidase 1 (HAO1) mRNA which encodes glycolate oxidase (GO), to reduce the hepatic conversion of glycolate to glyoxylate. This approach efficiently reduces GO mRNA and protein in the livers of mice and nonhuman primates. Reduction of hepatic GO leads to normalization of urine oxalate levels and reduces CaOx deposition in a preclinical mouse model of PH1. Our results support the use of DsiRNA to reduce liver GO levels as a potential therapeutic approach to treat PH1.


Cancer Biology & Therapy | 2006

EGFR-dependent downregulation of bim in epithelial cells requires MAPK and PKC-δ activities

Marlene R.D. Quadros; Sharon Connelly; Csaba Kari; Marc Abrams; Eric Wickstrom; Ulrich Rodeck

Activation of the epidermal growth factor receptor (EGFR) provides a measure of protection to immortalized epidermal keratinocytes (HaCaT cells) against apoptosis induced by diverse cellular stressors. This effect is due, in part, to sustained MAPK-dependent Bcl-xL expression. Here, we report a second EGFR/MAPK-dependent signaling event that protects HaCaT cells against apoptosis incurred during forced suspension culture (anoikis). This pathway targets Bim, a pro-apoptotic BH3-only Bcl-2 family member. Bim expression was functionally relevant to HaCaT cell survival as demonstrated by partial protection against anoikis provided by siRNA-induced Bim downregulation. Growth factor starvation of attached and suspended cells was associated with enhanced Bim expression whereas EGFR activation reduced Bim expression by inducing Bim phosphorylation and proteasomal degradation. EGFR-dependent Bim phosphorylation required MAPK activation. Furthermore, PKC-δ activity contributed to both MEK/MAPK phosphorylation and Bim phosphorylation as demonstrated using both pharmacological inhibitors of PKC-δ and siRNA-mediated PKC-δ knockdown. In addition to HaCaT cells, EGFR activation supported survival and induced Bim phosphorylation in several squamous carcinoma cell lines in a strictly MAPK-dependent fashion. These results establish that EGFR activation attenuates susceptibility of immortalized and malignant keratinocytes to apoptosis by posttranslational control of Bim-EL expression through a pathway requiring PKC-δ and MEK/MAPK activation.


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 showed > 90% 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.


Journal of Cancer Research and Clinical Oncology | 2005

Evaluation of glucocorticoid sensitivity in 697 pre-B acute lymphoblastic leukemia cells after overexpression or silencing of MAP kinase phosphatase-1

Marc Abrams; Noreen M. Robertson; Gerald Litwack; Eric Wickstrom

Purpose To determine the effect of modulating MAP kinase phosphatase-1 (MKP-1) expression levels on cell death induced by glucocorticoid (GC) or hydroxyurea (HU) treatment in the human pre-B acute lymphoblastic leukemia cell line 697.Methods Stable MKP-1 overexpressing transformants of the 697 pre-B acute lymphoblastic leukemia cell line were created and tested for sensitivity to the GC triamcinolone acetonide (TA) and HU, and compared to a control 697 cell line containing normal MKP-1 expression levels. Small interfering RNAs (siRNAs) were designed to inhibit MKP-1 expression and evaluated for their effect on GC-mediated cell death.Results MKP-1 overexpression caused a phenotype of partial resistance to HU-induced apoptosis but not to GC-induced apoptosis. Electroporation of siRNAs effectively silenced MKP-1 expression, and increased sensitivity to TA by 9.6±1.9%.Conclusions Because MKP-1 protects certain tumor cells from chemotherapy-induced apoptosis, its inhibition is being considered as a possible strategy for combination cancer therapy. However, this study suggests that while MKP-1 inhibition may improve the efficacy of DNA damaging agents, it may have only limited utility in combination with glucocorticoids.


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.

Collaboration


Dive into the Marc Abrams's collaboration.

Top Co-Authors

Avatar

Bob D. Brown

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shanthi Ganesh

University of California

View shared research outputs
Top Co-Authors

Avatar

Kevin Craig

Northeastern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dongyu Chen

Northeastern University

View shared research outputs
Top Co-Authors

Avatar

Eric Wickstrom

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge