Network


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

Hotspot


Dive into the research topics where Bridget A. Quinn is active.

Publication


Featured researches published by Bridget A. Quinn.


Expert Opinion on Investigational Drugs | 2011

Targeting Mcl-1 for the therapy of cancer

Bridget A. Quinn; Rupesh Dash; Belal Azab; Siddik Sarkar; Swadesh K. Das; Sachin Kumar; Regina A. Oyesanya; Santanu Dasgupta; Paul Dent; Steven Grant; Mohamed Rahmani; David T. Curiel; Igor Dmitriev; Michael Hedvat; Jun Wei; Bainan Wu; John L. Stebbins; John C. Reed; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

Introduction: Human cancers are genetically and epigenetically heterogeneous and have the capacity to commandeer a variety of cellular processes to aid in their survival, growth and resistance to therapy. One strategy is to overexpress proteins that suppress apoptosis, such as the Bcl-2 family protein Mcl-1. The Mcl-1 protein plays a pivotal role in protecting cells from apoptosis and is overexpressed in a variety of human cancers. Areas covered: Targeting Mcl-1 for extinction in these cancers, using genetic and pharmacological approaches, represents a potentially effectual means of developing new efficacious cancer therapeutics. Here we review the multiple strategies that have been employed in targeting this fundamental protein, as well as the significant potential these targeting agents provide in not only suppressing cancer growth, but also in reversing resistance to conventional cancer treatments. Expert opinion: We discuss the potential issues that arise in targeting Mcl-1 and other Bcl-2 anti-apoptotic proteins, as well problems with acquired resistance. The application of combinatorial approaches that involve inhibiting Mcl-1 and manipulation of additional signaling pathways to enhance therapeutic outcomes is also highlighted. The ability to specifically inhibit key genetic/epigenetic elements and biochemical pathways that maintain the tumor state represent a viable approach for developing rationally based, effective cancer therapies.


Expert Opinion on Therapeutic Targets | 2013

Targeting the Bcl-2 Family for Cancer Therapy

Shibu Thomas; Bridget A. Quinn; Swadesh K. Das; Rupesh Dash; Luni Emdad; Santanu Dasgupta; Xiang-Yang Wang; Paul Dent; John C. Reed; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

Introduction: Programmed cell death is well-orchestrated process regulated by multiple pro-apoptotic and anti-apoptotic genes, particularly those of the Bcl-2 gene family. These genes are well documented in cancer with aberrant expression being strongly associated with resistance to chemotherapy and radiation. Areas covered: This review focuses on the resistance induced by the Bcl-2 family of anti-apoptotic proteins and current therapeutic interventions currently in preclinical or clinical trials that target this pathway. Major resistance mechanisms that are regulated by Bcl-2 family proteins and potential strategies to circumvent resistance are also examined. Although antisense and gene therapy strategies are used to nullify Bcl-2 family proteins, recent approaches use small molecule inhibitors (SMIs) and peptides. Structural similarity of the Bcl-2 family of proteins greatly favors development of inhibitors that target the BH3 domain, called BH3 mimetics. Expert opinion: Strategies to specifically identify and inhibit critical determinants that promote therapy resistance and tumor progression represent viable approaches for developing effective cancer therapies. From a clinical perspective, pretreatment with novel, potent Bcl-2 inhibitors either alone or in combination with conventional therapies hold significant promise for providing beneficial clinical outcomes. Identifying SMIs with broader and higher affinities for inhibiting all of the Bcl-2 pro-survival proteins will facilitate development of superior cancer therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Apogossypol derivative BI-97C1 (Sabutoclax) targeting Mcl-1 sensitizes prostate cancer cells to mda-7/IL-24–mediated toxicity

Rupesh Dash; Belal Azab; Bridget A. Quinn; Xue-Ning Shen; Xiang-Yang Wang; Swadesh K. Das; Mohamed Rahmani; Jun Wei; Michael Hedvat; Paul Dent; Igor Dmitriev; David T. Curiel; Steven Grant; Bainan Wu; John L. Stebbins; Maurizio Pellecchia; John C. Reed; Devanand Sarkar; Paul B. Fisher

Limited options are available for treating patients with advanced prostate cancer (PC). Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), an IL-10 family cytokine, exhibits pleiotropic anticancer activities without adversely affecting normal cells. We previously demonstrated that suppression of the prosurvival Bcl-2 family member, myeloid cell leukemia-1 (Mcl-1), is required for mda-7/IL-24–mediated apoptosis of prostate carcinomas. Here we demonstrate that pharmacological inhibition of Mcl-1 expression with the unique Apogossypol derivative BI-97C1, also called Sabutoclax, is sufficient to sensitize prostate tumors to mda-7/IL-24–induced apoptosis, whereas ABT-737, which lacks efficacy in inhibiting Mcl-1, does not sensitize mda-7/IL-24–mediated cytotoxicity. A combination regimen of tropism-modified adenovirus delivered mda-7/IL-24 (Ad.5/3-mda-7) and BI-97C1 enhances cytotoxicity in human PC cells, including those resistant to mda-7/IL-24 or BI-97C1 alone. The combination regimen causes autophagy that facilitates NOXA- and Bim-induced and Bak/Bax-mediated mitochondrial apoptosis. Treatment with Ad.5/3-mda-7 and BI-97C1 significantly inhibits the growth of human PC xenografts in nude mice and spontaneously induced PC in Hi-myc transgenic mice. Tumor growth inhibition correlated with increased TUNEL staining and decreased Ki-67 expression in both PC xenografts and prostates of Hi-myc mice. These findings demonstrate that pharmacological inhibition of Mcl-1 with the Apogossypol derivative, BI-97C1, sensitizes human PCs to mda-7/IL-24–mediated cytotoxicity, thus potentially augmenting the therapeutic benefit of this combinatorial approach toward PC.


Cytokine & Growth Factor Reviews | 2010

mda-7/IL-24: A unique member of the IL-10 gene family promoting cancer-targeted toxicity

Rupesh Dash; Sujit K. Bhutia; Belal Azab; Zhao-zhong Su; Bridget A. Quinn; Timothy P. Kegelmen; Swadesh K. Das; Keetae Kim; Seok-Geun Lee; Margaret A. Park; Adly Yacoub; Mohammed Rahmani; Luni Emdad; Igor Dmitriev; Xiang-Yang Wang; Devanand Sarkar; Steven Grant; Paul Dent; David T. Curiel; Paul B. Fisher

Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) is a unique member of the IL-10 gene family that displays nearly ubiquitous cancer-specific toxicity, with no harmful effects toward normal cells or tissues. mda-7/IL-24 was cloned from human melanoma cells by differentiation induction subtraction hybridization (DISH) and promotes endoplasmic reticulum (ER) stress culminating in apoptosis or toxic autophagy in a broad-spectrum of human cancers, when assayed in cell culture, in vivo in human tumor xenograft mouse models and in a Phase I clinical trial in patients with advanced cancers. This therapeutically active cytokine also induces indirect antitumor activity through inhibition of angiogenesis, stimulation of an antitumor immune response, and sensitization of cancer cells to radiation-, chemotherapy- and antibody-induced killing.


Journal of Cellular Physiology | 2012

Enhanced delivery of mda‐7/IL‐24 using a serotype chimeric adenovirus (Ad.5/3) in combination with the apogossypol derivative BI‐97C1 (Sabutoclax) improves therapeutic efficacy in low CAR colorectal cancer cells

Belal Azab; Rupesh Dash; Swadesh K. Das; Sujit K. Bhutia; Xue-Ning Shen; Bridget A. Quinn; Siddik Sarkar; Xiang-Yang Wang; Michael Hedvat; Igor Dmitriev; David T. Curiel; Steven Grant; Paul Dent; John C. Reed; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

Adenovirus (Ad)‐based gene therapy represents a potentially viable strategy for treating colorectal cancer. The infectivity of serotype 5 adenovirus (Ad.5), routinely used as a transgene delivery vector, is dependent on Coxsackie‐adenovirus receptors (CAR). CAR expression is downregulated in many cancers thus preventing optimum therapeutic efficiency of Ad.5‐based therapies. To overcome the low CAR problem, a serotype chimerism approach was used to generate a recombinant Ad (Ad.5/3) that is capable of infecting cancer cells via Ad.3 receptors in a CAR‐independent manner. We evaluated the improved transgene delivery and efficacy of Ad.5/3 recombinant virus expressing melanoma differentiation associated gene‐7/interleukin‐24 (mda‐7/IL‐24), an effective wide‐spectrum cancer‐selective therapeutic. In low CAR human colorectal cancer cells RKO, wild‐type Ad.5 virus expressing mda‐7/IL‐24 (Ad.5‐mda‐7) failed to infect efficiently resulting in lack of expression of MDA‐7/IL‐24 or induction of apoptosis. However, a recombinant Ad.5/3 virus expressing mda‐7/IL‐24 (Ad.5/3‐mda‐7) efficiently infected RKO cells resulting in higher MDA‐7/IL‐24 expression and inhibition of cell growth both in vitro and in nude mice xenograft models. Addition of the novel Bcl‐2 family pharmacological inhibitor Apogossypol derivative BI‐97C1 (Sabutoclax) significantly augmented the efficacy of Ad.5/3‐mda‐7. A combination regimen of suboptimal doses of Ad.5/3‐mda‐7 and BI‐97C1 profoundly enhanced cytotoxicity in RKO cells both in vitro and in vivo. Considering the fact that Ad.5‐mda‐7 has demonstrated significant objective responses in a Phase I clinical trial for advanced solid tumors, Ad.5/3‐mda‐7 alone or in combination with BI‐97C1 would be predicted to exert significantly improved therapeutic efficacy in colorectal cancer patients. J. Cell. Physiol. 227: 2145–2153, 2012.


Frontiers in Bioscience | 2012

MDA-9/syntenin: a positive gatekeeper of melanoma metastasis.

Swadesh K. Das; Sujit K. Bhutia; Timothy P. Kegelman; Peachy L; Regina A. Oyesanya; Santanu Dasgupta; Upneet K. Sokhi; Belal Azab; Rupesh Dash; Bridget A. Quinn; Keetae Kim; Barral Pm; Zhao-zhong Su; Habib Boukerche; Devanand Sarkar; Paul B. Fisher

Melanoma differentiation associated gene-9 (MDA-9), synonymous with syntenin, is an adapter protein that provides a central role in regulating cell-cell and cell-matrix adhesion. MDA-9/syntenin transduces signals from the cell-surface to the interior through its interaction with a plethora of additional proteins and actively participates in intracellular trafficking and cell-surface targeting, synaptic transmission, and axonal outgrowth. Recent studies demarcate a seminal role of MDA-9/syntenin in cancer metastasis. In the context of melanoma, MDA-9/syntenin functions as a positive regulator of melanoma progression and metastasis through interactions with c-Src and promotes the formation of an active FAK/c-Src signaling complex leading to NF-k B and matrix metalloproteinase (MMP) activation. The present review provides a current perspective of our understanding of the important features of MDA-9/syntenin and its significant role in tumor cell metastasis with special focus on molecular mechanism of action.


Cancer Biology & Therapy | 2007

Magnetic resonance imaging for detection and determination of tumor volume in a genetically engineered mouse model of ovarian cancer

Harvey Hensley; Bridget A. Quinn; Ronald L. Wolf; Samuel Litwin; Seiji Mabuchi; Stephen J. Williams; Christine Williams; Thomas C. Hamilton; Denise C. Connolly

Our laboratory developed a transgenic mouse model of spontaneous epithelial ovarian cancer (EOC) in which tumors are initiated by expression of the early region of the Simian Virus 40 (SV40) under transcriptional control of the 5’ upstream regulatory region of the Müllerian inhibiting substance type II receptor (MISIIR) gene. Female TgMISIIR-TAg-DR26 transgenic mice develop bilateral ovarian tumors with variable latency and survive an average of 152 days. In the absence of reliable methods for disease detection and evaluation of therapeutic response, preclinical studies of this transgenic mouse model of EOC would be limited to longitudinal experiments involving large numbers of animals with euthanasia as the endpoint. Therefore, a non-invasive method for detecting tumors, measuring tumor volume and calculating parameters relevant to the evaluation of therapeutic or preventive interventions (i.e., tumor growth rates, tumor initiation, tumor regression and the time for tumors to reach a given size) is required. We developed and optimized a non-invasive Magnetic Resonance Imaging (MRI) scanning protocol to obtain high resolution abdominal images that is well tolerated by mice. Superior contrast and contrast to noise ratio (CNR) was found with Gd-DTPA contrast enhanced T1-weighted sequences. Image sets in both the axial and coronal orientations for redundant measurements of normal ovary and ovarian tumor volume can be acquired in approximately 20 minutes. Accuracy of MRI-based ovary and tumor volume determinations was verified by standard volume measurements at necropsy. Serial imaging studies were performed on 41 ovarian cancer bearing TgMISIIR-TAg-DR26 transgenic mice to quantitate tumor progression over time in this model. A chemotherapy study was conducted on TgMISIIR-TAg-DR26 transgenic mice using a standard combination therapy consisting of cisplatin and paclitaxel. Our results demonstrate that MRI is well tolerated and can be repeated in serial imaging studies, permitting quantitative analysis of tumor growth and progression and response to therapeutic interventions.


PLOS ONE | 2009

Induction of ovarian leiomyosarcomas in mice by conditional inactivation of Brca1 and p53.

Bridget A. Quinn; Tiffany Brake; Xiang Hua; Kimberly Baxter-Jones; Samuel Litwin; Lora Hedrick Ellenson; Denise C. Connolly

Background Approximately one out of every ten cases of epithelial ovarian cancer (EOC) is inherited. The majority of inherited cases of EOC result from mutations in the breast cancer associated gene 1 (BRCA1). In addition to mutation of BRCA1, mutation of the p53 gene is often found in patients with inherited breast and ovarian cancer syndrome. Methodology/Principal Findings We investigated the role of loss of function of BRCA1 and p53 in ovarian cancer development using mouse models with conditionally expressed alleles of Brca1 and/or p53. Our results show that ovary-specific Cre-recombinase-mediated conditional inactivation of both Brca1LoxP/LoxP and p53LoxP/LoxP resulted in ovarian or reproductive tract tumor formation in 54% of mice, whereas conditional inactivation of either allele alone infrequently resulted in tumors (≤5% of mice). In mice with conditionally inactivated Brca1LoxP/LoxP and p53LoxP/LoxP, ovarian tumors arose after long latency with the majority exhibiting histological features consistent with high grade leiomyosarcomas lacking expression of epithelial, follicular or lymphocyte markers. In addition, tumors with conditional inactivation of both Brca1LoxP/LoxP and p53LoxP/LoxP exhibited greater genomic instability compared to an ovarian tumor with inactivation of only p53LoxP/LoxP. Conclusions/Significance Although conditional inactivation of both Brca1 and p53 results in ovarian tumorigenesis, our results suggest that additional genetic alterations or alternative methods for targeting epithelial cells of the ovary or fallopian tube for conditional inactivation of Brca1 and p53 are required for the development of a mouse model of Brca1-associated inherited EOC.


Journal of Ovarian Research | 2010

Development of a syngeneic mouse model of epithelial ovarian cancer

Bridget A. Quinn; Fang Xiao; Laura E. Bickel; Lainie P. Martin; Xiang Hua; Andres J. Klein-Szanto; Denise C. Connolly

BackgroundMost cases of ovarian cancer are epithelial in origin and diagnosed at advanced stage when the cancer is widely disseminated in the peritoneal cavity. The objective of this study was to establish an immunocompetent syngeneic mouse model of disseminated epithelial ovarian cancer (EOC) to facilitate laboratory-based studies of ovarian tumor biology and preclinical therapeutic strategies.MethodsIndividual lines of TgMISIIR-TAg transgenic mice were phenotypically characterized and backcrossed to inbred C57BL/6 mice. In addition to a previously described line of EOC-prone mice, two lines (TgMISIIR-TAg-Low) were isolated that express the oncogenic transgene, but have little or no susceptibility to tumor development. Independent murine ovarian carcinoma (MOVCAR) cell lines were established from the ascites of tumor-bearing C57BL/6 TgMISIIR-TAg transgenic mice, characterized and tested for engraftment in the following recipient mice: 1) severe immunocompromised immunodeficient (SCID), 2) wild type C57BL/6, 3) oophorectomized tumor-prone C57BL/6 TgMISIIR-TAg transgenic and 4) non-tumor prone C57BL/6 TgMISIIR-TAg-Low transgenic. Lastly, MOVCAR cells transduced with a luciferase reporter were implanted in TgMISIIR-TAg-Low mice and in vivo tumor growth monitored by non-invasive optical imaging.ResultsEngraftment of MOVCAR cells by i.p. injection resulted in the development of disseminated peritoneal carcinomatosis in SCID, but not wild type C57BL/6 mice. Oophorectomized tumor-prone TgMISIIR-TAg mice developed peritoneal carcinomas with high frequency, rendering them unsuitable as allograft recipients. Orthotopic or pseudo-orthotopic implantation of MOVCAR cells in TgMISIIR-TAg-Low mice resulted in the development of disseminated peritoneal tumors, frequently accompanied by the production of malignant ascites. Tumors arising in the engrafted mice bore histopathological resemblance to human high-grade serous EOC and exhibited a similar pattern of peritoneal disease spread.ConclusionsA syngeneic mouse model of human EOC was created by pseudo-orthotopic and orthotopic implantation of MOVCAR cells in a susceptible inbred transgenic host. This immunocompetent syngeneic mouse model presents a flexible system that can be used to study the consequences of altered gene expression (e.g., by ectopic expression or RNA interference strategies) in an established MOVCAR tumor cell line within the ovarian tumor microenvironment and for the development and analysis of preclinical therapeutic agents including EOC vaccines and immunotherapeutic agents.


Cancer Research | 2014

Pancreatic cancer-specific cell death induced in vivo by cytoplasmic-delivered polyinosine-polycytidylic acid.

Praveen Bhoopathi; Bridget A. Quinn; Qin Gui; Xue-Ning Shen; Steven R. Grossman; Swadesh K. Das; Devanand Sarkar; Paul B. Fisher; Luni Emdad

Polyinosine-polycytidylic acid [pIC] is a synthetic dsRNA that acts as an immune agonist of TLR3 and RLR to activate dendritic and natural killer cells that can kill tumor cells. pIC can also trigger apoptosis in pancreatic ductal adenocarcinoma cells (PDAC) but its mechanism of action is obscure. In this study, we investigated the potential therapeutic activity of a formulation of pIC with polyethylenimine ([pIC](PEI)) in PDAC and investigated its mechanism of action. [pIC](PEI) stimulated apoptosis in PDAC cells without affecting normal pancreatic epithelial cells. Mechanistically, [pIC](PEI) repressed XIAP and survivin expression and activated an immune response by inducing MDA-5, RIG-I, and NOXA. Phosphorylation of AKT was inhibited by [pIC](PEI) in PDAC, and this event was critical for stimulating apoptosis through XIAP and survivin degradation. In vivo administration of [pIC](PEI) inhibited tumor growth via AKT-mediated XIAP degradation in both subcutaneous and quasi-orthotopic models of PDAC. Taken together, these results offer a preclinical proof-of-concept for the evaluation of [pIC](PEI) as an immunochemotherapy to treat pancreatic cancer.

Collaboration


Dive into the Bridget A. Quinn's collaboration.

Top Co-Authors

Avatar

Paul B. Fisher

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Devanand Sarkar

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Swadesh K. Das

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Luni Emdad

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Rupesh Dash

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Paul Dent

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Siddik Sarkar

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Xue-Ning Shen

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Xiang-Yang Wang

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Belal Azab

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge