Network


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

Hotspot


Dive into the research topics where Evrim Gurpinar is active.

Publication


Featured researches published by Evrim Gurpinar.


Clinical Cancer Research | 2014

NSAIDs Inhibit Tumorigenesis, but How?

Evrim Gurpinar; William E. Grizzle; Gary A. Piazza

Numerous epidemiologic studies have reported that the long-term use of nonsteroidal anti-inflammatory drugs (NSAID) is associated with a significant decrease in cancer incidence and delayed progression of malignant disease. The use of NSAIDs has also been linked with reduced risk from cancer-related mortality and distant metastasis. Certain prescription-strength NSAIDs, such as sulindac, have been shown to cause regression of precancerous lesions. Unfortunately, the extended use of NSAIDs for chemoprevention results in potentially fatal side effects related to their COX-inhibitory activity and suppression of prostaglandin synthesis. Although the basis for the tumor growth–inhibitory activity of NSAIDs likely involves multiple effects on tumor cells and their microenvironment, numerous investigators have concluded that the underlying mechanism is not completely explained by COX inhibition. It may therefore be possible to develop safer and more efficacious drugs by targeting such COX-independent mechanisms. NSAID derivatives or metabolites that lack COX-inhibitory activity, but retain or have improved anticancer activity, support this possibility. Experimental studies suggest that apoptosis induction and suppression of β-catenin–dependent transcription are important aspects of their antineoplastic activity. Studies show that the latter involves phosphodiesterase inhibition and the elevation of intracellular cyclic GMP levels. Here, we review the evidence for COX-independent mechanisms and discuss progress toward identifying alternative targets and developing NSAID derivatives that lack COX-inhibitory activity but have improved antineoplastic properties. Clin Cancer Res; 20(5); 1104–13. ©2013 AACR.


Molecular Cancer Therapeutics | 2013

Sulindac selectively inhibits colon tumor cell growth by activating the cGMP/PKG pathway to suppress Wnt/β-catenin signaling

Nan Li; Yaguang Xi; Heather N. Tinsley; Evrim Gurpinar; Bernard D. Gary; Bing Zhu; Yonghe Li; Xi Chen; Adam B. Keeton; Ashraf H. Abadi; Mary P. Moyer; William E. Grizzle; Wen-Chi L. Chang; Margie L. Clapper; Gary A. Piazza

Nonsteroidal anti-inflammatory drugs (NSAID) display promising antineoplastic activity for colorectal and other cancers, but toxicity from COX inhibition limits their long-term use for chemoprevention. Previous studies have concluded that the basis for their tumor cell growth inhibitory activity does not require COX inhibition, although the underlying mechanism is poorly understood. Here, we report that the NSAID sulindac sulfide inhibits cyclic guanosine 3′,5′-monophosphate phosphodiesterase (cGMP PDE) activity to increase intracellular cGMP levels and activate cGMP-dependent protein kinase (PKG) at concentrations that inhibit proliferation and induce apoptosis of colon tumor cells. Sulindac sulfide did not activate the cGMP/PKG pathway, nor affect proliferation or apoptosis in normal colonocytes. Knockdown of the cGMP-specific PDE5 isozyme by siRNA and PDE5-specific inhibitors tadalafil and sildenafil also selectively inhibited the growth of colon tumor cells that expressed high levels of PDE5 compared with colonocytes. The mechanism by which sulindac sulfide and the cGMP/PKG pathway inhibits colon tumor cell growth involves the transcriptional suppression of β-catenin to inhibit Wnt/β-catenin T-cell factor transcriptional activity, leading to downregulation of cyclin D1 and survivin. These observations suggest that safer and more efficacious sulindac derivatives can be developed for colorectal cancer chemoprevention by targeting PDE5 and possibly other cGMP-degrading isozymes. Mol Cancer Ther; 12(9); 1848–59. ©2013 AACR.


Frontiers in Oncology | 2013

COX-Independent Mechanisms of Cancer Chemoprevention by Anti-Inflammatory Drugs

Evrim Gurpinar; William E. Grizzle; Gary A. Piazza

Epidemiological and clinical studies suggest that non-steroidal anti-inflammatory drugs (NSAIDs), including cyclooxygenase (COX)-2 selective inhibitors, reduce the risk of developing cancer. Experimental studies in human cancer cell lines and rodent models of carcinogenesis support these observations by providing strong evidence for the antineoplastic properties of NSAIDs. The involvement of COX-2 in tumorigenesis and its overexpression in various cancer tissues suggest that inhibition of COX-2 is responsible for the chemopreventive efficacy of these agents. However, the precise mechanisms by which NSAIDs exert their antiproliferative effects are still a matter of debate. Numerous other studies have shown that NSAIDs can act through COX-independent mechanisms. This review provides a detailed description of the major COX-independent molecular targets of NSAIDs and discusses how these targets may be involved in their anticancer effects. Toxicities resulting from COX inhibition and the suppression of prostaglandin synthesis preclude the long-term use of NSAIDs for cancer chemoprevention. Furthermore, chemopreventive efficacy is incomplete and treatment often leads to the development of resistance. Identification of alternative NSAID targets and elucidation of the biochemical processes by which they inhibit tumor growth could lead to the development of safer and more efficacious drugs for cancer chemoprevention.


Oncogene | 2015

Phosphodiesterase 10A: a novel target for selective inhibition of colon tumor cell growth and β-catenin-dependent TCF transcriptional activity

Nan Li; Kevin Lee; Yaguang Xi; Bing Zhu; Bernard D. Gary; Veronica Ramirez-Alcantara; Evrim Gurpinar; Joshua C. Canzoneri; Alexandra Fajardo; Sara C. Sigler; John T. Piazza; Xi Chen; Joel Andrews; Meagan Thomas; Wenyan Lu; Yonghe Li; Danuel J. Laan; Mary P. Moyer; Suzanne Russo; Brian T. Eberhardt; Larry Yet; Adam B. Keeton; William E. Grizzle; Gary A. Piazza

The cyclic nucleotide phosphodiesterase 10A (PDE10) has been mostly studied as a therapeutic target for certain psychiatric and neurological conditions, although a potential role in tumorigenesis has not been reported. Here we show that PDE10 is elevated in human colon tumor cell lines compared with normal colonocytes, as well as in colon tumors from human clinical specimens and intestinal tumors from ApcMin/+ mice compared with normal intestinal mucosa, respectively. An isozyme and tumor-selective role of PDE10 were evident by the ability of small-molecule inhibitors and small interfering RNA knockdown to suppress colon tumor cell growth with reduced sensitivity of normal colonocytes. Stable knockdown of PDE10 by short hairpin RNA also inhibits colony formation and increases doubling time of colon tumor cells. PDE10 inhibition selectively activates cGMP/cGMP-dependent protein kinase signaling to suppress β-catenin levels and T-cell factor (TCF) transcriptional activity in colon tumor cells. Conversely, ectopic expression of PDE10 in normal and precancerous colonocytes increases proliferation and activates TCF transcriptional activity. These observations suggest a novel role of PDE10 in colon tumorigenesis and that inhibitors may be useful for the treatment or prevention of colorectal cancer.


Molecular Cancer Therapeutics | 2013

A Novel Sulindac Derivative Inhibits Lung Adenocarcinoma Cell Growth through Suppression of Akt/mTOR Signaling and Induction of Autophagy

Evrim Gurpinar; William E. Grizzle; John J. Shacka; Burton J. Mader; Nan Li; Nicholas A. Piazza; Suzanne Russo; Adam B. Keeton; Gary A. Piazza

Nonsteroidal anti-inflammatory drugs such as sulindac sulfide have shown promising antineoplastic activity in multiple tumor types, but toxicities resulting from COX inhibition limit their use in cancer therapy. We recently described a N,N-dimethylethyl amine derivative of sulindac sulfide, sulindac sulfide amide (SSA), that does not inhibit COX-1 or -2, yet displays potent tumor cell growth–inhibitory activity. Here, we studied the basis for the growth-inhibitory effects of SSA on human lung adenocarcinoma cell lines. SSA potently inhibited the growth of lung tumor cells with IC50 values of 2 to 5 μmol/L compared with 44 to 52 μmol/L for sulindac sulfide. SSA also suppressed DNA synthesis and caused a G0–G1 cell-cycle arrest. SSA-induced cell death was associated with characteristics of autophagy, but significant caspase activation or PARP cleavage was not observed after treatment at its IC50 value. siRNA knockdown of Atg7 attenuated SSA-induced autophagy and cell death, whereas pan-caspase inhibitor ZVAD was not able to rescue viability. SSA treatment also inhibited Akt/mTOR signaling and the expression of downstream proteins that are regulated by this pathway. Overexpression of a constitutively active form of Akt was able to reduce autophagy markers and confer resistance to SSA-induced cell death. Our findings provide evidence that SSA inhibits lung tumor cell growth by a mechanism involving autophagy induction through the suppression of Akt/mTOR signaling. This unique mechanism of action, along with its increased potency and lack of COX inhibition, supports the development of SSA or related analogs for the prevention and/or treatment of lung cancer. Mol Cancer Ther; 12(5); 663–74. ©2013 AACR.


Marine Drugs | 2010

Preclinical Pharmacology of BA-TPQ, a Novel Synthetic Iminoquinone Anticancer Agent

Scharri J. Ezell; Haibo Li; Hongxia Xu; Xiangrong Zhang; Evrim Gurpinar; Xu Zhang; Elizabeth R. Rayburn; Charnell I. Sommers; Xinyi Yang; Sadanandan E. Velu; Wei Wang; Ruiwen Zhang

Marine natural products and their synthetic derivatives represent a major source of novel candidate anti-cancer compounds. We have recently tested the anti-cancer activity of more than forty novel compounds based on an iminoquinone makaluvamine scaffold, and have found that many of the compounds exert potent cytotoxic activity against human cancer cell lines. One of the most potent compounds, BA-TPQ [(11,12),7-(benzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one], was active against a variety of human cancer cell lines, and inhibited the growth of breast and prostate xenograft tumors in mice. However, there was some toxicity noted in the mice following administration of the compound. In order to further the development of BA-TPQ, and in a search for potential sites of accumulation that might underlie the observed toxicity of the compound, we accomplished preclinical pharmacological studies of the compound. We herein report the in vitro and in vivo pharmacological properties of BA-TPQ, including its stability in plasma, plasma protein binding, metabolism by S9 enzymes, and plasma and tissue distribution. We believe these studies will be useful for further investigations, and may be useful for other investigators examining the use of similar compounds for cancer therapy.


Biomedical Chromatography | 2011

Development and validation of an HPLC method for quantitation of BA-TPQ, a novel iminoquinone anticancer agent, and an initial pharmacokinetic study in mice.

Haibo Li; Scharri J. Ezell; Xiangrong Zhang; Wei Wang; Hongxia Xu; Elizabeth R. Rayburn; Xu Zhang; Evrim Gurpinar; Xinyi Yang; Charnell I. Sommers; Sadanandan E. Velu; Ruiwen Zhang

We herein describe the development and validation of an HPLC method for the quantitation of 7-(benzylamino)-1,3,4,8-tetrahydropyrrolo [4,3,2-de]quinolin-8(1H)-one (BA-TPQ), a newly synthesized iminoquinone anticancer agent. BA-TPQ was extracted from plasma and tissue samples by first precipitating proteins with acetonitrile followed by a liquid-liquid extraction with ethyl acetate. Chromatographic separation was carried out using a gradient flow rate on a Zorbax SB C(18) column, and the effluent was monitored by UV detection at 346 nm. The method was found to be precise, accurate, and specific, with a linear range of 3.91-1955.0  ng/mL in plasma, 19.55-1955.0  ng/mL in spleen, brain, and liver homogenates and 19.55-3910.0  ng/mL in heart, lung and kidney homogenates. The method was stable under all relevant conditions. Using this method, we also carried out an initial study determining plasma pharmacokinetics and tissue distribution of BA-TPQ in mice following intravenous administration. In summary, this simple and sensitive HPLC method can be used in future preclinical and clinical studies of BA-TPQ.


Oncotarget | 2017

Phosphodiesterase 10A is overexpressed in lung tumor cells and inhibitors selectively suppress growth by blocking β-catenin and MAPK signaling

Bing Zhu; Ashley S. Lindsey; Nan Li; Kevin Lee; Veronica Ramirez-Alcantara; Joshua C. Canzoneri; Alexandra M. Fajardo; Luciana Madeira da Silva; Meagan Thomas; John T. Piazza; Larry Yet; Brian T. Eberhardt; Evrim Gurpinar; Dennis Otali; William E. Grizzle; Jacob Valiyaveettil; Xi Chen; Adam B. Keeton; Gary A. Piazza

Phosphodiesterase 10A (PDE10) is a cyclic nucleotide (e.g. cGMP) degrading enzyme highly expressed in the brain striatum where it plays an important role in dopaminergic neurotransmission, but has limited expression and no known physiological function outside the central nervous system. Here we report that PDE10 mRNA and protein levels are strongly elevated in human non-small cell lung cancer cells and lung tumors compared with normal human airway epithelial cells and lung tissue, respectively. Genetic silencing of PDE10 or inhibition by small molecules such as PQ10 was found to selectively inhibit the growth and colony formation of lung tumor cells. PQ10 treatment of lung tumor cells rapidly increased intracellular cGMP levels and activated cGMP-dependent protein kinase (PKG) at concentrations that inhibit lung tumor cell growth. PQ10 also increased the phosphorylation of β-catenin and reduced its levels, which paralleled the suppression of cyclin D1 and survivin but preceded the activation of PARP and caspase cleavage. PQ10 also suppressed RAS-activated RAF/MAPK signaling within the same concentration range and treatment period as required for cGMP elevation and PKG activation. These results show that PDE10 is overexpressed during lung cancer development and essential for lung tumor cell growth in which inhibitors can selectively induce apoptosis by increasing intracellular cGMP levels and activating PKG to suppress oncogenic β-catenin and MAPK signaling.


Cancer Research | 2015

Abstract 950: Antitumor activity of a novel phosphodiesterase 10 inhibitor in an orthotopic mouse model of lung cancer

Veronica Ramirez-Alcantara; Michele Schuler; Bing Zhu; Nan Li; Evrim Gurpinar; Dennis Otali; Joshua C. Canzoneri; Adam B. Keeton; Bernard D. Gary; Suzanne Russo; Lori Coward; Gregory S. Gorman; William E. Grizzle; Xi Chen; Michael R. Boyd; Gary A. Piazza

BACKGROUND: We recently reported that phosphodiesterase 10A (PDE10) is elevated in certain tumor types and essential for tumor cell growth (Li et al., Oncogene 2014). Here we evaluate anti-tumor efficacy, pharmacokinetics, tissue distribution, and tolerance of a novel PDE10 inhibitor, ADT-048, in mice. METHODS: ADT-048 was orally administrated to C57BL/6 mice and free levels quantified in plasma and tissues by LC-MS. Anti-tumor activity was evaluated in athymic nude-Foxn1nu mice after inoculating the left lung with 1×106 A549 lung tumor cells transfected with luciferase. Tumor growth was monitored by in situ bioluminescence using the In Vivo Imaging System and efficacy was evaluated by necropsy grading. RESULTS: ADT-048 inhibited A549 lung tumor cell growth with an IC50 value of 0.7 μM, while appreciably higher concentrations were required to inhibit the growth of normal airway epithelial cells (IC50 = 35 μM). Pharmacokinetic studies demonstrated a half-life suitable for once a day dosing, while tissue distribution studies demonstrated a unique capacity to achieve high concentrations in lungs relative to plasma and other tissues. ADT-048, at a dose of 100 mg/Kg, displayed significant anti-tumor activity as evident by reduced luminescence and tumor grading following 28 days of treatment. CONCLUSIONS: A novel PDE10 inhibitor, ADT-048, was found to display highly potent and selective tumor cell growth inhibitory activity in vitro. ADT-048 was well tolerated in mice and had attractive pharmaceutical properties with a unique capacity to achieve high lung concentrations following oral administration. Efficacy studies demonstrate strong anti-tumor activity of ADT-048, which represents a candidate for clinical development. Supported by NIH grants 1R01CA155638 and 1R01CA131378 (Piazza). Citation Format: Veronica Ramirez-Alcantara, Michele Schuler, Bing Zhu, Nan Li, Evrim Gurpinar, Dennis Otali, Joshua Canzoneri, Adam Keeton, Bernard Gary, Suzanne Russo, Lori Coward, Gregory Gorman, William Grizzle, Xi Chen, Michael Boyd, Gary Piazza. Antitumor activity of a novel phosphodiesterase 10 inhibitor in an orthotopic mouse model of lung cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 950. doi:10.1158/1538-7445.AM2015-950


Cancer Research | 2014

Abstract 1773: Phosphodiesterase 10A: A novel target for selective inhibition of colon tumor cell growth and Wnt/β-catenin signaling

Nan Li; Kevin Lee; Yaguang Xi; Bing Zhu; Bernary D. Gary; Veronica Ramirez-Alcantara; Evrim Gurpinar; Joshua C. Canzoneri; Alexandra M. Fajardo; Sara Sigler; John T. Piazza; Xi Chen; Joel Andrews; Wenyan Lu; Yonghe Li; Suzanne Russo; Larry Yet; Adam B. Keeton; William E. Grizzle; Gary A. Piazza

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The cyclic nucleotide phosphodiesterase10A (PDE10) has been studied as a therapeutic target for certain psychiatric and neurological conditions, although a potential role in tumorigenesis has not been reported. Here we report that PDE10 is elevated in human colon tumor cell lines compared with normal colonocytes. Similarly, PDE10 is elevated in colon tumors from human clinical specimens as well as intestinal tumors from the ApcMin/+ mouse model compared with non-involved tissue or normal intestinal mucosa, respectively. An isozyme and tumor-selective role of PDE10 was evident by the ability of specific inhibitors and siRNA knockdown to suppress colon tumor cell growth without significantly affecting the growth of normal colonocytes. Stable knockdown of PDE10 by shRNA also inhibits colony formation and increases doubling time of colon tumor cells. Inhibition of PDE10 results in the selective activation of cGMP/PKG signaling in colon tumor cells to suppress β-catenin levels and T-cell factor (TCF) transcriptional activity. Conversely, ectopic expression of PDE10 in normal and precancerous colonocytes increases proliferation and activates TCF transcriptional activity. These observations demonstrate that PDE10 is essential for colon tumor cell growth and suggest that inhibitors may have therapeutic potential for the treatment or prevention of colorectal cancer. Citation Format: Nan Li, Kevin Lee, Yaguang Xi, Bing Zhu, Bernary D. Gary, Veronica Ramirez-Alcantara, Evrim Gurpinar, Joshua C. Canzoneri, Alexandra Fajardo, Sara Sigler, John T. Piazza, Xi Chen, Joel Andrews, Wenyan Lu, Yonghe Li, Suzanne Russo, Larry Yet, Adam B. Keeton, William E. Grizzle, Gary A. Piazza. Phosphodiesterase 10A: A novel target for selective inhibition of colon tumor cell growth and Wnt/β-catenin signaling. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1773. doi:10.1158/1538-7445.AM2014-1773

Collaboration


Dive into the Evrim Gurpinar's collaboration.

Top Co-Authors

Avatar

Gary A. Piazza

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Adam B. Keeton

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

William E. Grizzle

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Nan Li

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Bernard D. Gary

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Bing Zhu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Xi Chen

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suzanne Russo

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge