Network


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

Hotspot


Dive into the research topics where Kevin J. Gaffney is active.

Publication


Featured researches published by Kevin J. Gaffney.


Molecular Cancer | 2008

COX-2 inhibition is neither necessary nor sufficient for celecoxib to suppress tumor cell proliferation and focus formation in vitro

Huan-Ching Chuang; Adel Kardosh; Kevin J. Gaffney; Nicos A. Petasis; Axel H. Schönthal

BackgroundAn increasing number of reports is challenging the notion that the antitumor potential of the selective COX-2 inhibitor celecoxib (Celebrex®) is mediated primarily via the inhibition of COX-2. We have investigated this issue by applying two different analogs of celecoxib that differentially display COX-2-inhibitory activity: the first analog, called unmethylated celecoxib (UMC), inhibits COX-2 slightly more potently than its parental compound, whereas the second analog, 2,5-dimethyl-celecoxib (DMC), has lost the ability to inhibit COX-2.ResultsWith the use of glioblastoma and pancreatic carcinoma cell lines, we comparatively analyzed the effects of celecoxib, UMC, and DMC in various short-term (≤48 hours) cellular and molecular studies, as well as in long-term (≤3 months) focus formation assays. We found that DMC exhibited the most potent antitumor activity; celecoxib was somewhat less effective, and UMC clearly displayed the overall weakest antitumor potential in all aspects. The differential growth-inhibitory and apoptosis-stimulatory potency of these compounds in short-term assays did not at all correlate with their capacity to inhibit COX-2, but was closely aligned with their ability to trigger endoplasmic reticulum stress (ERS), as indicated by the induction of the ERS marker CHOP/GADD153 and activation of the ERS-associated caspase 7. In addition, we found that these compounds were able to restore contact inhibition and block focus formation during long-term, chronic drug exposure of tumor cells, and this was achieved at sub-toxic concentrations in the absence of ERS or inhibition of COX-2.ConclusionThe antitumor activity of celecoxib in vitro did not involve the inhibition of COX-2. Rather, the drugs ability to trigger ERS, a known effector of cell death, might provide an alternative explanation for its acute cytotoxicity. In addition, the newly discovered ability of this drug to restore contact inhibition and block focus formation during chronic drug exposure, which involved neither ERS nor COX-2, suggests a novel, as yet unrecognized mechanism of celecoxib action.


Cancer Letters | 2012

Preferential killing of triple-negative breast cancer cells in vitro and in vivo when pharmacological aggravators of endoplasmic reticulum stress are combined with autophagy inhibitors

Simmy Thomas; Natasha Sharma; Encouse B. Golden; Hee-Yeon Cho; Puneet Agarwal; Kevin J. Gaffney; Nicos A. Petasis; Thomas C. Chen; Florence M. Hofman; Stan G. Louie; Axel H. Schönthal

The cellular processes of autophagy and endoplasmic reticulum stress (ERS) appear to be interconnected, and it has been proposed that autophagy may serve to reduce ERS via removal of terminally misfolded and aggregated proteins. Conversely, there are indications that blockage of autophagy may increase ERS. Based on earlier work demonstrating that pharmacologically aggravated ERS can result in tumor cell killing, we investigated whether blockage of autophagy would enhance this effect in a therapeutically useful manner. We therefore combined chloroquine (CQ), a pharmacological inhibitor of autophagy, with other drugs known to act as ERS aggravators (ERSA), namely nelfinavir (an HIV protease inhibitor) and celecoxib (a cyclooxygenase-2 inhibitor) or its non-coxib analog 2,5-dimethyl-celecoxib (DMC), and investigated combination drug effects in a variety of breast cancer cell lines. We found that the addition of CQ resulted in synergistic enhancement of tumor cell killing by ERSA compounds, particularly in triple-negative breast cancer (TNBC) cells. This combination effect could also be confirmed in an in vivo model, where CQ boosted low-dose ERSA effects, resulting in rapid deterioration of xenografted tumors in mice. Altogether, our results indicate that combinations of an autophagy inhibitor with pharmacological ERSA (i.e. compounds that lead to ER stress aggravation) should be further explored for potential therapy of otherwise difficult-to-treat TNBC.


Cancer Letters | 2009

Enhanced killing of chemo-resistant breast cancer cells via controlled aggravation of ER stress

Hee-Yeon Cho; Simmy Thomas; Encouse B. Golden; Kevin J. Gaffney; Florence M. Hofman; Thomas C. Chen; Stan G. Louie; Nicos A. Petasis; Axel H. Schönthal

Moderate activity of the endoplasmic reticulum (ER) stress response system exerts anti-apoptotic function and supports tumor cell survival and chemoresistance, whereas its more severe aggravation may exceed the protective capacity of this system and turn on its pro-apoptotic module. In this study, we investigated whether the combination of two pharmacologic agents with known ability to trigger ER stress via different mechanisms would synergize and lead to enhanced tumor cell death. We combined the HIV protease inhibitor nelfinavir (Viracept) and the cyclooxygenase 2 (COX-2) inhibitor celecoxib (Celebrex) and investigated their combined effect on ER stress and on the viability of breast cancer cells. We found that this drug combination aggravated ER stress and caused pronounced toxicity in human breast cancer cell lines, inclusive of variants that were highly resistant to other therapeutic treatments, such as doxorubicin, paclitaxel, or trastuzumab. The anti-tumor effects of celecoxib were mimicked at increased potency by its non-coxib analog, 2,5-dimethyl-celecoxib (DMC), but were substantially weaker in the case of unmethylated-celecoxib (UMC), a derivative with superior COX-2 inhibitory efficacy. We conclude that the anti-tumor effects of nelfinavir can be enhanced by celecoxib analogs in a COX-2 independent fashion via the aggravation of ER stress, and such drug combinations should be considered as a beneficial adjunct to the treatment of drug-resistant breast cancers.


Nucleic Acids Research | 2012

Inhibition of the function of class IIa HDACs by blocking their interaction with MEF2

Nimanthi Jayathilaka; Aidong Han; Kevin J. Gaffney; Raja Dey; Jamie A. Jarusiewicz; Kaori Noridomi; Michael A. Philips; Xiao Lei; Ju He; Jun Ye; Tao Gao; Nicos A. Petasis; Lin Chen

Enzymes that modify the epigenetic status of cells provide attractive targets for therapy in various diseases. The therapeutic development of epigenetic modulators, however, has been largely limited to direct targeting of catalytic active site conserved across multiple members of an enzyme family, which complicates mechanistic studies and drug development. Class IIa histone deacetylases (HDACs) are a group of epigenetic enzymes that depends on interaction with Myocyte Enhancer Factor-2 (MEF2) for their recruitment to specific genomic loci. Targeting this interaction presents an alternative approach to inhibiting this class of HDACs. We have used structural and functional approaches to identify and characterize a group of small molecules that indirectly target class IIa HDACs by blocking their interaction with MEF2 on DNA.Weused X-ray crystallography and 19F NMRto show that these compounds directly bind to MEF2. We have also shown that the small molecules blocked the recruitment of class IIa HDACs to MEF2-targeted genes to enhance the expression of those targets. These compounds can be used as tools to study MEF2 and class IIa HDACs in vivo and as leads for drug development.


Cancer Letters | 2011

Enhancement of photodynamic therapy by 2,5-dimethyl celecoxib, a non-cyclooxygenase-2 inhibitor analog of celecoxib

Angela Ferrario; Sophia Lim; Frank Xu; Marian Luna; Kevin J. Gaffney; Nicos A. Petasis; Axel H. Schönthal; Charles J. Gomer

Photodynamic therapy (PDT) effectiveness can be improved by employing combined modality approaches involving pharmaceuticals targeting the tumor microenvironment and/or tumor cell death pathways. In one approach, combining PDT with celecoxib improves long-term tumoricidal activity without increasing normal tissue photosensitization. However, side effects arising from the use of coxib based cyclooxygenase-2 (COX-2) inhibitors, including cardiovascular injury, decreases the clinical applications of this class of compounds. A growing number of studies demonstrate that the tumoricidal actions of coxibs such as celecoxib involve non-COX-2 mediated mechanisms. The celecoxib analog, 2,5-dimethyl celecoxib (DMC), lacks COX-2 inhibitory activity but exhibits cytotoxic properties comparable to the COX-2 inhibitor celecoxib. We compared the effectiveness of DMC and celecoxib in modulating PDT response at both the in vitro and in vivo level using a C3H/BA murine mammary carcinoma model. Both DMC and celecoxib blocked PDT induced expression of the pro-survival protein survivin, enhanced the endoplasmic reticulum stress (ERS) response of PDT, and increased both apoptosis and cytotoxicity in BA cells exposed to combination protocols. DMC enhanced the in vivo tumoricidal responsiveness of PDT without altering PGE2 levels. Our data demonstrates that DMC improved PDT by increasing apoptosis and tumoricidal activity without modulating COX-2 catalytic activity. Our results also suggest that celecoxib mediated enhancement of PDT may involve both COX-2 dependent and independent mechanisms.


Molecular Cancer Therapeutics | 2010

Antiangiogenic Activities of 2,5-Dimethyl-Celecoxib on the Tumor Vasculature

Jenilyn J. Virrey; Zhi Liu; Hee-Yeon Cho; Adel Kardosh; Encouse B. Golden; Stan G. Louie; Kevin J. Gaffney; Nicos A. Petasis; Axel H. Schönthal; Thomas C. Chen; Florence M. Hofman

Our laboratory has previously shown that a novel compound, 2,5-dimethyl-celecoxib (DMC), which is structurally similar to the cyclooxygenase-2 (COX-2) inhibitor celecoxib but lacks the COX-2–inhibitory function, mimics the antitumor effects of celecoxib. Most studies on DMC, however, focused on its effects on tumor cells. Here, we investigated the activities of DMC as an antiangiogenic agent in both in vitro and in vivo systems. Using primary cultures of human glioma specimens, we found that DMC treatment was cytotoxic to tumor-associated brain endothelial cells (TuBEC), which was mediated through the endoplasmic reticulum stress pathway. In contrast, confluent cultures of quiescent human BEC did not undergo cell death. DMC potently suppressed the proliferation and migration of the TuBEC. DMC caused no apparent effects on the secretion of vascular endothelial growth factor and interleukin-8 but inhibited the secretion of endothelin-1 in tumor-associated EC. DMC treatment of glioma xenografts in mice resulted in smaller tumors with a pronounced reduction in microvessel density compared with untreated mice. In vitro and in vivo analyses confirmed that DMC has antivascular activity. Considering that DMC targets both tumor cells and tumor-associated ECs, this agent is a promising anticancer drug. Mol Cancer Ther; 9(3); 631–41


Cancer Research | 2014

Abstract 1811: Evaluation of anti-CXCR2 small molecule inhibitors as novel chemotherapy targeting the Interleukin-8 pathway in colorectal cancer

Yinghui Jane E. Huang; Kevin J. Gaffney; Ethan Gerdts; Nicos A. Petasis; Heinz-Josef Lenz; Melissa J. LaBonte

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Despite new and improved chemotherapy treatments for colorectal cancer (CRC) contributing to enhanced treatment response rates and patient prognosis, it has been suggested that an overwhelming 90% of metastatic CRC patients encounter treatment failure due to drug resistance. The Interleukin-8/CXCR2 pathway has been validated as a promising target for novel chemotherapy due to its overarching effects on various cell types within the tumor microenvironment that ultimately drive tumor growth, proliferation, survival, chemoresistance, metastasis and angiogenesis. A series of homologous small molecule inhibitors, which competitively bind to the active site of CXCR2, was developed and evaluated in terms of their efficacy in inhibiting the various cellular responses induced by IL-8/CXCR2 signaling, utilizing MTS cell proliferation, angiogenesis tube formation, colony formation, Western blot, cell migration and invasion assays. The anti-CXCR2 compounds developed produce potent repression of proliferation and colony forming capacity in HCT116, HT29, SW620 and LoVo CRC cell lines, achieving IC50 values as low as 9.6 μM in HCT116 and 9.6 μM in HT29, along with colony formation fold changes as low as .163 in HCT116 and .255 in HT29. Additionally, the compounds demonstrated inhibition of angiogenesis, migration and invasion and induced significant down-regulation of pathways downstream of CXCR2, including NF-κB, MAPK and Akt. The proposed targeting of the IL-8 pathway offers an attractive and promising approach to CRC therapy. Based on the structure and functional relationship of these compounds, new inhibitors will be designed and validated in hopes of having improved drug properties by utilizing our in vitro and in vivo assay systems. Citation Format: Yinghui Jane E. Huang, Kevin J. Gaffney, Ethan Gerdts, Nicos A. Petasis, Heinz-Josef Lenz, Melissa J. LaBonte. Evaluation of anti-CXCR2 small molecule inhibitors as novel chemotherapy targeting the Interleukin-8 pathway in colorectal cancer. [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 1811. doi:10.1158/1538-7445.AM2014-1811


Blood | 2009

Green tea polyphenols block the anticancer effects of bortezomib and other boronic acid-based proteasome inhibitors

Encouse B. Golden; Philip Y. Lam; Adel Kardosh; Kevin J. Gaffney; Enrique Cadenas; Stan G. Louie; Nicos A. Petasis; Thomas C. Chen; Axel H. Schönthal


Leukemia Research | 2010

Cytotoxic effects of celecoxib on Raji lymphoma cells correlate with aggravated endoplasmic reticulum stress but not with inhibition of cyclooxygenase-2.

Szu-Ting Chen; Simmy Thomas; Kevin J. Gaffney; Stan G. Louie; Nicos A. Petasis; Axel H. Schönthal


Organic and Biomolecular Chemistry | 2015

Molecular characterization of the boron adducts of the proteasome inhibitor bortezomib with epigallocatechin-3-gallate and related polyphenols

Stephen J. Glynn; Kevin J. Gaffney; Marcos A. Sainz; Stan G. Louie; Nicos A. Petasis

Collaboration


Dive into the Kevin J. Gaffney's collaboration.

Top Co-Authors

Avatar

Nicos A. Petasis

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Stan G. Louie

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Axel H. Schönthal

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas C. Chen

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Adel Kardosh

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Florence M. Hofman

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Hee-Yeon Cho

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Simmy Thomas

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Gere S. diZerega

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge