1
|
Li J, Wang X, Zhang H, Hu X, Peng X, Jiang W, Zhuo L, Peng Y, Zeng G, Wang Z. Fenamates: Forgotten treasure for cancer treatment and prevention: Mechanisms of action, structural modification, and bright future. Med Res Rev 2024. [PMID: 39171404 DOI: 10.1002/med.22079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Fenamates as classical nonsteroidal anti-inflammatory agents are widely used for relieving pain. Preclinical studies and epidemiological data highlight their chemo-preventive and chemotherapeutic potential for cancer. However, comprehensive reviews of fenamates in cancer are limited. To accelerate the repurposing of fenamates, this review summarizes the results of fenamates alone or in combination with existing chemotherapeutic agents. This paper also explores targets of fenamates in cancer therapy, including COX, AKR family, AR, gap junction, FTO, TEAD, DHODH, TAS2R14, ion channels, and DNA. Besides, this paper discusses other mechanisms, such as regulating Wnt/β-catenin, TGF-β, p38 MAPK, and NF-κB pathway, and the regulation of the expressions of Sp, EGR-1, NAG-1, ATF-3, ErbB2, AR, as well as the modulation of the tumor immune microenvironment. Furthermore, this paper outlined the structural modifications of fenamates, highlighting their potential as promising leads for anticancer drugs.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoling Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weifan Jiang
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guo Zeng
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Abdelhaleem EF, Kassab AE, El-Nassan HB, Khalil OM. Design, synthesis, and biological evaluation of new celecoxib analogs as apoptosis inducers and cyclooxygenase-2 inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200190. [PMID: 35976138 DOI: 10.1002/ardp.202200190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022]
Abstract
Series of new celecoxib analogs were synthesized to assess their anticancer activity against the MCF-7 cell line. Four compounds, 3a, 3c, 5b, and 5c, showed 1.4-9.2-fold more potent anticancer activity than celecoxib. The antiproliferative activity of the most potent compounds, 3c, 5b, and 5c, seems to be associated well with their ability to induce apoptosis in MCF-7 cells (18-24-fold). This evidence was supported by an increase in the expression of the tumor suppressor gene p53 (4-6-fold), the elevation in the Bax/BCL-2 ratio, and a significant increase in the level of active caspase-7 (4-7-fold). Moreover, compounds 3c and 5c showed significant cyclooxygenase-2 (COX-2) inhibitory activity. They were also docked into the crystal structure of the COX-2 enzyme (PDB ID: 3LN1) to understand their mode of binding.
Collapse
Affiliation(s)
- Eman F Abdelhaleem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala B El-Nassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Omneya M Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Abdelhaleem EF, Kassab AE, El-Nassan HB, Khalil OM. Design and Synthesis of Novel Celecoxib Analogues with Potential Cytotoxic and Pro-apoptotic Activity Against Breast Cancer Cell Line MCF-7. Med Chem 2022; 18:903-914. [PMID: 35264093 DOI: 10.2174/1573406418666220309123648] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/11/2022] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast cancer is currently the leading cause of worldwide cancer incidence exceeding lung cancer. In addition, breast cancer accounts for 1 in 4 cancer cases and 1 in 6 cancer deaths among women. Cytotoxic chemotherapy is still the main therapeutic approach for patients with metastatic breast cancer. OBJECTIVE To synthesize a series of novel celecoxib analogues to evaluate their anticancer activity against MCF-7 cell line. METHOD Our design of target compounds was based on preserving the pyrazole moiety of celecoxib attached to two phenyl rings, one of them having polar hydrogen bonding group (sulfonamide or methoxy group). The methyl group of the second phenyl ring was replaced with chlorine or bromine atom. Finally, the trifluoromethyl group was replaced with arylidene hydrazine-1-carbonyl moiety, which is substituted either with fluoro or methoxy group, offering various electronic and lipophilic environments. These modifications were carried out to investigate their effects on the anti-proliferative activity of the newly synthesized celecoxib analogues and to provide a valuable structure activity relationship. RESULTS Four compounds namely (4e-h) exhibited significant antitumor activity. Compounds 4e, 4f and 4h showed 1.2-2 folds more potent anticancer activity than celecoxib. Celecoxib analogue 4f showed the most potent anti-proliferative activity. Its anti-proliferative activity seems to associate well with its ability to inhibit BCL-2. Moreover, activation of damage response pathway of the DNA leads to cell cycle arrest at G2/M phase, accumulation of cells in pre-G1 phase, indicating that cell death proceeds through an apoptotic mechanism. Compound 4f exhibited potent pro-apoptotic effect via induction of the intrinsic mitochondrial pathway of apoptosis. This mechanistic pathway was proved by a significant increase in the expression of the tumor suppressor gene p53, elevation in Bax/BCL-2 ratio and a significant increase in the level of active caspase-7. Furthermore, compound 4f showed moderate COX-2 inhibitory activity. CONCLUSION Celecoxib analogue 4f is a promising multi-targeted lead for the design and synthesis of potent anticancer agents.
Collapse
Affiliation(s)
- Eman F Abdelhaleem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Hala B El-Nassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Omneya M Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| |
Collapse
|
4
|
Metal complexes of NSAIDs as potent anti-tumor chemotherapeutics: Mechanistic insights into cytotoxic activity via multiple pathways primarily by inhibition of COX–1 and COX–2 enzymes. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214316] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
5
|
Emerging role of ferroptosis in breast cancer: New dawn for overcoming tumor progression. Pharmacol Ther 2021; 232:107992. [PMID: 34606782 DOI: 10.1016/j.pharmthera.2021.107992] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer has become a serious threat to women's health. Cancer progression is mainly derived from resistance to apoptosis induced by procedures or therapies. Therefore, new drugs or models that can overcome apoptosis resistance should be identified. Ferroptosis is a recently identified mode of cell death characterized by excess reactive oxygen species-induced lipid peroxidation. Since ferroptosis is distinct from apoptosis, necrosis and autophagy, its induction successfully eliminates cancer cells that are resistant to other modes of cell death. Therefore, ferroptosis may become a new direction around which to design breast cancer treatment. Unfortunately, the complete appearance of ferroptosis in breast cancer has not yet been fully elucidated. Furthermore, whether ferroptosis inducers can be used in combination with traditional anti- breast cancer drugs is still unknown. Moreover, a summary of ferroptosis in breast cancer progression and therapy is currently not available. In this review, we discuss the roles of ferroptosis-associated modulators glutathione, glutathione peroxidase 4, iron, nuclear factor erythroid-2 related factor-2, superoxide dismutases, lipoxygenase and coenzyme Q in breast cancer. Furthermore, we provide evidence that traditional drugs against breast cancer induce ferroptosis, and that ferroptosis inducers eliminate breast cancer cells. Finally, we put forward prospect of using ferroptosis inducers in breast cancer therapy, and predict possible obstacles and corresponding solutions. This review will deepen our understanding of the relationship between ferroptosis and breast cancer, and provide new insights into breast cancer-related therapeutic strategies.
Collapse
|
6
|
AĞDAŞ F, ERYILMAZ A, GÖKMEN YILMAZ E, ERGİN K. THE EFFECTS OF SULINDAC ON CELL VIABILITY, CELL CYCLE AND ANJIOGENEZİS IN PHARYNGEAL CANCER CELL. ENT UPDATES 2020. [DOI: 10.32448/entupdates.750434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
7
|
Kumari R, Banerjee S, Roy P, Nath M. Organotin(IV) complexes of NSAID, ibuprofen, X‐ray structure of Ph
3
Sn(IBF), binding and cleavage interaction with DNA and
in vitro
cytotoxic studies of several organotin complexes of drugs. Appl Organomet Chem 2019. [DOI: 10.1002/aoc.5283] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Ranjana Kumari
- Department of ChemistryIndian Institute of Technology Roorkee Roorkee 247667 India
| | - Somesh Banerjee
- Department of BiotechnologyIndian Institute of Technology Roorkee Roorkee 247667 India
| | - Partha Roy
- Department of BiotechnologyIndian Institute of Technology Roorkee Roorkee 247667 India
| | - Mala Nath
- Department of ChemistryIndian Institute of Technology Roorkee Roorkee 247667 India
| |
Collapse
|
8
|
Chikara S, Nagaprashantha LD, Singhal J, Horne D, Awasthi S, Singhal SS. Oxidative stress and dietary phytochemicals: Role in cancer chemoprevention and treatment. Cancer Lett 2017; 413:122-134. [PMID: 29113871 DOI: 10.1016/j.canlet.2017.11.002] [Citation(s) in RCA: 323] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022]
Abstract
Several epidemiological observations have shown an inverse relation between consumption of plant-based foods, rich in phytochemicals, and incidence of cancer. Phytochemicals, secondary plant metabolites, via their antioxidant property play a key role in cancer chemoprevention by suppressing oxidative stress-induced DNA damage. In addition, they modulate several oxidative stress-mediated signaling pathways through their anti-oxidant effects, and ultimately protect cells from undergoing molecular changes that trigger carcinogenesis. In several instances, however, the pro-oxidant property of these phytochemicals has been observed with respect to cancer treatment. Further, in vitro and in vivo studies show that several phytochemicals potentiate the efficacy of chemotherapeutic agents by exacerbating oxidative stress in cancer cells. Therefore, we reviewed multiple studies investigating the role of dietary phytochemicals such as, curcumin (turmeric), epigallocatechin gallate (EGCG; green tea), resveratrol (grapes), phenethyl isothiocyanate (PEITC), sulforaphane (cruciferous vegetables), hesperidin, quercetin and 2'-hydroxyflavanone (2HF; citrus fruits) in regulating oxidative stress and associated signaling pathways in the context of cancer chemoprevention and treatment.
Collapse
Affiliation(s)
- Shireen Chikara
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Lokesh Dalasanur Nagaprashantha
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Jyotsana Singhal
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sanjay Awasthi
- Department of Medical Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sharad S Singhal
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
9
|
Takhar H, Singhal N, Mislang A, Kumar R, Kim L, Selva-Nayagam S, Pittman K, Karapetis C, Borg M, Olver IN, Brown MP. Phase II study of celecoxib with docetaxel chemoradiotherapy followed by consolidation chemotherapy docetaxel plus cisplatin with maintenance celecoxib in inoperable stage III nonsmall cell lung cancer. Asia Pac J Clin Oncol 2017; 14:91-100. [DOI: 10.1111/ajco.12749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 06/22/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Harminder Takhar
- Cancer Clinical Trials Unit; Royal Adelaide Hospital; Adelaide South Australia Australia
| | - Nimit Singhal
- Cancer Clinical Trials Unit; Royal Adelaide Hospital; Adelaide South Australia Australia
| | - Anna Mislang
- Cancer Clinical Trials Unit; Royal Adelaide Hospital; Adelaide South Australia Australia
| | - Raj Kumar
- Department of Medical Oncology; Flinders Medical Centre and Flinders University; Adelaide South Australia Australia
| | - Laurence Kim
- Cancer Clinical Trials Unit; Royal Adelaide Hospital; Adelaide South Australia Australia
| | - Sid Selva-Nayagam
- Cancer Clinical Trials Unit; Royal Adelaide Hospital; Adelaide South Australia Australia
| | - Ken Pittman
- Department of Medical Oncology; The Queen Elizabeth Hospital; Woodville South Australia Australia
| | - Chris Karapetis
- Department of Medical Oncology; Flinders Medical Centre and Flinders University; Adelaide South Australia Australia
| | - Martin Borg
- Adelaide Radiotherapy Centre; Adelaide South Australia Australia
| | - Ian N. Olver
- Sansom Institute; University of South Australia; Adelaide South Australia Australia
| | - Michael P. Brown
- Cancer Clinical Trials Unit; Royal Adelaide Hospital; Adelaide South Australia Australia
- Sansom Institute; University of South Australia; Adelaide South Australia Australia
- Centre for Cancer Biology; SA Pathology and University of South Australia; Adelaide South Australia Australia
- Discipline of Medicine; University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|
10
|
Kumari R, Nath M. Tri- and diorganotin(IV) derivatives of non-steroidal anti-inflammatory drug sulindac: Characterization, electronic structures (DFT), DNA binding and plasmid cleavage studies. Appl Organomet Chem 2017. [DOI: 10.1002/aoc.3661] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Ranjana Kumari
- Department of Chemistry; Indian Institute of Technology Roorkee; Roorkee 247667 India
| | - Mala Nath
- Department of Chemistry; Indian Institute of Technology Roorkee; Roorkee 247667 India
| |
Collapse
|
11
|
Kim J, Shim M. COX-2 inhibitor NS-398 suppresses doxorubicin-induced p53 accumulation through inhibition of ROS-mediated Jnk activation. Mol Carcinog 2016; 55:2156-2167. [PMID: 26756900 DOI: 10.1002/mc.22458] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/13/2015] [Accepted: 12/28/2015] [Indexed: 12/26/2022]
Abstract
Cyclooxygenase-2 (COX-2) is one of the isoforms of cyclooxygenase, a rate-limiting enzyme in the arachidonic acid cascade. COX-2 protein expression is highly induced by numerous factors and it has been reportedly overexpressed in various human malignancies. Although anti-tumorigenic effects of COX-2 inhibitors have been shown, several lines of evidence suggest that COX-2 inhibitors antagonize the cytotoxicity of chemotherapeutic agents. In this study, we investigated the effect of NS-398, a COX-2 inhibitor, on modulation of doxorubicin (DOX)-induced p53 accumulation. Non-selective and selective COX-2 inhibitors attenuated DOX-induced accumulation of wild type (WT) but not mutant p53. Nutlin-3α or MG132 abolished the suppressive effect of a COX-2 inhibitor on DOX-induced p53 increase. Moreover, the DOX-induced increase in p53 protein levels was reduced in COX-2 knockout (KO) mouse embryonic fibroblasts (MEFs) compared to those in WT or COX-1 KO MEFs. DOX-induced accumulation of p53 was attenuated by a specific inhibitor or knockdown of Jun-N-terminal kinase (Jnk). In addition, DOX-induced Jnk activation was decreased in COX-2 KO MEFs or by COX-2 inhibition, suggesting that Jnk stabilizes p53 by a mechanism that involves COX-2. Pre-treatment with a reactive oxygen species (ROS) scavenger, N-acetylcysteine, attenuated DOX-induced Jnk activation and subsequent p53 accumulation. Furthermore, the absence or inhibition of COX-2 resulted in suppression of DOX-induced increase in ROS levels. These results suggest that COX-2 activates Jnk through modulation of ROS levels, leading to accumulation of p53. Our study identifies a putative novel cross-talk between COX-2 and p53. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joohwee Kim
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Minsub Shim
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
12
|
Hosseinimehr SJ, Nobakht R, Ghasemi A, Pourfallah TA. Radioprotective effect of mefenamic acid against radiation-induced genotoxicity in human lymphocytes. Radiat Oncol J 2015; 33:256-60. [PMID: 26484310 PMCID: PMC4607580 DOI: 10.3857/roj.2015.33.3.256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/20/2015] [Accepted: 06/29/2015] [Indexed: 12/23/2022] Open
Abstract
Purpose Mefenamic acid (MEF) as a non-steroidal anti-inflammatory drug is used as a medication for relieving of pain and inflammation. Radiation-induced inflammation process is involved in DNA damage and cell death. In this study, the radioprotective effect of MEF was investigated against genotoxicity induced by ionizing radiation in human blood lymphocytes. Materials and Methods Peripheral blood samples were collected from human volunteers and incubated with MEF at different concentrations (5, 10, 50, or 100 µM) for two hours. The whole blood was exposed to ionizing radiation at a dose 1.5 Gy. Lymphocytes were cultured with mitogenic stimulation to determine the micronuclei in cytokinesis blocked binucleated lymphocyte. Results A significant decreasing in the frequency of micronuclei was observed in human lymphocytes irradiated with MEF as compared to irradiated lymphocytes without MEF. The maximum decreasing in frequency of micronuclei was observed at 100 µM of MEF (38% decrease), providing maximal protection against ionizing radiation. Conclusion The radioprotective effect of MEF is probably related to anti-inflammatory property of MEF on human lymphocytes.
Collapse
Affiliation(s)
- Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reyhaneh Nobakht
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Arash Ghasemi
- Department of Radiology and Radiation Oncology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Tayyeb Allahverdi Pourfallah
- Department of Biochemistry and Biophysics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
13
|
|
14
|
Dimiza F, Fountoulaki S, Papadopoulos AN, Kontogiorgis CA, Tangoulis V, Raptopoulou CP, Psycharis V, Terzis A, Kessissoglou DP, Psomas G. Non-steroidal antiinflammatory drug-copper(II) complexes: structure and biological perspectives. Dalton Trans 2011; 40:8555-68. [PMID: 21805007 DOI: 10.1039/c1dt10714c] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Copper(II) complexes with the non-steroidal antiinflammatory drug mefenamic acid in the presence of aqua or nitrogen donor heterocyclic ligands (2,2'-bipyridine, 1,10-phenanthroline, 2,2'-bipyridylamine or pyridine) have been synthesized and characterized. The crystal structures of [(2,2'-bipyridine)bis(mefenamato)copper(II)], 2, [(2,2'-bipyridylamine)bis(mefenamato)copper(II)], 4, and [bis(pyridine)bis(methanol)bis(mefenamato)copper(II)], 5, have been determined by X-ray crystallography. UV study of the interaction of the complexes with calf-thymus DNA (CT DNA) has shown that the complexes can bind to CT DNA and [bis(aqua)tetrakis(mefenamato)dicopper(II)] exhibits the highest binding constant to CT DNA. The cyclic voltammograms of the complexes in the presence of CT DNA solution have shown that the complexes can bind to CT DNA by the intercalative binding mode verified also by DNA solution viscosity measurements. Competitive studies with ethidium bromide (EB) indicate that the complexes can displace the DNA-bound EB suggesting strong competition with EB. Mefenamic acid and its complexes exhibit good binding propensity to human or bovine serum albumin protein having relatively high binding constant values. All the compounds have been tested for their antioxidant and free radical scavenging activity as well as for their in vitro inhibitory activity against soybean lipoxygenase showing significant activity.
Collapse
Affiliation(s)
- Filitsa Dimiza
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, P.O. Box 135, GR-54124, Thessaloniki, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Khan MNA, Lee YS. Cyclooxygenase inhibitors: scope of their use and development in cancer chemotherapy. Med Res Rev 2011; 31:161-201. [PMID: 19967720 DOI: 10.1002/med.20182] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The traditional nonsteroidal anti-inflammatory drugs (NSAIDs) exert their effect by inhibition of cyclooxygenase-1 (COX-1) as well as COX-2 enzymes. As COX-1 is responsible for maintaining normal biological functions, the nonselective inhibition of these enzymes caused side effects including gastrointestinal (GI) problems. Recently developed selective COX-2 inhibitors could reduce these adverse effects, but the evidence of cardiovascular side effects including an increased risk of myocardial infarction began to emerge, and some of the COX-2 inhibitors were eventually withdrawn from the market and this led to the downfall of this research. So, the discovery of novel COX-2 inhibitors with their safety profile became the biggest challenge in pharmaceutical research. However, recent mechanistic and clinical studies revolutionized this area by indicating the fact that COX-2 is involved in apoptosis resistance, angiogenesis, and tumor progression. Epidemiological data suggest that selective COX-2 inhibitors might prevent the development of cancers. Moreover, COX-2 is found to be overexpressed in many cancers thus making it an attractive therapeutic target for the prevention and treatment of a number of malignancies. The purpose of this review is to focus on the medicinal chemistry aspects of COX-2 inhibitors in cancer chemotherapy and recent reports on these inhibitors as anticancer agents. We attempted to cover only the COX inhibitors that showed anticancer activity, although a number of potent COX-2 inhibitors have been reported without their anticancer effects. Furthermore, structure-activity relationships (SAR) of different classes of compounds for COX-2 inhibition as well as anticancer activity, and their future applications are discussed.
Collapse
Affiliation(s)
- Mohammed Naseer A Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 1 Hoegi-dong, Dongdaemoon-ku, Seoul 130-701, Republic of Korea
| | | |
Collapse
|
16
|
Ghosh N, Chaki R, Mandal V, Mandal SC. COX-2 as a target for cancer chemotherapy. Pharmacol Rep 2010; 62:233-44. [PMID: 20508278 DOI: 10.1016/s1734-1140(10)70262-0] [Citation(s) in RCA: 230] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 08/21/2009] [Indexed: 01/06/2023]
Abstract
Cyclooxygenase-1 and -2 (COX-1/2) catalyze the initial step in the formation of prostaglandins. Very recently their role in carcinogenesis has become more evident. They influence apoptosis, angiogenesis, and invasion, and play a key role in the production of carcinogens. Usually, a high level of COX-2 expression is found in cancer cells. Large epidemiological trials studying users and non-users of aspirin have shown that cyclooxygenase inhibitors and non-steroidal anti-inflammatory drugs (NSAIDs) could be of benefit against the development and growth of malignancies. Moreover, clinical trials in patients with familial adenomatosis polyposis syndrome have shown the efficacy of selective COX-2 inhibitors in the reduction of the number and size of colorectal polyps. Several preclinical studies show promising results with combinatorial treatments of either chemotherapy or radiotherapy with COX inhibitors. Preclinical studies with the simultaneous use of inhibitors of the epidermal growth factor receptor and COX-2 inhibitors have shown also promising results. Encouraging results from the first clinical trials combining chemotherapy with COX-2 inhibitors in patients with cancer in the advanced and neoadjuvant setting have recently been reported. Thus, it appears that targeting the COX-2 pathway is a promising strategy in the prevention and treatment of solid tumors.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, India.
| | | | | | | |
Collapse
|
17
|
Randomized double-blind phase II trial comparing gemcitabine plus LY293111 versus gemcitabine plus placebo in advanced adenocarcinoma of the pancreas. Cancer J 2010; 15:339-43. [PMID: 19672152 DOI: 10.1097/ppo.0b013e3181b36264] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND LY293111 (LY) is a novel oral anticancer agent with leukotriene B4 receptor antagonist and peroxisome proliferator-activated receptor gamma agonist properties, producing promising results alone and in combination with gemcitabine in pancreatic cancer xenograft models. A phase I study proved that the combination (gemcitabine plus LY) is safe and well tolerated. PATIENTS AND METHODS Chemotherapy-naive patients with histologically confirmed locally advanced or metastatic adenocarcinoma of the pancreas were randomly assigned to gemcitabine 1000 mg/m on days 1, 8, and 15 of a 28-day cycle and continuously administered LY 600 mg twice daily or gemcitabine 1000 mg/m on days 1, 8, and 15 of a 28-day cycle and daily oral placebo. Arms were balanced for Eastern Cooperative Oncology Group performance status and disease stage. The primary end point was 6-month survival; secondary objectives include response rate (RR), progression-free survival, and overall survival. RESULTS Six-month survival was not different between groups (P>0.2, 1-sided); progression-free survival and RR were not different (P>0.05, 2-sided). RR was also not impacted. LY did not increase grades 3-4 hematologic toxicities, but was associated with a trend toward more, grades 3-4 diarrhea. CONCLUSIONS These results do not demonstrate any benefit to adding LY to gemcitabine in unpretreated patients with advanced pancreatic carcinoma.
Collapse
|
18
|
Agarwal S, Reddy GV, Reddanna P. Eicosanoids in inflammation and cancer: the role of COX-2. Expert Rev Clin Immunol 2010; 5:145-65. [PMID: 20477063 DOI: 10.1586/1744666x.5.2.145] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Eicosanoids, a family of oxygenated metabolites of eicosapolyenoic fatty acids, such as arachidonic acid, formed via the lipoxygenase, cyclooxygenase (COX) and epoxygenase pathways, play an important role in the regulation of various pathophysiological processes, including inflammation and cancer. COX-2, the inducible isoform of COX, has emerged as the key enzyme regulating inflammation, and promises to play a considerable role in cancer. Although NSAIDs have been in use for centuries, the COX-2 selective inhibitors - coxibs - have emerged as potent anti-inflammatory drugs with fewer gastric side effects. As COX-2 plays a major role in neoplastic transformation and cancer growth, by downregulating apoptosis and promoting angiogenesis, invasion and metastasis, coxibs have a potential role in the prevention and treatment of cancer. Recent studies indicate their possible application in overcoming drug resistance by downregulating the expression of MDR-1. However, the cardiac side effects of some of the coxibs have limited their application in treating various inflammatory disorders and warrant the development of COX-2 inhibitors without side effects. This review will focus on the role of COX-2 in inflammation and cancer, with an emphasis on novel approaches to the development of COX-2 inhibitors without side effects.
Collapse
Affiliation(s)
- Smita Agarwal
- Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India.
| | | | | |
Collapse
|
19
|
Rocconi RP, Kirby TO, Seitz RS, Beck R, Straughn JM, Alvarez RD, Huh WK. Lipoxygenase Pathway Receptor Expression in Ovarian Cancer. Reprod Sci 2008; 15:321-6. [DOI: 10.1177/1933719108316390] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rodney P. Rocconi
- Department of Obstetrics and Gynecology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama,
| | - Tyler O. Kirby
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Alabama
| | | | - Rod Beck
- Applied Genomics, Inc, Huntsville Alabama
| | - J. Michael Straughn
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Alabama
| | - Ronald D. Alvarez
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Alabama
| | - Warner K. Huh
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama at Birmingham, Alabama
| |
Collapse
|
20
|
Pidgeon GP, Lysaght J, Krishnamoorthy S, Reynolds JV, O'Byrne K, Nie D, Honn KV. Lipoxygenase metabolism: roles in tumor progression and survival. Cancer Metastasis Rev 2008; 26:503-24. [PMID: 17943411 DOI: 10.1007/s10555-007-9098-3] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The metabolism of arachidonic acid through lipoxygenase pathways leads to the generation of various biologically active eicosanoids. The expression of these enzymes vary throughout the progression of various cancers, and thereby they have been shown to regulate aspects of tumor development. Substantial evidence supports a functional role for lipoxygenase-catalyzed arachidonic and linoleic acid metabolism in cancer development. Pharmacologic and natural inhibitors of lipoxygenases have been shown to suppress carcinogenesis and tumor growth in a number of experimental models. Signaling of hydro[peroxy]fatty acids following arachidonic or linoleic acid metabolism potentially effect diverse biological phenomenon regulating processes such as cell growth, cell survival, angiogenesis, cell invasion, metastatic potential and immunomodulation. However, the effects of distinct LOX isoforms differ considerably with respect to their effects on both the individual mechanisms described and the tumor being examined. 5-LOX and platelet type 12-LOX are generally considered pro-carcinogenic, with the role of 15-LOX-1 remaining controversial, while 15-LOX-2 suppresses carcinogenesis. In this review, we focus on the molecular mechanisms regulated by LOX metabolism in some of the major cancers. We discuss the effects of LOXs on tumor cell proliferation, their roles in cell cycle control and cell death induction, effects on angiogenesis, migration and the immune response, as well as the signal transduction pathways involved in these processes. Understanding the molecular mechanisms underlying the anti-tumor effect of specific, or general, LOX inhibitors may lead to the design of biologically and pharmacologically targeted therapeutic strategies inhibiting LOX isoforms and/or their biologically active metabolites, that may ultimately prove useful in the treatment of cancer, either alone or in combination with conventional therapies.
Collapse
Affiliation(s)
- Graham P Pidgeon
- Department of Clinical Surgery, Trinity College Dublin, St. James Hospital, Dublin 8, Ireland.
| | | | | | | | | | | | | |
Collapse
|
21
|
Horikawa Y, Otaka M, Komatsu K, Jin M, Odashima M, Wada I, Matsuhashi T, Ohba R, Oyake J, Hatakeyama N, Dubois RN, Watanabe S. MEK activation suppresses CPT11-induced apoptosis in rat intestinal epithelial cells through a COX-2-dependent mechanism. Dig Dis Sci 2007; 52:2757-65. [PMID: 17393318 DOI: 10.1007/s10620-007-9798-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Accepted: 01/29/2007] [Indexed: 12/14/2022]
Abstract
Resistance to chemotherapeutic agents is one of the distinct features of cancer cells. We evaluate the role of activated MEK-ERK signaling in Camptotecin/irinotecan (CPT-11)-induced cell death using constitutively activated MEK1-transfected normal rat intestinal epithelial cells (IEC-caMEK cells). A CPT-11-induced inhibitory concentration of 50% was determined by WST assay. Apoptosis was evaluated by DNA staining and fragmented DNA analysis. Protein expressions were analyzed by western blotting. We also examined the role of cyclooxygenase-2 in the cell systems. IEC-caMEK cells possessed survival advantages compared to control cells. Apoptosis was remarkably suppressed in IEC-caMEK cells. Western blot analysis revealed increased expression of Bcl-2, Bcl-xL, Mcl-1, and COX-2 and decreased expression of Bak in IEC-caMEK cells. The COX-2 selective inhibitor ameliorated the antiapoptotic nature of IEC-caMEK cells. MEK activation suppressed CPT-11-induced apoptosis in IEC-caMEK cells via a COX-2- dependent mechanism. Therefore, MEK-ERK signaling may contribute to the drug-resistant nature of cancer cells.
Collapse
Affiliation(s)
- Youhei Horikawa
- Department of Gastroenterology/Internal Medicine, Akita University School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Scheper MA, Nikitakis NG, Chaisuparat R, Montaner S, Sauk JJ. Sulindac induces apoptosis and inhibits tumor growth in vivo in head and neck squamous cell carcinoma. Neoplasia 2007; 9:192-9. [PMID: 17401459 PMCID: PMC1838577 DOI: 10.1593/neo.06781] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 01/18/2007] [Accepted: 01/19/2007] [Indexed: 12/28/2022] Open
Abstract
Sulindac has antineoplastic effects on various cancer cell lines; consequently, we assessed sulindac's effects on laryngeal squamous cell carcinoma (SCC) cells in vitro and in vivo. In vitro, SCC (HEP-2) cells treated with various cyclooxygenase inhibitors or transfected with constitutively active signal transducer and activator of transcription 3 (Stat3) or survivin vectors were analyzed using Western blot analysis, annexin V assay, and cell proliferation assay. In parallel, nude mice injected subcutaneously with HEP-2 cells were either treated intraperitoneally with sulindac or left untreated, and analyzed for tumor weight, survivin expression, and tyrosine-phosphorylated Stat3 expression. In vitro studies confirmed the selective antiproliferative and proapoptotic effects of sulindac, which also downregulated Stat3 and survivin protein expression. Stat3 or survivin forced expression partially rescued the antiproliferative effects of sulindac. In vivo studies showed significant repression of HEP-2 xenograft growth in sulindactreated mice versus controls, with near-complete resolution at 10 days. Additionally, tumor specimens treated with sulindac showed downregulation of phosphorylated tyrosine-705 Stat3 and survivin expression. Taken together, our data suggest, for the first time, a specific inhibitory effect of sulindac on tumor growth and survivin expression in laryngeal cancer, both in vitro and in vivo, in a Stat3-dependent manner, suggesting a novel therapeutic approach to head and neck cancer.
Collapse
Affiliation(s)
- Mark A Scheper
- Department of Diagnostic Sciences and Pathology, University of Maryland, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
23
|
de Groot DJA, de Vries EGE, Groen HJM, de Jong S. Non-steroidal anti-inflammatory drugs to potentiate chemotherapy effects: from lab to clinic. Crit Rev Oncol Hematol 2006; 61:52-69. [PMID: 16945549 DOI: 10.1016/j.critrevonc.2006.07.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Revised: 06/30/2006] [Accepted: 07/06/2006] [Indexed: 11/20/2022] Open
Abstract
Most solid tumors express the cyclooxygenase-2 (COX-2) protein, a target of NSAIDs. COX-2 overexpression in tumorsis considered a predictor of more advanced stage disease and of worse prognosis in a number of studies investigating solid malignancies. Therefore, NSAIDs are evaluated as anti-cancer drugs. NSAIDs inhibit proliferation, invasiveness of tumors, and angiogenesis and overcome apoptosis resistance in a COX-2 dependent and independent manner. This review will focus on the rationale behind NSAIDs, including selective COX-2 inhibitors, in combination with conventional chemotherapeutic drugs or novel molecular targeted drugs. Studies investigating anti-cancer effects of NSAIDs on cell lines and xenograft models have shown modulation of the Akt, NF-kappaB, tyrosine kinase and the death receptor-mediated apoptosis pathways. COX-2 expression in tumors is not yet used as biomarker in the clinic. Despite the increased risk on cardiovascular toxicity induced by selective COX-2 inhibitors, several ongoing clinical trials are still investigating the therapeutic benefits of NSAIDs in oncology. The anti-tumor effects in these trials balanced with the side effects data will define the precise role of selective COX-2 inhibitors in the treatment of cancer patients.
Collapse
Affiliation(s)
- D J A de Groot
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | | | | | | |
Collapse
|
24
|
Méric JB, Rottey S, Olaussen K, Soria JC, Khayat D, Rixe O, Spano JP. Cyclooxygenase-2 as a target for anticancer drug development. Crit Rev Oncol Hematol 2006; 59:51-64. [PMID: 16531064 DOI: 10.1016/j.critrevonc.2006.01.003] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2005] [Revised: 01/10/2006] [Accepted: 01/10/2006] [Indexed: 02/02/2023] Open
Abstract
The two isoforms cyclooxygenase-1 and -2 catalyze the initial step in the formation of prostaglandins in a variety of pathophysiological processes. More recently their role in carcinogenesis has become more evident. They seem to influence apoptosis, angiogenesis, and invasion, and play a role in the production of carcinogens. Usually, a high level of COX-2 expression is found in cancer cells. However, low COX-2 expression is observed in some cancers like prostate or breast cancer. This phenomenon is quite surprising and should influence on clinical trial designs. Large epidemiological trials studying users and non-users of aspirin have shown that cyclooxygenase (COX) inhibitors and non-steroidal anti-inflammatory drugs (NSAIDs) could be of benefit against the development and growth of malignancies. Moreover, clinical trials in patients with familial adenomatosis polyposis syndrome have shown too the efficacy of non-selective COX inhibitors and recently also of selective COX-2 inhibitors in the reduction of the number and the size of colorectal polyps. However, a primary chemopreventive effect has not been demonstrated yet. NSAIDs are also supposed to have a preventive and growth inhibitory effect in extra-colonic epithelial malignancies. Several preclinical studies show promising results with combination treatments of either chemotherapy or radiotherapy with COX inhibitors. Preclinical studies with the simultaneous use of inhibitors of the epidermal growth factor receptor and COX-2 inhibitors have shown also promising results. Encouraging results with the first clinical trials combining chemotherapy with COX-2 inhibitors in patients with cancer in the advanced and neoadjuvant setting have recently been reported. However, NSAIDs effects in cancer cells are mediated not only by COX enzymes but also by interactions with downstream effectors of COX-2. Hence, we can state that targeting the COX-2 pathway is a promising strategy in the prevention and treatment of solid tumors. Ongoing trials are expected to answer - at least partly - the remaining questions concerning COX-2 and cancer.
Collapse
Affiliation(s)
- Jean-Baptiste Méric
- Service d'Oncologie Médicale, Hôpital Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75651 Paris Cedex 13, France.
| | | | | | | | | | | | | |
Collapse
|
25
|
Shaik MS, Chatterjee A, Jackson T, Singh M. Enhancement of antitumor activity of docetaxel by celecoxib in lung tumors. Int J Cancer 2006; 118:396-404. [PMID: 16052515 PMCID: PMC2907249 DOI: 10.1002/ijc.21325] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Our study investigates the effect of a highly selective cyclooxygenase-2 (COX-2) inhibitor, celecoxib, on the cytotoxicity of docetaxel in nude mice bearing A549 tumor xenografts and elucidates the molecular mechanisms of the antitumor effect of this combination. Female nu/nu mice, xenografted with s.c. A549 tumors were treated with either celecoxib (150 mg/kg/day), docetaxel (10 mg/kg) or a combination of both. The tumor tissues were quantified for the induction of apoptosis, intratumor levels/expressions of prostaglandin E2 (PGE2), 15 deoxy prostaglandin J2 (15-d PGJ2), microsomal prostaglandin E synthase (mPGES) and cytoplasmic phospholipase A2 (cPLA2). The combination of celecoxib with docetaxel significantly inhibited the tumor growth (p < 0.03) as compared to celecoxib or docetaxel alone, decreased the levels of PGE2 by 10-fold and increased the 15-d PGJ2 levels by 4-fold as compared to control. The combination also enhanced the peroxisome proliferator-activated receptor (PPAR)-gamma expression, decreased the expression of cPLA2, mPGES and vascular endothelial growth factor (VEGF), but had no effect on the expression of COX-1 or COX-2 in tumor tissues. TUNEL staining of the tumor tissues showed a marked increase in the apoptosis in the combination group as compared to the celecoxib- or docetaxel-treated groups and this was associated with an increase in the intratumor p53 expression. In conclusion, the combination of celecoxib with docetaxel produces a greater antitumor effect in s.c. A549 tumors as compared to celecoxib or docetaxel alone and this effect is associated with concomitant alterations in the intratumor levels of PGE2 and 15-d PGJ2.
Collapse
Affiliation(s)
| | | | - Tanise Jackson
- College of Pharmacy, Florida A&M University, Tallahassee, FL, USA
| | - Mandip Singh
- College of Pharmacy, Florida A&M University, Tallahassee, FL, USA
| |
Collapse
|
26
|
Christofis P, Katsarou M, Papakyriakou A, Sanakis Y, Katsaros N, Psomas G. Mononuclear metal complexes with Piroxicam: Synthesis, structure and biological activity. J Inorg Biochem 2005; 99:2197-210. [PMID: 16176832 DOI: 10.1016/j.jinorgbio.2005.07.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 07/28/2005] [Accepted: 07/30/2005] [Indexed: 12/21/2022]
Abstract
Piroxicam (=Hpir) is a non-steroidal anti-inflammatory and an anti-arthritic drug. VO(2+), Mn(2+), Fe(3+), MoO(2)(2+) and UO(2)(2+) complexes with deprotonated piroxicam have been prepared and characterized with the use of infrared, UV-Vis, nuclear magnetic resonance and electron paramagnetic resonance spectroscopies. The experimental data suggest that piroxicam acts as a deprotonated bidentate ligand in all complexes and is coordinated to the metal ion through the pyridine nitrogen and the amide oxygen. Molecular mechanics calculations in the gas state have been performed in order to propose a model for the Fe(3+), VO(2+) and MoO(2)(2+) complexes. Potential anticancer cytostatic and cytotoxic effects of piroxicam complexes with VO(2+), Mn(2+) and MoO(2)(2+) on human promyelocytic leukemia HL-60 cells have been investigated. Among all complexes, only VO(pir)(2)(H(2)O) clearly induces apoptosis after 24-h incubation, whereas piroxicam induces apoptosis after 57-h incubation.
Collapse
Affiliation(s)
- Petros Christofis
- Institute of Physical Chemistry, NCSR Demokritos, 15310 Aghia Paraskevi Attikis, GR15310 Athens, Greece
| | | | | | | | | | | |
Collapse
|
27
|
Schwartz GK, Weitzman A, O'Reilly E, Brail L, de Alwis DP, Cleverly A, Barile-Thiem B, Vinciguerra V, Budman DR. Phase I and pharmacokinetic study of LY293111, an orally bioavailable LTB4 receptor antagonist, in patients with advanced solid tumors. J Clin Oncol 2005; 23:5365-73. [PMID: 15939925 DOI: 10.1200/jco.2005.02.766] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE LY293111, a novel diaryl ether carboxylic acid derivative, is a potent and selective inhibitor of the lipoxygenase pathway either directly through 5'-lipoxygenase or via antagonism of the leukotriene B4 (LTB4) receptor. More recently it has been determined to have peroxisome proliferator activated receptor-gamma agonist (PPARgamma) activity. LY293111 has antineoplastic activity in a variety of preclinical models. The tolerability and pharmacokinetics of LY293111 administered continuously, by mouth, BID for repeat cycles of 21 days was evaluated. PATIENTS AND METHODS Thirty-eight patients with advanced solid tumors were treated at five dose levels (200 to 800 mg BID) for a total of 102 cycles. RESULTS The most common toxicity was diarrhea (76%). One patient at 600 mg BID (n = 11) and two at 800 mg BID (n = 8), experienced dose-limiting grade 3 diarrhea. Dose reductions and/or delays were infrequent. Increases in steady-state maximum plasma concentration (Cmax,ss) and area under the steady-state plasma concentration time curve 0 to 12 hours (AUCtau,ss) on day 8 could be considered to be dose-proportional over the four-fold-dose range. Interpatient variability in Cmax,ss and AUCtau,ss was estimated to be 65% and 71% respectively. There was a small increase in AUC (1.37; 90% CI, 0.85 to 2.21) between single and multiple doses. Two patients with progressive chondrosarcoma and melanoma had stable disease lasting approximately 336 and 168 days, respectively. CONCLUSION LY293111 can be administered safely by continuous oral therapy with mild toxicities. Diarrhea is dose-limiting. The recommended phase II dose will be 600 mg BID. The steady-state concentrations in humans exceed relevant levels observed in preclinical models.
Collapse
Affiliation(s)
- Gary K Schwartz
- Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Haynes A, Shaik MS, Chatterjee A, Singh M. Evaluation of an aerosolized selective COX-2 inhibitor as a potentiator of doxorubicin in a non-small-cell lung cancer cell line. Pharm Res 2004; 20:1485-95. [PMID: 14567645 PMCID: PMC2892336 DOI: 10.1023/a:1025774630993] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE To evaluate the in vitro effects of an aerosolized cyclooxygenase-2 (COX-2) inhibitor, nimesulide, on the cytotoxicity and apoptotic response of doxorubicin against the human lung adenocarcinoma cell line A549. METHODS Nimesulide was formulated into a metered dose inhaler (MDI) formulation and characterized for aerodynamic particle size and medication delivery. The in vitro cytotoxicity of nimesulide-MDI in the presence or absence of doxorubicin was assessed by using the six-stage viable impactor by an already standardized method. Induction of apoptosis in A549 cells by nimesulide (nonaerosolized or aerosolized) in combination with doxorubicin was evaluated by established techniques such as caspase-3 estimation and terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) staining. Finally, to understand the mechanism of action, the influence of different treatments on the expression of COX-2 and peroxisome proliferator-activated receptor-gamma (PPAR-gamma) in A549 cells was studied by immunoblotting. RESULTS The nimesulide-MDI formulation had a mass median aerodynamic diameter (MMAD) of 1.1 microm, (GSD = 2.8) and a medication delivery of 51 microg/shot. Nimesulide-MDI (40 shots) in combination with doxorubicin (0.01 microg/ml) had a cell kill of more than 60% as determined by in vitro cytotoxicity assay. The specific caspase-3 activity in A549 cells treated with nimesulide (40 microg/ml) and doxorubicin (0.25 microg/ml) in combination was 3 and 5 times higher than doxorubicin and nimesulide, respectively. Further, TUNEL staining showed apoptosis in over 30% of A549 cells treated with aerosolized nimesulide and doxorubicin combination vs. negligible as seen in cells treated individually. The expression of COX-2 was not altered in control or treatments, whereas PPAR-gamma was expressed only in the combination treatment. CONCLUSION Our results indicate that aerosolized nimesulide significantly enhances doxorubicin activity against A549 cells, and the enhanced cytotoxicity was probably mediated via a COX-2-independent mechanism.
Collapse
Affiliation(s)
- Alfred Haynes
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307
| | - Madhu Sudhan Shaik
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307
| | - Abhijit Chatterjee
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307
| | - M. Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307
- To whom correspondence should be addressed. ()
| |
Collapse
|
29
|
Davis TW, O'Neal JM, Pagel MD, Zweifel BS, Mehta PP, Heuvelman DM, Masferrer JL. Synergy between celecoxib and radiotherapy results from inhibition of cyclooxygenase-2-derived prostaglandin E2, a survival factor for tumor and associated vasculature. Cancer Res 2004; 64:279-85. [PMID: 14729635 DOI: 10.1158/0008-5472.can-03-1168] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Previous work has demonstrated that selective cyclooxygenase-2 (COX-2) inhibitors can act synergistically with radiotherapy to improve tumor debulking and control in preclinical models. The underlying mechanism of this remarkable activity has not yet been determined. Here, we report that radiation can elevate intratumoral levels of COX-2 protein and its products, particularly prostaglandin E(2) (PGE(2)). Furthermore, inhibition of COX-2 activity or neutralization of PGE(2) activity enhances radiotherapy even in tumors where COX-2 expression is restricted to the tumor neovasculature. Direct assessment of vascular function by direct contrast enhancement-magnetic resonance imaging showed that the combination of radiation and celecoxib lead to enhanced vascular permeability. These observations suggest that an important mechanism of celecoxib-induced radiosensitization involves inhibition of COX-2-derived PGE(2), thus removing a survival factor for the tumor and its vasculature.
Collapse
Affiliation(s)
- Thomas W Davis
- Oncology Discovery Research, Pfizer Corporation, St. Louis, Missouri 63017, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Davis TW, Hunter N, Trifan OC, Milas L, Masferrer JL. COX-2 inhibitors as radiosensitizing agents for cancer therapy. Am J Clin Oncol 2003; 26:S58-61. [PMID: 12902857 DOI: 10.1097/01.coc.0000074158.59269.9f] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Prostaglandins have long been known to impact the radiosensitivity of cells and tissues, and many studies have centered on exploiting nonspecific prostaglandin inhibitors such as NSAIDs for therapeutic gain. These studies have ultimately been unsuccessful due to the lack of targeted specificity against the tumor. The discovery of the inducible cyclooxygenase enzyme (COX-2) and development of some highly selective inhibitors (which spare the constitutive COX-1 activity) has renewed excitement for modulating tumor prostaglandins as a method of specific radiosensitization of tumors, while sparing normal tissues. This review discusses these new data and generates a rationale for use of COX-2 inhibitors as radiosensitizing agents in cancer therapy.
Collapse
|
31
|
Abstract
Non-steroidal anti-inflammatory agents (NSAIDs) inhibit the conversion of arachadonic acid to a class of inflammatory mediators known as eicosanoids. These minimally toxic drugs have demonstrated important anti-cancer properties in colorectal cancer and pre-clinical models have shown their potential for use in the treatment of non-small cell lung cancer (NSCLC). Clinical trials are underway investigating the efficacy of eicosanoid inhibitors alone and in combination with radiation and chemotherapy.
Collapse
Affiliation(s)
- Janessa J Laskin
- Department of Medical Oncology and Hematology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, 777 PRB, Nashville, TN 37215-6307, USA
| | | |
Collapse
|
32
|
Gibson AD. Cyclooxygenase-2: A Target for Chemoprevention and Treatment of Non–Small-Cell Lung Cancer. Clin Lung Cancer 2003; 4:273-6. [PMID: 14609443 DOI: 10.1016/s1525-7304(11)70292-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
33
|
Abstract
Lung cancer is one of the most frequent causes of cancer deaths worldwide. Non-small cell lung cancer (NSCLC) accounts for approximately 80% of cases and no curative treatment is available for the advanced stages of disease (stages III and IV), which comprise the majority of cases. Current treatment regimens with standard chemotherapy offer only a limited survival benefit, and, therefore, the development of new therapeutic strategies is needed. Novel chemotherapeutic drugs such as the epothilones, MEN 10755 and S-1 are being studied in patients with advanced stages of disease. Furthermore, a large number of therapies targeted against critical biological abnormalities in NSCLC are being investigated in clinical trials. The latter approach includes inhibition of growth factors, interference with abnormal signal transduction, inhibition of angiogenesis and gene replacement therapy. Promising results have thus far been obtained with some of these therapies. This review describes the role of new therapeutic agents in the treatment of NSCLC.
Collapse
Affiliation(s)
- Linda E Bröker
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | |
Collapse
|
34
|
Abstract
Following two decades of research on the biology of cancer and in particular of lung cancer, we have now a large number of molecular targets that can be utilized to create specific medicines against these cancers. Non-small cell lung cancer represents numerically the most important solid tumor in Western world, and is poorly affected by current therapies, where surgery represents almost the only curative therapy for about 25% of patients who are resectable at diagnosis. An abundant number of targeted therapies are being investigated in NSCLC. Among them are the metalloproteinase inhibitors, several tyrosine kinase inhibitors and several attempts of gene replacement have also been made. Promising results have so far been obtained with some of these approaches, and the outcome of large randomized studies is awaited. Small cell lung cancer (SCLC) represents about 20% of lung carcinomas, and several of the novel approaches that are being attempted for NSCLC, are also being investigated for SCLC. All these novel therapies open a new era of anticancer treatment that will likely complement the currently available therapies in the near future.
Collapse
Affiliation(s)
- Giuseppe Giaccone
- Division of Medical Oncology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Kovala-Demertzi D, Dokorou V, Ciunik Z, Kourkoumelis N, Demertzis MA. Organotin mefenamic complexes?preparations, spectroscopic studies and crystal structure of a triphenyltin ester of mefenamic acid: novel anti-tuberculosis agents. Appl Organomet Chem 2002. [DOI: 10.1002/aoc.308] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
36
|
Hattori K, Matsushita R, Kimura K, Abe Y, Nakashima E. Synergistic effect of indomethacin with adriamycin and cisplatin on tumor growth. Biol Pharm Bull 2001; 24:1214-7. [PMID: 11642337 DOI: 10.1248/bpb.24.1214] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, we have examined the antitumor effect of combined administrations of indomethacin (IND) with chemotherapeutic drugs on tumor growth. Colon 26 clone 20 (C20) cells and monocyte chemotactant protein-1 (MCP-1) transfected C20 cells (C20betaA-2-1) were used and these cells were inoculated into the footpad of BALB/c mice. At day 1 after tumor inoculation, treatment with 0.001% IND via the drinking water was commenced. At days 4, 6, and 8, adriamycin or cisplatin was administered intravenously at a dose of 5 mg/kg or intraperitoneally at a dose of 2 mg/kg, respectively. Although IND, adriamycin and cisplatin only partially reduced the growth of the C20 tumors after treatment with each drug on its own, a marked synergistic effect was observed when they were given in combination. A synergistic effect between IND and cisplatin on C20betaA-2-1 was also observed. However, IND itself showed no suppression of C20betaA-2-1 tumor growth. These results suggest that combination of indomethacin with chemotherapeutic drugs could be an effective form of cancer chemotherapy. The observed effects may be dependent on the expression of MCP-1.
Collapse
Affiliation(s)
- K Hattori
- Department of Fundamental Experimental Pharmaco-Science, Kyoritsu College of Pharmacy, Tokyo, Japan
| | | | | | | | | |
Collapse
|
37
|
Synthesis, crystal structures and spectroscopic studies of diorganotin derivatives with mefenamic acid. Crystal and molecular structures of 1,2:3,4-di-μ2-2-[(2,3-dimethylphenyl)amino]-benzoato-O,O-1,3-bis-2-[(-[(2,3-dimethylphenyl)amino]benzoato-O-1,2,4:2,3,4-di-μ3-oxo-tetrakis[di-methyltin(IV)] and 1,2:3,4-di-μ2-2-[(-[(-[(2,3-dimethylphenyl)amino]-benzoato-O,O-1,3-bis-2-[(-[(-[(2,3-dimethylphenyl)amino]benzoato-O-1,2,4:2,3,4-di-μ3-oxo-tetrakis[di-n-butyltin(IV)]. J Organomet Chem 2001. [DOI: 10.1016/s0022-328x(01)01026-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Gołab J, Kozar K, Kamiński R, Czajka A, Marczak M, Switaj T, Giermasz A, Stokłosa T, Lasek W, Zagozdzon R, Mucha K, Jakóbisiak M. Interleukin 12 and indomethacin exert a synergistic, angiogenesis-dependent antitumor activity in mice. Life Sci 2000; 66:1223-30. [PMID: 10737417 DOI: 10.1016/s0024-3205(00)00427-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Nonsteroidal anti-inflammatory drugs have been shown to reduce the incidence and mortality from colorectal cancer. It has recently been demonstrated that these drugs are capable of suppressing the production of pro-angiogenic factors from tumor cells. The mechanisms of antitumor action of interleukin 12 include the enforced secretion of anti-angiogenic factors and stimulation of antitumor immunity. Therefore, we hypothesized that the combination of a model nonsteroidal anti-inflammatory drug--indomethacin and interleukin 12--would result in enhanced angiogenesis-dependent antitumor effects against a colon-26 carcinoma cells transplanted into syngeneic mice. As expected the combined administration of both agents simultaneously resulted in a strengthened antitumor activity that was manifested as a retardation of tumor growth and prolongation of mouse survival. Importantly some mice were completely cured after the combined treatment. As administration of interleukin 12 and indomethacin resulted in enhanced inhibition of angiogenesis it seems possible that prevention of new blood vessel formation is one of the mechanisms responsible for the observed antitumor effects.
Collapse
Affiliation(s)
- J Gołab
- Department of Immunology, Institute of Biostructure, The Medical University of Warsaw, Poland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Verheul HM, Panigrahy D, Yuan J, D'Amato RJ. Combination oral antiangiogenic therapy with thalidomide and sulindac inhibits tumour growth in rabbits. Br J Cancer 1999; 79:114-8. [PMID: 10408702 PMCID: PMC2362163 DOI: 10.1038/sj.bjc.6690020] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neovascularization facilitates tumour growth and metastasis formation. In our laboratory, we attempt to identify clinically available oral efficacious drugs for antiangiogenic activity. Here, we report which non-steroidal anti-inflammatory drugs (NSAIDs) can inhibit corneal neovascularization, induced by basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF). This antiangiogenic activity may contribute to the known effects of NSAIDs on gastric ulcers, polyps and tumours. We found that sulindac was one of the most potent antiangiogenic NSAIDs, inhibiting bFGF-induced neovascularization by 50% and VEGF-induced neovascularization by 55%. Previously, we reported that thalidomide inhibited growth factor-induced corneal neovascularization. When we combined sulindac with thalidomide, we found a significantly increased inhibition of bFGF- or VEGF-induced corneal neovascularization (by 63% or 74% respectively) compared with either agent alone (P < 0.01). Because of this strong antiangiogenic effect, we tested the oral combination of thalidomide and sulindac for its ability to inhibit the growth of V2 carcinoma in rabbits. Oral treatment of thalidomide or sulindac alone inhibited tumour growth by 55% and 35% respectively. When given together, the growth of the V2 carcinoma was inhibited by 75%. Our results indicated that oral antiangiogenic combination therapy with thalidomide and sulindac may be a useful non-toxic treatment for cancer.
Collapse
Affiliation(s)
- H M Verheul
- Department of Surgery, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
40
|
Duffy CP, Elliott CJ, O'Connor RA, Heenan MM, Coyle S, Cleary IM, Kavanagh K, Verhaegen S, O'Loughlin CM, NicAmhlaoibh R, Clynes M. Enhancement of chemotherapeutic drug toxicity to human tumour cells in vitro by a subset of non-steroidal anti-inflammatory drugs (NSAIDs). Eur J Cancer 1998; 34:1250-9. [PMID: 9849488 DOI: 10.1016/s0959-8049(98)00045-8] [Citation(s) in RCA: 193] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The effect on cytotoxicity of combining a range of clinically important non-steroidal anti-inflammatory drugs (NSAIDs) with a variety of chemotherapeutic drugs was examined in the human lung cancer cell lines DLKP, A549, COR L23P and COR L23R and in a human leukaemia line HL60/ADR. A specific group of NSAIDs (indomethacin, sulindac, tolmetin, acemetacin, zomepirac and mefenamic acid) all at non-toxic levels, significantly increased the cytotoxicity of the anthracyclines (doxorubicin, daunorubicin and epirubicin), as well as teniposide, VP-16 and vincristine, but not the other vinca alkaloids vinblastine and vinorelbine. A substantial number of other anticancer drugs, including methotrexate, 5-fluorouracil, cytarabine, hydroxyurea, chlorambucil, cyclophosphamide, cisplatin, carboplatin, mitoxantrone, actinomycin D, bleomycin, paclitaxel and camptothecin, were also tested, but displayed no synergy in combination with the NSAIDs. The synergistic effect was concentration dependent. The effect appears to be independent of the cyclo-oxygenase inhibitory ability of the NSAIDs, as (i) the synergistic combination could not be reversed by the addition of prostaglandins D2 or E2; (ii) sulindac sulphone, a metabolite of sulindac that does not inhibit the cyclooxygenase enzyme, was positive in the combination assay: and (iii) many NSAIDs known to be cyclo-oxygenase inhibitors, e.g. meclofenamic acid, diclofenac, naproxen, fenoprofen, phenylbutazone, flufenamic acid, flurbiprofen, ibuprofen and ketoprofen, were inactive in the combination assay. The enhancement of cytotoxicity was observed in a range of drug sensitive tumour cell lines, but did not occur in P-170-overexpressing multidrug resistant cell lines. However, in the HL60/ADR and COR L23R cell lines, in which multidrug resistance is due to overexpression of the multidrug resistance-associated protein MRP, a significant increase in cytotoxicity was observed in the presence of the active NSAIDs. Subsequent Western blot analysis of the drug sensitive parental cell lines, DLKP and A549, revealed that they also expressed MRP and reverse-transcription-polymerase chain reaction studies demonstrated that mRNA for MRP was present in both cell lines. It was found that the positive NSAIDs were among the more potent inhibitors of [3H]-LTC4 transport into inside-out plasma membrane vesicles prepared from MRP-expressing cells, of doxorubicin efflux from preloaded cells and of glutathione-S-transferase activity. The NSAIDs did not enhance cellular sensitivity to radiation. The combination of specific NSAIDs with anticancer drugs reported here may have potential clinical applications, especially in the circumvention of MRP-mediated multidrug resistance.
Collapse
Affiliation(s)
- C P Duffy
- National Cell and Tissue Culture Centre, Dublin City University, Glasnevin, Ireland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kasahara K, Fujimura M, Bando T, Shibata K, Shirasaki H, Matsuda T. Modulation of sensitivity to cis-diamminedichloroplatinum (II) by thromboxane A2 receptor antagonists in non-small-cell lung cancer cell lines. Br J Cancer 1996; 74:1553-8. [PMID: 8932334 PMCID: PMC2074842 DOI: 10.1038/bjc.1996.588] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We examined the effect of selective thromboxane A2 (TXA2) receptor antagonists, calcium 5(Z)-1R, 2S, 3S, 4S-7-[3-phenylsulphonylaminobicyclo [2.2.1] hept-2-yl]-5-heptonoate hydrate (S-1452) and +/- -7-(3,5,6,-trimethyl-1,4-benzoquinon-2-yl)-7-phenylhaptanoic acid (AA-2414), on sensitivity to cis-diamminedichloroplatinum (II) (CDDP) in non-small-cell lung cancer cell lines. IC50 values to CDDP using MTT assay were decreased 2.1- and 4.6-fold respectively by treatment with 250 or 500 microM S-1452, for a 2 h simultaneous drug exposure, and those of PC-9/CDDP, a CDDP-resistant cell line, were decreased 3.1- and 6.1-fold. Sensitivity to carboplatin was also enhanced by the treatment with S-1452. IC50 values to CDDP and carboplatin were decreased by treatment with AA-2414 in a dose-dependent manner. Isobologram analysis showed that the combination of CDDP with S-1452 or AA-2414 produced supra-additive or additive effects in each cell line. Neither glutathione content nor glutathione S-transferase activity was changed in either cell line by treatment with 500 microM S-1452. Accumulation of platinum into PC-9 and PC-9/CDDP was increased by the treatment in a dose-dependent manner. Na+, K+-ATPase activity of PC-9 and PC-9/CDDP was enhanced by the treatment of S-1452 in a dose-dependent manner. These data show that the TXA2 receptor antagonists may enhance the sensitivity of non-small-cell lung cancer cell lines to platinum agents. Increase in Na+, K+-ATPase activity induced by S-1452 may be the mechanism of its sensitising effect through increase in platinum accumulation.
Collapse
Affiliation(s)
- K Kasahara
- The Third Department of Internal Medicine, Kanazawa University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Affiliation(s)
- G Ara
- Division of Cancer Pharmacology, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | | |
Collapse
|
43
|
Affiliation(s)
- B A Teicher
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
44
|
Huang P, Heimbrook DC. Monthly Update: Cyclooxygenase and 5–lipoxygenase inhibitors for the prevention and treatment of cancer: Oncologic, Endocrine & Metabolic. Expert Opin Investig Drugs 1995. [DOI: 10.1517/13543784.4.3.243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Teicher BA, Holden SA, Chen YN, Ara G, Korbut TT, Northey D. CAI: effects on cytotoxic therapies in vitro and in vivo. Cancer Chemother Pharmacol 1994; 34:515-21. [PMID: 7923563 DOI: 10.1007/bf00685664] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
CAI (NSC 609974; L651582), a new agent that has demonstrated antimetastatic activity in vitro and in vivo, was not very cytotoxic toward EMT-6 mouse mammary carcinoma cells in culture or toward FSaIIC fibrosarcoma cells in vivo. Coexposure of EMT-6 cells to CAI and antitumor alkylating agents under various environmental conditions did not markedly increase the cytotoxicity of cisplatin (CDDP), melphalan, or carmustine (BCNU). However, the combination of CAI and 4-hydroperoxycyclophosphamide (4-HC) produced much greater than additive killing of EMT-6 cells. CAI also increased the sensitivity of hypoxic EMT-6 cells to X-rays. CAI increased the cytotoxicity of cyclophosphamide toward FSaIIC tumor cells when animals were treated with single doses of both drugs. The effect of CAI on tumor cell killing by cyclophosphamide was greatest at high doses of the antitumor alkylating agent. CAI administration appeared to result in increased serum levels of prostaglandin E2 and leukotriene B4 in animals bearing the Lewis lung tumor. Administration of CAI on days 4-18 did not alter the growth of the Lewis lung carcinoma but did result in an increase in the tumor-growth delay produced by treatment with CDDP, cyclophosphamide, melphalan, BCNU, and fractionated radiation. Although CAI did not reduce the number of lung metastases present in Lewis lung carcinoma-bearing mice on day 20, it did appear to reduce the number of large (vascularized) metastases present on that day.
Collapse
Affiliation(s)
- B A Teicher
- Dana-Farber Cancer Institute, Boston, MA 02115
| | | | | | | | | | | |
Collapse
|