1
|
Lee KJ, Chang WCL, Chen X, Valiyaveettil J, Ramirez-Alcantara V, Gavin E, Musiyenko A, Madeira da Silva L, Annamdevula NS, Leavesley SJ, Ward A, Mattox T, Lindsey AS, Andrews J, Zhu B, Wood C, Neese A, Nguyen A, Berry K, Maxuitenko Y, Moyer MP, Nurmemmedov E, Gorman G, Coward L, Zhou G, Keeton AB, Cooper HS, Clapper ML, Piazza GA. Suppression of Colon Tumorigenesis in Mutant Apc Mice by a Novel PDE10 Inhibitor that Reduces Oncogenic β-Catenin. Cancer Prev Res (Phila) 2021; 14:995-1008. [PMID: 34584001 DOI: 10.1158/1940-6207.capr-21-0208] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/12/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022]
Abstract
Previous studies have reported that phosphodiesterase 10A (PDE10) is overexpressed in colon epithelium during early stages of colon tumorigenesis and essential for colon cancer cell growth. Here we describe a novel non-COX inhibitory derivative of the anti-inflammatory drug, sulindac, with selective PDE10 inhibitory activity, ADT 061. ADT 061 potently inhibited the growth of colon cancer cells expressing high levels of PDE10, but not normal colonocytes that do not express PDE10. The concentration range by which ADT 061 inhibited colon cancer cell growth was identical to concentrations that inhibit recombinant PDE10. ADT 061 inhibited PDE10 by a competitive mechanism and did not affect the activity of other PDE isozymes at concentrations that inhibit colon cancer cell growth. Treatment of colon cancer cells with ADT 061 activated cGMP/PKG signaling, induced phosphorylation of oncogenic β-catenin, inhibited Wnt-induced nuclear translocation of β-catenin, and suppressed TCF/LEF transcription at concentrations that inhibit cancer cell growth. Oral administration of ADT 061 resulted in high concentrations in the colon mucosa and significantly suppressed the formation of colon adenomas in the Apc+/min-FCCC mouse model of colorectal cancer without discernable toxicity. These results support the development of ADT 061 for the treatment or prevention of adenomas in individuals at risk of developing colorectal cancer. PREVENTION RELEVANCE: PDE10 is overexpressed in colon tumors whereby inhibition activates cGMP/PKG signaling and suppresses Wnt/β-catenin transcription to selectively induce apoptosis of colon cancer cells. ADT 061 is a novel PDE10 inhibitor that shows promising cancer chemopreventive activity and tolerance in a mouse model of colon cancer.
Collapse
Affiliation(s)
- Kevin J Lee
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Wen-Chi L Chang
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Xi Chen
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Jacob Valiyaveettil
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | | | - Elaine Gavin
- Gynecologic Oncology Research Division, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Alla Musiyenko
- Gynecologic Oncology Research Division, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Luciana Madeira da Silva
- Gynecologic Oncology Research Division, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Naga S Annamdevula
- Department of Chemical and Biomedical Engineering, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Silas J Leavesley
- Department of Chemical and Biomedical Engineering, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama.,Department of Pharmacology, University of South Alabama, Mobile, Alabama
| | - Antonio Ward
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Tyler Mattox
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Ashley S Lindsey
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Joel Andrews
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Bing Zhu
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Charles Wood
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Ashleigh Neese
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Ashley Nguyen
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Kristy Berry
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Yulia Maxuitenko
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | | | | | | | | | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | - Harry S Cooper
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Margie L Clapper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama.
| |
Collapse
|
2
|
Song B, Shu ZB, Du J, Ren JC, Feng Y. Anti-cancer effect of low dose of celecoxib may be associated with lnc-SCD-1:13 and lnc-PTMS-1:3 but not COX-2 in NCI-N87 cells. Oncol Lett 2017; 14:1775-1779. [PMID: 28789408 PMCID: PMC5529947 DOI: 10.3892/ol.2017.6316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 02/23/2017] [Indexed: 12/21/2022] Open
Abstract
In order to investigate the mechanism of celecoxib and whether long non-coding RNAs (lncRNAs) were involved in the effects of celecoxib treatment in NCI-N87 cells, NCI-N87 cells were treated with 15, 30 and 60 µM celecoxib and an MTT assay was performed to assess cell viability. Following treatment with 15 µM celecoxib, the cell cycle and apoptosis were analyzed by flow cytometry, and the mRNA levels of lnc-SCD-1:13, lnc-PTMS-1:3, cyclooxygenase-2 (COX-2), integrin α3 (ITGA3) and DSH homolog 1 (DVL1) were detected by reverse transcription quantitative PCR (RT-qPCR) in NCI-N87 cells. MTT analysis demonstrated that celecoxib significantly inhibited cell viability in treated cells compared with untreated cells. Flow cytometry analysis revealed that, compared with untreated cells, the percentage of cells in the G0/G1 phase was significantly increased, and the percentage of cells in the S and G2 phase was decreased. In addition, the percentage of early and late apoptotic cells was increased in cells treated with 15 µM celecoxib compared with the control. RT-qPCR analysis also demonstrated that the mRNA levels of lnc-SCD-1:13, lnc-PTMS-1:3, ITGA3 and DVL1 were increased following treatment with celecoxib (15 µM; P<0.05). However, there were no significant differences in the expression of COX-2 mRNA between cells treated with celecoxib (15 µM) and untreated cells. The present study demonstrated that a low dose of celecoxib may be involved in regulating cell growth independent of COX-2 in NCI-N87 cells. Furthermore, ITGA3 and/or DVL1 co-expressed with lnc-SCD-1:13 and lnc-PTMS-1:3 may be associated with the effects of treatment with a low dose of celecoxib in NCI-N87 cells.
Collapse
Affiliation(s)
- Bin Song
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhen-Bo Shu
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Juan Du
- Internal Medicine 2, The Tumor Hospital of Jilin, Changchun, Jilin 130012, P.R. China
| | - Ji-Chen Ren
- Internal Medicine 2, The Tumor Hospital of Jilin, Changchun, Jilin 130012, P.R. China
| | - Ye Feng
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
3
|
Abstract
Numerous epidemiologic studies have reported that the long-term use of nonsteroidal anti-inflammatory drugs (NSAID) is associated with a significant decrease in cancer incidence and delayed progression of malignant disease. The use of NSAIDs has also been linked with reduced risk from cancer-related mortality and distant metastasis. Certain prescription-strength NSAIDs, such as sulindac, have been shown to cause regression of precancerous lesions. Unfortunately, the extended use of NSAIDs for chemoprevention results in potentially fatal side effects related to their COX-inhibitory activity and suppression of prostaglandin synthesis. Although the basis for the tumor growth-inhibitory activity of NSAIDs likely involves multiple effects on tumor cells and their microenvironment, numerous investigators have concluded that the underlying mechanism is not completely explained by COX inhibition. It may therefore be possible to develop safer and more efficacious drugs by targeting such COX-independent mechanisms. NSAID derivatives or metabolites that lack COX-inhibitory activity, but retain or have improved anticancer activity, support this possibility. Experimental studies suggest that apoptosis induction and suppression of β-catenin-dependent transcription are important aspects of their antineoplastic activity. Studies show that the latter involves phosphodiesterase inhibition and the elevation of intracellular cyclic GMP levels. Here, we review the evidence for COX-independent mechanisms and discuss progress toward identifying alternative targets and developing NSAID derivatives that lack COX-inhibitory activity but have improved antineoplastic properties.
Collapse
Affiliation(s)
- Evrim Gurpinar
- Authors' Affiliations: Department of Pharmacology and Toxicology; Department of Pathology, The University of Alabama at Birmingham, Birmingham; and Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | | | | |
Collapse
|
4
|
The proapoptotic effect of traditional and novel nonsteroidal anti-inflammatory drugs in mammalian and yeast cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:504230. [PMID: 23983899 PMCID: PMC3747411 DOI: 10.1155/2013/504230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/08/2013] [Indexed: 12/16/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have long been used to treat pain, fever, and inflammation. However, mounting evidence shows that NSAIDs, such as aspirin, have very promising antineoplastic properties. The chemopreventive, antiproliferative behaviour of NSAIDs has been associated with both their inactivation of cyclooxygenases (COX) and their ability to induce apoptosis via pathways that are largely COX-independent. In this review, the various proapoptotic pathways induced by traditional and novel NSAIDs such as phospho-NSAIDs, hydrogen sulfide-releasing NSAIDs and nitric oxide-releasing NSAIDs in mammalian cell lines are discussed, as well as the proapoptotic effects of NSAIDs on budding yeast which retains the hallmarks of mammalian apoptosis. The significance of these mechanisms in terms of the role of NSAIDs in effective cancer prevention is considered.
Collapse
|
5
|
Whitt JD, Li N, Tinsley HN, Chen X, Zhang W, Li Y, Gary BD, Keeton AB, Xi Y, Abadi AH, Grizzle WE, Piazza GA. A novel sulindac derivative that potently suppresses colon tumor cell growth by inhibiting cGMP phosphodiesterase and β-catenin transcriptional activity. Cancer Prev Res (Phila) 2012; 5:822-33. [PMID: 22556201 DOI: 10.1158/1940-6207.capr-11-0559] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been widely reported to inhibit tumor growth by a COX-independent mechanism, although alternative targets have not been well defined or used to develop improved drugs for cancer chemoprevention. Here, we characterize a novel sulindac derivative referred to as sulindac benzylamine (SBA) that does not inhibit COX-1 or COX-2, yet potently inhibits the growth and induces the apoptosis of human colon tumor cells. The basis for this activity appears to involve cyclic guanosine 3',5',-monophosphate phosphodiesterase (cGMP PDE) inhibition as evident by its ability to inhibit cGMP hydrolysis in colon tumor cell lysates and purified cGMP-specific PDE5, increase intracellular cGMP levels, and activate cGMP-dependent protein kinase G at concentrations that suppress tumor cell growth. PDE5 was found to be essential for colon tumor cell growth as determined by siRNA knockdown studies, elevated in colon tumor cells as compared with normal colonocytes, and associated with the tumor selectivity of SBA. SBA activation of PKG may suppress the oncogenic activity of β-catenin as evident by its ability to reduce β-catenin nuclear levels, Tcf (T-cell factor) transcriptional activity, and survivin levels. These events preceded apoptosis induction and appear to result from a rapid elevation of intracellular cGMP levels following cGMP PDE inhibition. We conclude that PDE5 and possibly other cGMP degrading isozymes can be targeted to develop safer and more efficacious NSAID derivatives for colorectal cancer chemoprevention.
Collapse
Affiliation(s)
- Jason D Whitt
- Department of Pharmacology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Martin JE, Le Leu RK, Hu Y, Young GP. R-flurbiprofen suppresses distal nonmucin-producing colorectal tumors in azoxymethane-treated rats, without suppressing eicosanoid production. Am J Physiol Gastrointest Liver Physiol 2010; 298:G860-4. [PMID: 20338924 DOI: 10.1152/ajpgi.00330.2009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Toxicity and gastrointestinal side effects limits the use of nonsteroidal anti-inflammatory drugs (NSAIDs) as agents to prevent colorectal cancer. These undesirable effects appear to be related to the inhibition of cyclooxygenase-associated pathways. Using the azoxymethane (AOM)-rat model of carcinogenesis, we aimed to test the potency of a low-toxicity R-flurbiprofen and whether NSAIDs have differing effects on regional tumor subtypes. Groups of 50 rats were gavaged 6 days a week with drug. After 1 and 2 wk on drug, rats were given intraperitoneal injection of AOM (15 mg/kg body wt). Groups were controls, sulindac (nonselective cyclooxygenase inhibitor) 5 and 20 mg/kg body wt per day, and R-flurbiprofen 30 mg/kg body wt per day. Tumor location, size, and histological subtype (either mucinous or nonmucinous) were recorded after 30 wk. The incidence of colon tumors was significantly reduced in the sulindac 20 mg (P < 0.001) and the R-flurbiprofen groups (P < 0.03) compared with the control group. The sulindac 20 mg and R-flurbiprofen groups also showed a reduced number of distal colon tumors (P < 0.03), whereas proximal tumors were not affected. Tumors only of the nonmucinous subtype were significantly reduced with the sulindac 20 mg and R-flurbiprofen groups (P < 0.001). Tumor size was not significantly different between all groups. Only the sulindac 20 mg group showed a reduced colonic prostaglandin E2 concentration. The sulindac groups showed a dose-dependant reduction in body weight gain (P < 0.001). In conclusion, R-flurbiprofen at a dose of 30 mg/kg body wt per day was well tolerated by the animals and, along with sulindac at 30 mg/day body wt, showed protection against the development of colon cancer in the rat-AOM model.
Collapse
Affiliation(s)
- Jonathan E Martin
- Flinders Ctr. for Cancer Prevention and Control, Flinders Univ., South Australia, 5042
| | | | | | | |
Collapse
|
7
|
Kim N, Kim CH, Ahn DW, Lee KS, Cho SJ, Park JH, Lee MK, Kim JS, Jung HC, Song IS. Anti-gastric cancer effects of celecoxib, a selective COX-2 inhibitor, through inhibition of Akt signaling. J Gastroenterol Hepatol 2009; 24:480-7. [PMID: 18823436 DOI: 10.1111/j.1440-1746.2008.05599.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM Previously, we showed that treatment with celecoxib significantly reduced the number of viable gastric cancer cells, in a dose- and time-dependent manner. However, the specific anti-cancer effects of celecoxib on gastric cancer cells have not been clarified. The present in vitro study was carried out to investigate the mechanism involved in the anti-gastric cancer effects of celecoxib. METHODS 3-(4,5-Dimethyl-2 thiazoyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was carried out after treating AGS cells (human gastric cancer cell line, ATCC CRL 1739) with celecoxib or indomethacin, and the effect of prostaglandin E(2) or LY294002 (PI3K inhibitor) was evaluated. Western blot analysis of tAkt (total Akt), pAkt (phosphorylated Akt), pGSK3beta (phosphorylated glycogen synthase kinase-3beta), pFKHR (phosphorylated forkhead transcriptional factor), and caspase-9 was carried out at various concentrations (0, 5, 10, 25, or 50 micromol/L) of celecoxib or indomethacin-treatment for 24 or 48 h in AGS cells. RESULTS Celecoxib- or LY294002-induced cell death was found to occur in a dose-dependent manner in AGS cells, and these decreases were slightly recovered by the addition of PGE(2) (25 or 50 micromol/L). The expression of pAkt but not tAkt was lower in the celecoxib treated-AGS cells and the response was dose dependent (P < 0.05). The expression of pGSK3beta and pFKHR was also significantly decreased in the celecoxib treated-AGS cells. Procaspase 9 (47 kDa) was frequently cleaved into 37, 35 and 17 kDa fragments in the celecoxib-treatment group. However, these changes in cell signal transduction were not observed in the indomethacin treated-AGS cells. CONCLUSION The anti-cancer effects of celecoxib on gastric cancer cells might be partly mediated by downregulation of Akt, GSK3beta, FKHR, and upregulation of caspase-9, in the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Nayoung Kim
- Department of Internal Medicine, Seoul National University, Bundang Hospital, Seongnam, Gyeonggi-do, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Cho SJ, Kim N, Kim JS, Jung HC, Song IS. The anti-cancer effect of COX-2 inhibitors on gastric cancer cells. Dig Dis Sci 2007; 52:1713-21. [PMID: 17393325 DOI: 10.1007/s10620-007-9787-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 01/18/2007] [Indexed: 12/21/2022]
Abstract
Epidemiologic studies have shown that nonsteroidal anti-inflammatory drugs could reduce the risk of cancer development including gastric cancer. This study was performed to identify the antineoplastic mechanism in gastric cancer cells affected by celecoxib, a selective COX-2 inhibitor. MTT assay, ELISA for prostaglandin E(2) (PGE(2)), cell-cycle analyses, immunofluorescent staining, and flow cytometry were performed after treating human gastric cancer cell lines (AGS and MKN-45) with celecoxib or indomethacin. The viabilities of celecoxib-treated cells decreased in a dose- and time-dependent manner compared with indomethacin. Drop of PGE(2) levels was more prominent in the presence of indomethacin than in that of celecoxib. Celecoxib arrested the cell cycle in the G(0)-G(1) phase, which reduced cell numbers in the S phase. Moreover, celecoxib increased the apoptotic cell proportions, a 4-fold increase over control cells. The anticancer effects of celecoxib on gastric cancer cells appear to be mediated by cell-cycle arrest and apoptosis, and not by COX-2 or PGE(2) suppression alone.
Collapse
Affiliation(s)
- Soo-Jeong Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | | | | |
Collapse
|
9
|
Bos CL, Kodach LL, van den Brink GR, Diks SH, van Santen MM, Richel DJ, Peppelenbosch MP, Hardwick JCH. Effect of aspirin on the Wnt/beta-catenin pathway is mediated via protein phosphatase 2A. Oncogene 2006; 25:6447-56. [PMID: 16878161 DOI: 10.1038/sj.onc.1209658] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nonsteroidal anti-inflammatory drugs show chemopreventive efficacy in colon cancer, but the mechanism behind this remains unclear. Elucidating this mechanism is seen as vital to the development of new chemopreventive agents. We studied the effects of aspirin on the oncogenic Wnt/beta-catenin pathway activity in colorectal cancer cell lines and observed that aspirin dose-dependently decreased the activity of this pathway, as judged by TCF-driven luciferase activity, reduced Wnt target gene expression and increased phosphorylation of beta-catenin by immunoblotting. Furthermore, the ubiquitination and cytoplasmic levels of beta-catenin were assessed by immunoblotting, and also the localization of beta-catenin was shown by green fluorescent protein-tagged beta-catenin and time-lapse fluorescent imaging. Importantly, aspirin treatment caused increased phosphorylation of protein phosphatase 2A (PP2A), an event associated with inhibition of PP2A enzymatic activity, which was confirmed by a reduction in enzymatic PP2A activity. Moreover, this inhibition of PP2A enzymatic activity was essential for the effects of aspirin on the Wnt/beta-catenin pathway as shown by transient transfection with PP2A constructs. The findings in this article provide a molecular explanation for the efficacy of aspirin in chemoprevention of colorectal cancer and shows biochemical evidence that PP2A is an important regulator of Wnt/beta-catenin pathway activity in these cells.
Collapse
Affiliation(s)
- C L Bos
- Laboratory of Experimental Oncology and Radiobiology, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Din FVN, Stark LA, Dunlop MG. Aspirin-induced nuclear translocation of NFkappaB and apoptosis in colorectal cancer is independent of p53 status and DNA mismatch repair proficiency. Br J Cancer 2005; 92:1137-43. [PMID: 15770215 PMCID: PMC2361954 DOI: 10.1038/sj.bjc.6602455] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Substantial evidence indicates nonsteroidal anti-inflammatory drugs (NSAIDs) protect against colorectal cancer (CRC). However, the molecular basis for this anti-tumour activity has not been fully elucidated. We previously reported that aspirin induces signal-specific IκBα degradation followed by NFκB nuclear translocation in CRC cells, and that this mechanism contributes substantially to aspirin-induced apoptosis. We have also reported the relative specificity of this aspirin-induced NFκB-dependent apoptotic effect for CRC cells, in comparison to other cancer cell types. It is now important to establish whether there is heterogeneity within CRC, with respect to the effects of aspirin on the NFκB pathway and apoptosis. p53 signalling and DNA mismatch repair (MMR) are known to be deranged in CRC and have been reported as potential molecular targets for the anti-tumour activity of NSAIDs. Furthermore, both p53 and MMR dysfunction have been shown to confer resistance to chemotherapeutic agents. Here, we set out to determine the p53 and hMLH1 dependency of the effects of aspirin on NFκB signalling and apoptosis in CRC. We specifically compared the effects of aspirin treatment on cell viability, apoptosis and NFκB signalling in an HCT-116 CRC cell line with the p53 gene homozygously disrupted (HCT-116p53−/−) and an HCT-116 cell line rendered MMR proficient by chromosomal transfer (HCT-116+ch3), to the parental HCT-116 CRC cell line. We found that aspirin treatment induced apoptosis following IκBα degradation, NFκB nuclear translocation and repression of NFκB-driven transcription, irrespective of p53 and DNA MMR status. These findings are relevant for design of both novel chemopreventative agents and chemoprevention trials in CRC.
Collapse
Affiliation(s)
- F V N Din
- Department of Oncology, Division of Clinical and Molecular Medicine and MRC Human Genetics Unit, Western General Hospital, University of Edinburgh, Crewe Rd, Edinburgh EH4 2XU, Scotland, UK
| | | | | |
Collapse
|
11
|
Yu J, Tang BD, Leung WK, To KF, Bai AH, Zeng ZR, Ma PK, Go MY, Hu PJ, Sung JJ. Different cell kinetic changes in rat stomach cancer after treatment with celecoxib or indomethacin: Implications on chemoprevention. World J Gastroenterol 2005; 11:41-5. [PMID: 15609394 PMCID: PMC4205381 DOI: 10.3748/wjg.v11.i1.41] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Mechanisms underlying the chemopreventive effects of cyclooxygenase (COX) inhibitors remain elusive. We have previously shown that celecoxib but not indomethacin could prevent carcinogen-induced gastric cancer development in Wistar rats. This chemopreventive effect appeared to be independent of COX-2 and prostaglandin (PG) E2 suppression since the lowest PGE2 was obtained in indomethacin group. This study compared the cell kinetic changes in stomachs of rats after treatment with celecoxib (5, 10, 20 mg/(kg·d)) or indomethacin (3 mg/(kg·d)) to gain more insights into the chemopreventive mechanism.
METHODS: The apoptosis and proliferation indexes in gastric tumor, adjacent non-cancer tissues and normal gastric tissues were determined. Apoptosis was quantified by apoptotic nuclei counting and TUNEL, whereas proliferation was determined by Ki67 immunostaining.
RESULTS: Treatment with either celecoxib or indomethacin inhibited gastric tumor proliferation by more than 65% (P<0.02). However, celecoxib caused a dose-dependent increase in apoptosis (P<0.05) which was not seen in indomethacin-treated tumors (P = 0.54). The highest apoptosis to proliferation ratio was seen in tumors treated with celecoxib at 10 mg/(kg·d). Treatment with this dose of celecoxib was associated with the lowest incidence of gastric cancer development.
CONCLUSION: Our findings suggest that the difference in chemopreventive effects of indomethacin and celecoxib in this animal model of gastric carcinogenesis is largely due to the differential cell kinetic changes, which does not correlate with the degree of COX-2 and PG suppression.
Collapse
Affiliation(s)
- Jun Yu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Perse M, Zebic A, Cerar A. Rofecoxib does not inhibit aberrant crypt foci formation but inhibits later steps in the development of experimental colorectal cancer: rofecoxib in experimental colon cancer. Scand J Gastroenterol 2005; 40:61-7. [PMID: 15841716 DOI: 10.1080/00365520410009519] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Cyclooxygenase-2 (COX-2) has been shown to have an important role in carcinogenesis. Elevated COX-2 expression has been reported in several human tumours, including colorectal cancer (CRC) and appears to correlate with survival inversely. Regular use of non-steroidal anti-inflammatory drugs (NSAIDs) can decrease the incidence of CRC, but prolonged administration may cause side effects. Selective COX-2 inhibitors have comparable effects to NSAIDs with reduced side effects. The aim of the present study was to analyse the potential therapeutic role of rofecoxib, a selective COX-2 inhibitor, in experimentally induced rat colorectal tumours. MATERIAL AND METHODS Sixty-three Wistar male rats (33 in the experimental group and 30 in the control group) received subcutaneous injections of 1,2-dimethylhydrazine (DMH). Synchronal with the first injection of DMH in the experimental group, rats were given rofecoxib orally for 6 months, when autopsy was done. Colorectal tumours were evaluated quantitatively and histopathologically for the presence of aberrant crypt foci (ACF), adenomas and adenocarcinomas. RESULTS No statistically significant differences were found in the number of dysplastic ACF between the experimental and control groups (p > 0.05). However, a significant lower incidence of adenomas (p < 0.05), adenocarcinomas (p < 0.05) and decreased volume of macroscopically visible tumours (by 42%) was found in the experimental group. Furthermore, no significant differences were evaluated between the groups according to the degree of dysplasia and Dukes stage. In the experimental group, chronic ulcerations were found in the upper gastrointestinal tract in 9% of the rats. CONCLUSION Our data suggest that rofecoxib effectively inhibits tumour growth and progression but not tumour initiation.
Collapse
Affiliation(s)
- Martina Perse
- Institute of Pathology, Medical Experimental Centre, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.
| | | | | |
Collapse
|
13
|
Mori H, Yamada Y, Kuno T, Hirose Y. Aberrant crypt foci and beta-catenin accumulated crypts; significance and roles for colorectal carcinogenesis. Mutat Res 2004; 566:191-208. [PMID: 15082237 DOI: 10.1016/j.mrrev.2003.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2002] [Revised: 08/19/2003] [Accepted: 08/21/2003] [Indexed: 12/23/2022]
Abstract
Preneoplastic or precancerous lesions in the large bowel have been characterized in terms of morphology and histochemical phenotype. However, the detailed histogenesis and relation of particular lesions to malignancies has not yet to be unequivocally clarified. Aberrant crypt foci (ACF), identified in whole-mount preparations of colonic mucosa in rodents and also recognized in human colon, are now frequently used as effective surrogate biomarkers for experimentally detection of chemopreventive agents against colorectal cancers, but the preneoplastic or precancerous nature of ACF in rodents and humans still remains inconclusive. Relatively recently, early appearing beta-catenin accumulated crypts (BCAC) have been described in en face preparations of colonic mucosa in rodents which differ from ACF in many features. BCAC are suggested to be premalignant rather than preneoplastic. The pathological significance of both lesions, including their advantages and disadvantages as surrogate end points for large bowel neoplasms, and roles in colorectal carcinogenesis are discussed here.
Collapse
Affiliation(s)
- Hideki Mori
- Department of Tumor Pathology, Gifu University School of Medicine, 40 Tsukasa-machi, Gifu 500-8705, Japan.
| | | | | | | |
Collapse
|
14
|
Babbar N, Ignatenko NA, Casero RA, Gerner EW. Cyclooxygenase-independent induction of apoptosis by sulindac sulfone is mediated by polyamines in colon cancer. J Biol Chem 2003; 278:47762-75. [PMID: 14506281 DOI: 10.1074/jbc.m307265200] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sulindac, a non-steroidal anti-inflammatory prodrug, is metabolized into pharmacologically active sulfide and sulfone derivatives. Sulindac sulfide, but not sulindac sulfone, inhibits cyclooxygenase (COX) enzyme activities, yet both derivatives have growth inhibitory effects on colon cancer cells. Microarray analysis was used to detect COX-independent effects of sulindac on gene expression in human colorectal cells. Spermidine/sperm-ine N1-acetyltransferase (SSAT) gene, which encodes a polyamine catabolic enzyme, was induced by clinically relevant sulindac sulfone concentrations. Northern blots confirmed increased SSAT RNA levels in these colon cancer cells. Deletion analysis and mutational studies were done to map the sulindac sulfone-dependent response sequences in the SSAT 5'-flanking sequences. This led us to the identification of two peroxisome proliferator-activated receptor (PPAR) response elements (PPREs) in the SSAT gene. PPRE-2, at +48 bases relative to the transcription start site, is required for the induction of SSAT by sulindac sulfone and is specifically bound by PPAR gamma in the Caco-2 cells as shown by transfection and gel shift experiments. PPRE-1, at-323 bases relative to the start site, is not required for the induction of SSAT by sulindac sulfone but can be bound by both PPAR delta and PPAR gamma. Sulindac sulfone reduced cellular polyamine contents in the absence but not in the presence of verapamil, an inhibitor of the export of monoacetyl diamines, inhibited cell proliferation and induced apoptosis. The induced apoptosis could be partially rescued by exogenous putrescine. These data suggest that apoptosis induced by sulindac sulfone is mediated, in part, by the COX-independent, PPAR-dependent transcriptional activation of SSAT, leading to reduced tissue polyamine contents in human colon cancer cells.
Collapse
MESH Headings
- Acetyltransferases/genetics
- Apoptosis
- Blotting, Northern
- Blotting, Western
- Caco-2 Cells
- Cell Membrane/metabolism
- Cell Survival
- Colonic Neoplasms/metabolism
- Cyclooxygenase 2
- DNA, Complementary/metabolism
- Dose-Response Relationship, Drug
- Humans
- Immunoblotting
- Isoenzymes/metabolism
- Membrane Proteins
- Models, Biological
- Models, Genetic
- Oligonucleotide Array Sequence Analysis
- Oligonucleotides/chemistry
- Plasmids/metabolism
- Polyamines/chemistry
- Promoter Regions, Genetic
- Prostaglandin-Endoperoxide Synthases/metabolism
- Protein Binding
- Protein Biosynthesis
- Putrescine/chemistry
- RNA/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Response Elements
- Spermidine/metabolism
- Sulindac/analogs & derivatives
- Sulindac/pharmacology
- Time Factors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Naveen Babbar
- Arizona Cancer Center, and Biochemistry, Molecular and Cellular Biology Graduate Program, The University of Arizona, Tucson, 85724, USA
| | | | | | | |
Collapse
|
15
|
Nikitakis NG, Hebert C, Lopes MA, Reynolds MA, Sauk JJ. PPARgamma-mediated antineoplastic effect of NSAID sulindac on human oral squamous carcinoma cells. Int J Cancer 2002; 98:817-23. [PMID: 11948457 DOI: 10.1002/ijc.10278] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is strong evidence that nonsteroidal antiinflammatory drug (NSAID) sulindac may exert a significant antineoplastic effect. The purpose of our study was to explore the effects of sulindac on human oral squamous cell carcinoma (SCCa) cells and to elucidate the underlying molecular mechanisms. The changes that sulindac treatment induced on growth, apoptosis and cell cycle distribution of human oral SCCa cell lines were assessed by cell growth and flow cytometry experiments. Utilizing quantitative RT-PCR and immunocytochemistry, we determined the effect of sulindac treatment on mRNA and protein expression of different sulindac's targets. Also, PPARgamma expression was selectively targeted by antisense oligonucleotide treatment. Both sulfide and sulfone metabolites of sulindac, which differ in the ability to cause COX-2 inhibition, induced a significant dose- and time-dependent cell growth reduction accompanied by increase in apoptosis without concomitant cell cycle arrest. Sulindac treatment also caused upregulation of the protein and mRNA expression levels of COX-2 and PPARs. Treatment with antisense PPARgamma oligonucleotides abolished sulindac's growth inhibitory effect. Our results are consistent with a significant growth inhibitory effect of NSAID sulindac on human oral SCCa cells, which is mediated, at least partially, through induction of apoptosis. We suggest that upregulation of PPARgamma expression and activation may be, at least partially, responsible for sulindac's antiproliferative effect.
Collapse
Affiliation(s)
- Nikolaos G Nikitakis
- Department of Diagnostic Sciences and Pathology, University of Maryland, Baltimore, MD 21201-1586, USA.
| | | | | | | | | |
Collapse
|
16
|
Brown WA, Skinner SA, Malcontenti-Wilson C, Vogiagis D, O'Brien PE. Non-steroidal anti-inflammatory drugs with activity against either cyclooxygenase 1 or cyclooxygenase 2 inhibit colorectal cancer in a DMH rodent model by inducing apoptosis and inhibiting cell proliferation. Gut 2001; 48:660-6. [PMID: 11302965 PMCID: PMC1728280 DOI: 10.1136/gut.48.5.660] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Standard non-steroidal anti-inflammatory drugs (NSAIDs) reduce the risk of colorectal cancer by 40-60% but the mechanism by which this occurs is uncertain. Selective cyclooxygenase 2 inhibitors are potentially ideal chemopreventive agents as they are less toxic than standard NSAIDs. No study has compared the efficacy of these drugs at clinically relevant doses in a tumour model. AIMS To assess the efficacy of a range of NSAIDs with varying activity against the two cyclooxygenase isoforms in a rodent colorectal carcinogen model at anti-inflammatory doses and to explore the effect of NSAIDs on the rate of tumour apoptosis and proliferation. METHODS Colorectal tumours were induced in six week old Sprague-Dawley rats with five weekly doses of 1,2 dimethylhydrazine. Test agents were: indomethacin 2 mg/kg/day, meloxicam 0.6 mg/kg/day, celecoxib 6 mg/kg/day, and sulindac sulphone 40 mg/kg/day. Sulindac was tested at its chemoprotective dose of 20 mg/kg/day. After 23 weeks the number and volume of tumours per animal were recorded. Histology was performed. Tumour apoptosis was quantified on haematoxylin-eosin sections. Tumour proliferation was quantified using an immunohistochemical stain for bromodexoyuridine incorporation. RESULTS Test agents effectively reduced the number and volume of tumours developing in the treatment period. In all groups there was an increase in the rate of tumour apoptosis and a reduced rate of proliferation. CONCLUSIONS These data suggest that the chemopreventive effect of NSAIDs is independent of their cyclooxygenase inhibitory profile. One potential mechanism for their action may be through induction of apoptosis and inhibition of proliferation.
Collapse
Affiliation(s)
- W A Brown
- Monash University Department of Surgery, Alfred Hospital, Melbourne, Australia.
| | | | | | | | | |
Collapse
|
17
|
Hardwick JC, van den Brink GR, Offerhaus GJ, van Deventer SJ, Peppelenbosch MP. NF-kappaB, p38 MAPK and JNK are highly expressed and active in the stroma of human colonic adenomatous polyps. Oncogene 2001; 20:819-27. [PMID: 11314016 DOI: 10.1038/sj.onc.1204162] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2000] [Revised: 11/30/2000] [Accepted: 12/05/2000] [Indexed: 12/11/2022]
Abstract
The factors that govern the progression from colonic adenomatous polyp to colon cancer are poorly understood. The observation that NSAIDs act as chemopreventative agents and reduce the size of colonic polyps suggests the involvement of inflammatory signalling, but inflammatory signalling in colonic polyps has not been studied. We investigated the expression of the active forms of NF-kappaB, JNK and p38 MAPK using immunohistochemistry with activation specific antibodies in human colonic adenomas. We show that active NF-kappaB is seen in stromal macrophages that also express COX-2 and TNF-alpha, active JNK is seen in stromal and intraepithelial T-lymphocytes and periendothelial cells of new blood vessels, and active p38 MAPK is most highly expressed in macrophages and other stromal cells. These results demonstrate the presence of active inflammatory signal transduction in colonic polyps and that these are predominantly in the stroma. In the case of NF-kappaB this coincides with the cellular localisation of COX-2. These results support evidence that NSAIDs may act through effects on stromal cells rather than epithelial cells.
Collapse
Affiliation(s)
- J C Hardwick
- Department of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
18
|
Brown WA, Skinner SA, Malcontenti-Wilson C, Misajon A, DeJong T, Vogiagis D, O'Brien PE. Non-steroidal anti-inflammatory drugs with different cyclooxygenase inhibitory profiles that prevent aberrant crypt foci formation but vary in acute gastrotoxicity in a rat model. J Gastroenterol Hepatol 2000; 15:1386-92. [PMID: 11197048 DOI: 10.1046/j.1440-1746.2000.02370.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Standard non-steroidal anti-inflammatory drugs (NSAIDs) reduce the risk of colorectal cancer; however, their use as preventive agents is limited by their inherent toxicity. Drugs that selectively inhibit cyclooxygenase-2 (COX-2) may be useful in this setting as they are supposedly less toxic. No study has directly compared the ability of standard NSAIDs and selective COX-2 inhibitors to inhibit colorectal cancer at clinically relevant doses. METHODS Aberrant crypt foci (ACF) were induced in Sprague-Dawley rats by using 1,2-dimethylhydrazine (DMH). Test agents or vehicle were then administered for 3 weeks, twice daily through orogastric gavage. At the end of this period, the number and multiplicity of ACF were determined. The agents tested at equivalent anti-inflammatory doses were: sulindac and indomethacin (standard NSAIDs), meloxicam (selective COX-2 inhibitor), celecoxib (specific COX-2 inhibitor) and sulindac sulfone (no known COX activity). Acute gastrotoxicity of NSAID in rats was compared by using quantitative histology. RESULTS All test agents reduced the number of ACF. There was a 42% reduction with indomethacin, 46% with sulindac, 46% with meloxicam, 22% with celecoxib and 36% with sulindac sulfone. Only the COX-2 inhibitors caused no significant gastrotoxicity in rats. CONCLUSIONS Cyclooxygenase-2 inhibitors are potentially ideal chemopreventive agents as they inhibit ACF and are not gastrotoxic.
Collapse
Affiliation(s)
- W A Brown
- Monash University Department of Surgery, Alfred Hospital, Commercial Road, Prahran, Victoria 3181, Australia.
| | | | | | | | | | | | | |
Collapse
|
19
|
Tonelli F, Valanzano R, Messerini L, Ficari F. Long-term treatment with sulindac in familial adenomatous polyposis: is there an actual efficacy in prevention of rectal cancer? J Surg Oncol 2000; 74:15-20. [PMID: 10861602 DOI: 10.1002/1096-9098(200005)74:1<15::aid-jso4>3.0.co;2-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND OBJECTIVES Ileorectal anastomosis (IRA) is still used in the treatment of familial adenomatous polyposis (FAP). Sulindac appears to induce regression of colorectal adenomas; however, its effects in long-term therapy and in preventing carcinoma remain unclear. METHODS Fifteen FAP patients treated by IRA received sulindac (200 mg/day) for a mean period of 48.6 +/- 28.7 (range 12-124) months. Number, size, and type of rectal polyps were assessed by endoscopic and histological evaluation every 6 months. RESULTS Significant regression of polyps was observed in all patients after 6 months (P < 0.02). However, after a mean of 48.6 +/- 28.7 months, both number and size of polyps increased again, showing no statistical difference with baseline values. Minute polyps appeared reddish, while the largest lesions were flat or slightly elevated. Endoscopic polypectomy was necessary in 9 patients and transanal surgical excision in 3. Two patients were submitted to restorative proctectomy because of a large polyp with severe dysplasia and a rectal cancer, respectively. CONCLUSIONS Sulindac appears to influence the morphological appearance of polyps in FAP patients, inducing apparent regression. However, at a dose of 200 mg, it does not influence the progression of polyps toward a malignant pattern.
Collapse
Affiliation(s)
- F Tonelli
- Department of Clinical Physiopathology, Surgical Unit, University of Florence, Italy.
| | | | | | | |
Collapse
|
20
|
Affiliation(s)
- K Krishnan
- Department of Internal Medicine, East Tennessee State University, Johnson City 37614-70622, USA
| | | | | |
Collapse
|
21
|
de Jong TA, Skinner SA, Malcontenti-Wilson C, Vogiagis D, Bailey M, van Driel IR, O'Brien PE. Inhibition of rat colon tumors by sulindac and sulindac sulfone is independent of K-ras (codon 12) mutation. Am J Physiol Gastrointest Liver Physiol 2000; 278:G266-72. [PMID: 10666051 DOI: 10.1152/ajpgi.2000.278.2.g266] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonsteroidal anti-inflammatory drug (NSAID) use reduces the risk of colorectal cancer by 40-50%. Previous studies suggest that effective inhibition of colorectal cancer by NSAIDs may be dependent on the presence or absence of a K-ras mutation. This study was aimed at determining the relationship between inhibition of colorectal cancer by sulindac and sulindac sulfone and the presence of activating K-ras mutations in the 1,2-dimethylhydrazine dihydrochloride rat model. Sulindac (20 mg x kg(-1) x day(-1)), sulindac sulfone (40 mg x kg(-1) x day(-1)), or vehicle was administered orally to male Sprague-Dawley rats for a 4-wk period beginning 20 wk after tumor induction. Tumor number and volume were measured before treatment by laparotomy and colonoscopy and again after treatment. Sulindac and sulindac sulfone treatment significantly reduced the number and volume of colorectal tumors compared with control rats. For K-ras (codon 12) mutation detection, frozen tumor tissue was collected at the endpoint. We found K-ras codon 12 mutations in 11 of 21 (52%) control tumors. The proportion of tumors with K-ras mutations in the sulindac-treated group [5 of 8 (62%); odds ratio = 1.51 (95% confidence interval = 0.29, 8.33)] and the proportion of sulindac sulfone-treated tumors [9 of 14 (64%); odds ratio = 1.63 (95% confidence interval = 0.41, 6.66)] were not significantly different from controls. Tumor inhibition did not correlate with K-ras (codon 12) mutation status, which suggests that the mechanism of inhibition of rat colorectal cancer by sulindac and sulindac sulfone is independent of K-ras mutation.
Collapse
Affiliation(s)
- T A de Jong
- Department of Surgery, Monash University Medical School, Prahran, Melbourne 3181, Australia
| | | | | | | | | | | | | |
Collapse
|
22
|
Yang VW, Geiman DE, Hubbard WC, Spannhake EW, Hylind LM, Hamilton SR, Giardiello FM. Tissue prostanoids as biomarkers for chemoprevention of colorectal neoplasia: correlation between prostanoid synthesis and clinical response in familial adenomatous polyposis. Prostaglandins Other Lipid Mediat 2000; 60:83-96. [PMID: 10680778 PMCID: PMC2714803 DOI: 10.1016/s0090-6980(99)00054-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent studies indicate that sulindac, a nonsteroidal anti-inflammatory drug (NSAID), lowers mucosal prostanoid levels and regresses colorectal adenomas in patients with familial adenomatous polyposis (FAP). To determine whether they are biomarkers for sulindac-mediated chemoprevention of colorectal adenomas, levels of 5 prostanoids [prostaglandin (PG) D2, PGE2, PGF2alpha, thromboxane B2, and 6-keto-PGF1alpha] in the normal-appearing rectal mucosa from 7 FAP patients with a history of subtotal colectomy and ileorectal anastomosis and 4 FAP patients without surgery, were measured in the absence or presence of exogenously added arachidonic acid before the initiation and at the end of 3 months of sulindac treatment. The addition of arachidonic acid resulted in a uniform increase in the levels of all 5 prostanoids although this increase was selectively attenuated in patients with ileorectal anastomosis who took sulindac. In the latter patients, arachidonic acid also augmented the inhibition of prostanoid synthesis by sulindac. In contrast, sulindac failed to attenuate the increase in prostanoid levels resulting from arachidonic acid in patients without previous surgery. Importantly, when measured in the presence of arachidonic acid, the reduction in the levels of all 5 prostanoids due to sulindac was statistically correlated with a reduction in the size and number of adenomas in the two groups of patients combined. These results suggest that tissue prostanoids measured in the presence of arachidonic acid may serve as sensitive and reliable biomarkers in monitoring the clinical responsiveness of FAP patients undergoing chemoprevention for colorectal neoplasia with NSAIDs.
Collapse
Affiliation(s)
- V W Yang
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Matsuhashi N, Nakajima A, Shinohara K, Oka T, Yazaki Y. Rectal cancer after sulindac therapy for a sporadic adenomatous colonic polyp. Am J Gastroenterol 1998; 93:2261-6. [PMID: 9820411 DOI: 10.1111/j.1572-0241.1998.00630.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Like adenomatous polyps in familial adenomatous polyposis, some sporadic colorectal polyps have been reported to regress in response to sulindac administration. However, a rapidly growing invasive rectal cancer developed in one of 15 patients with sulindac-treated sporadic adenomatous colorectal polyps 16 months after sulindac treatment. In this patient, both the adenomatous polyp that responded partially to sulindac and the rectal cancer developing after sulindac therapy showed immunostaining for cyclooxygenase-2. Although short term sulindac therapy seems to be able to cause some adenomatous colorectal polyps to regress, 4 months of sulindac therapy may not reliably prevent colorectal cancer development in these patients.
Collapse
Affiliation(s)
- N Matsuhashi
- Third Department of Internal Medicine, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | | | |
Collapse
|
24
|
Picariello L, Brandi ML, Formigli L, Orlandini SZ, Dolara P, Caderni G, Raimondi L, Tonelli F. Apoptosis induced by sulindac sulfide in epithelial and mesenchymal cells from human abdominal neoplasms. Eur J Pharmacol 1998; 360:105-12. [PMID: 9845279 DOI: 10.1016/s0014-2999(98)00665-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We investigated whether the therapeutic action of sulindac, used for the treatment of familial adenomatous polyposis, desmoid tumors, and against colon cancer, could be mediated by its active metabolite, sulindac sulfide, in cell growth and apoptosis on cell lines derived from abdominal neoplasms. Sulindac sulfide actions on cell growth and apoptosis were evaluated in epithelial human colon tumor 8 (HCT8) cell line and mesenchymal cell lines (bovine bone endothelial (BBE) cell line, desmoid tumor-derived cells, human colorectal cancer-derived fibroblasts). Sulindac sulfide (0.1-60 microg/ml) induced a dose-dependent inhibition of cell proliferation of all cell lines tested. Apoptosis was induced at doses of 20 and 40 microg/ml, respectively, in BBE and HCT8 cells with no effect on desmoid tumor cells and colorectal cancer-derived fibroblasts. Since mesenchymal cells respond to clinically effective concentrations of the compound, its preferential action on the stromal compartment of intestinal polyps, desmoid tumors and colon cancer can be proposed, with consequent regression of the tumor.
Collapse
Affiliation(s)
- L Picariello
- Department of Clinical Physiopathology, University of Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Krishnan K, Ruffin MT, Brenner DE. Clinical models of chemoprevention for colon cancer. Hematol Oncol Clin North Am 1998; 12:1079-113, viii. [PMID: 9888022 DOI: 10.1016/s0889-8588(05)70042-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Colon cancer is a common malignancy in the westernized world and is incurable in its advanced stages. This article summarizes the currently available information on colorectal cancer chemoprevention. A brief outline of the incidence and etiologic factors is followed by a discussion of the evidence on which chemopreventive strategies for colon cancer are modeled. This includes a description of the development of surrogate endpoint biomarkers and experimental models to study colorectal cancer chemopreventives, a review of the promising colorectal cancer chemopreventives, and a discussion of the issues to be addressed in the design of future chemoprevention trials. The article concludes with an emphasis on the development and validation of biomarkers and selection of high-risk cohorts using genetic and epidemiologic tools as the main goals of future colon cancer chemoprevention trials before large-scale, risk-reduction trials are conducted.
Collapse
Affiliation(s)
- K Krishnan
- Department of Internal Medicine, East Tennessee State University, Johnson City, USA
| | | | | |
Collapse
|
26
|
Lefebvre AM, Chen I, Desreumaux P, Najib J, Fruchart JC, Geboes K, Briggs M, Heyman R, Auwerx J. Activation of the peroxisome proliferator-activated receptor gamma promotes the development of colon tumors in C57BL/6J-APCMin/+ mice. Nat Med 1998; 4:1053-7. [PMID: 9734399 DOI: 10.1038/2036] [Citation(s) in RCA: 463] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The development of colorectal cancer, one of the most frequent cancers, is influenced by prostaglandins and fatty acids. Decreased prostaglandin production, seen in mice with mutations in the cyclooxygenase 2 gene or in animals and humans treated with cyclooxygenase inhibitors, prevents or attenuates colon cancer development. There is also a strong correlation between the intake of fatty acids from animal origin and colon cancer. Therefore, the peroxisome proliferator-activated receptor gamma (PPARgamma), a downstream transcriptional mediator for prostaglandins and fatty acids which is highly expressed in the colon may be involved in this process. Activation of PPARgamma by two different synthetic agonists increased the frequency and size of colon tumors in C57BL/6J-APCMin/+ mice, an animal model susceptible to intestinal neoplasia. Tumor frequency was only increased in the colon, and did not change in the small intestine, coinciding with the colon-restricted expression of PPARgamma. Treatment with PPARgamma agonists increased beta-catenin levels both in the colon of C57BL/61-APCMin/+ mice and in HT-29 colon carcinoma cells. Genetic abnormalities in the Wnt/wingless/APC pathway, which enhance the transcriptional activity of the beta-catenin-T-cell factor/lymphoid enhancer factor 1 transcription complex, often underly the development of colon tumors. Our data indicate that PPARgamma activation modifies the development of colon tumors in C57BL/61-APCMin/+ mice.
Collapse
Affiliation(s)
- A M Lefebvre
- LBRE, U.325 INSERM, Département d'Athérosclérose, Institut Pasteur, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|