1
|
Baldini E, Cardarelli S, Lori E, Bonati E, Gagliardi F, Pironi D, Fallahi P, Antonelli A, D’Andrea V, Ulisse S, Sorrenti S. The Potential Therapeutic Value of Aspirin in Anaplastic Thyroid Cancer. Cancers (Basel) 2024; 16:4203. [PMID: 39766102 PMCID: PMC11674608 DOI: 10.3390/cancers16244203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Background: several experimental findings and epidemiological observations indicated that aspirin/acetylsalicylic acid (ASA) may be endowed with anticancer effects against a variety of human malignancies, including thyroid carcinomas. Among these, undifferentiated/anaplastic thyroid carcinoma (ATC) is one of the most aggressive and lethal human cancers, refractory to all currently available therapies. Methods: we here evaluated in a preclinical setting the effects of ASA on a panel of three ATC-derived cell lines: the CAL-62, the 8305C, and the 8505C. Results: the data obtained demonstrated the ability of ASA to inhibit, in a dose- and time-dependent manner, the proliferation of all ATC cell lines investigated, with IC50 values comprised between 2.0 and 4.3 mM. Cell growth was restrained with the same efficacy when the ASA treatment was applied to three-dimensional soft-agar cultures. In addition, ASA significantly reduced migration and invasion in two of the three ATC cell lines. We finally investigated the effects of ASA on the MAPK and PI3K/Akt signaling pathways, which are often altered in ATC. The results showed that the phosphorylation status of the Akt1/2/3 kinases was significantly reduced following ASA treatment, while ERK1/2 phosphorylation was either unaffected or slightly upregulated. Conclusions: our findings support epidemiological evidence on the anticancer potential of ASA. On this basis, further investigations should be carried out to assess the usefulness of ASA as adjuvant therapy in patients affected by ATC.
Collapse
Affiliation(s)
- Enke Baldini
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Silvia Cardarelli
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Eleonora Lori
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Elena Bonati
- General Surgery Unit, Department of Medicine and Surgery, Parma University Hospital, 43126 Parma, Italy;
| | - Federica Gagliardi
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Daniele Pironi
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Alessandro Antonelli
- Department of Surgery, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy;
| | - Vito D’Andrea
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Salvatore Ulisse
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| | - Salvatore Sorrenti
- Department of Surgery, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.C.); (E.L.); (F.G.); (D.P.); (V.D.); (S.S.)
| |
Collapse
|
2
|
Ezenkwa US, Omenai SA, Iyapo O, Ezekekwu CA, Adetona AE, Akunwata CU, Ale AO, Ebili HO. Low prostaglandin-endoperoxide synthase-2 gene expression in colorectal carcinomas may predict poorer survival. Ecancermedicalscience 2024; 18:1814. [PMID: 40171466 PMCID: PMC11959140 DOI: 10.3332/ecancer.2024.1814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Indexed: 04/03/2025] Open
Abstract
Introduction Prostaglandin-endoperoxide synthase-2 (ptgs2), otherwise called Cyclooxygenase 2, is overexpressed in colorectal carcinoma (CRC) compared to normal tissues. However, the impact of differential expression among ptgs2-positive tumours on CRC prognosis has not been well investigated. By sub-stratifying positive tumour expression, this study determined its potential influence on patients' outcomes. Methods The Cancer Genome Atlas database was explored to determine CRC cases with RNA-Sequence (RNA-Seq) transcript data and matched clinicopathological data alongside gene copy number variation and methylation status. Descriptive, chi-square, Fisher exact, Linear-by-Linear associations, logistic and Kaplan-Meier statistics were used to determine proportions, associations, predictors and survival between ptgs2 and tumour parameters using Statistical Package for Social Sciences version 20. Two-tailed p-value <0.05 was accepted as statistically significant. Results There were 534 CRC classified predominantly as adenocarcinoma not otherwise specified (86.3%) and mucinous carcinoma (12.4) histologically included in this study. Marker (ptgs2) expression ranged from 0.02 FPKM-131.89 FPKM, (Median 1.4 FPKM). The majority of the cases (53.4%) were diagnosed at an early stage and showed high ptgs2 RNA-Sequence (RNA-seq) expression in 51.5% (275/534). Significant associations were seen between ptgs2 expression and histological subtype (p < 0.001), lymphovascular invasion (p = 0.013), pN2 stage (> 6 positive lymph nodes) (p = 0.011) and American Joint Committee on Cancer Staging stage (p = 0.028), and these all had lower ptgs2 expression. On regression analysis, histological differentiation emerged as a predictor of ptgs2 expression (Odds ratio 2.749, 95% confidence interval 1.479-5.108, p < 0.001). Also, gene methylation was associated with reduced ptgs2 expression. Overall survival was significantly inferior among individuals with low ptgs2 tumours (p = 0.018) while that for disease-free survival was non-significant (p = 0.327). Conclusion CRCs with low ptgs2 transcripts are associated with poorer survival. This finding suggests a need for closer follow up and tailored adjuvant therapy for these patients.
Collapse
Affiliation(s)
- Uchenna Simon Ezenkwa
- Department of Histopathology, Federal University of Health Sciences Azare, Azare 751101, Bauchi, Nigeria
- Department of Pathology, Federal Medical Centre Azare, Azare 751101, Bauchi, Nigeria
| | | | - Oluwadamilare Iyapo
- Department of Pathology, Federal Medical Centre, Ebute Metta, Lagos 101212, Nigeria
| | - Chinedu Anthony Ezekekwu
- Bristol Haematology and Oncology Centre, University Hospital, Bristol and Weston NHS Trust, Bristol BS28ED, UK
| | - Adesoji E Adetona
- Department of Morbid Anatomy & Histopathology, Olabisi Onabanjo University, Ago-Iwoye 121101, Ogun State, Nigeria
| | - Chima Uzoma Akunwata
- Department of Haematology, University College Hospital Ibadan, Ibadan 200221, Oyo, Nigeria
| | - Ayotunde Oladunmi Ale
- Department of Medicine, Olabisi Onabanjo University/University Teaching Hospital, Sagamu 121101, Ogun, Nigeria
| | - Henry Okwuchukwu Ebili
- Department of Morbid Anatomy & Histopathology, Olabisi Onabanjo University, Ago-Iwoye 121101, Ogun State, Nigeria
| |
Collapse
|
3
|
Yadav M, Abdalla M, Madhavi M, Chopra I, Bhrdwaj A, Soni L, Shaheen U, Prajapati L, Sharma M, Sikarwar MS, Albogami S, Hussain T, Nayarisseri A, Singh SK. Structure-Based Virtual Screening, Molecular Docking, Molecular Dynamics Simulation and Pharmacokinetic modelling of Cyclooxygenase-2 (COX-2) inhibitor for the clinical treatment of Colorectal Cancer. MOLECULAR SIMULATION 2022. [DOI: 10.1080/08927022.2022.2068799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Manasi Yadav
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, PR People’s Republic of China
| | - Maddala Madhavi
- Department of Zoology, Osmania University, Hyderabad, Telangana State, India
| | - Ishita Chopra
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
- Bioinformatics Research Laboratory, LeGene Biosciences Pvt Ltd, Indore, Madhya Pradesh, India
| | - Anushka Bhrdwaj
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Lovely Soni
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Uzma Shaheen
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Leena Prajapati
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | - Megha Sharma
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
| | | | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Tajamul Hussain
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
- Center of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anuraj Nayarisseri
- In silico Research Laboratory, Eminent Biosciences, Indore, Madhya Pradesh, India
- Bioinformatics Research Laboratory, LeGene Biosciences Pvt Ltd, Indore, Madhya Pradesh, India
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Designing and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
4
|
Shuey MM, Xiang RR, Moss ME, Carvajal BV, Wang Y, Camarda N, Fabbri D, Rahman P, Ramsey J, Stepanian A, Sebastiani P, Wells QS, Beckman JA, Jaffe IZ. Systems Approach to Integrating Preclinical Apolipoprotein E-Knockout Investigations Reveals Novel Etiologic Pathways and Master Atherosclerosis Network in Humans. Arterioscler Thromb Vasc Biol 2022; 42:35-48. [PMID: 34758633 PMCID: PMC8887835 DOI: 10.1161/atvbaha.121.317071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Animal models of atherosclerosis are used extensively to interrogate molecular mechanisms in serial fashion. We tested whether a novel systems biology approach to integration of preclinical data identifies novel pathways and regulators in human disease. Approach and Results: Of 716 articles published in ATVB from 1995 to 2019 using the apolipoprotein E knockout mouse to study atherosclerosis, data were extracted from 360 unique studies in which a gene was experimentally perturbed to impact plaque size or composition and analyzed using Ingenuity Pathway Analysis software. TREM1 (triggering receptor expressed on myeloid cells) signaling and LXR/RXR (liver X receptor/retinoid X receptor) activation were identified as the top atherosclerosis-associated pathways in mice (both P<1.93×10-4, TREM1 implicated early and LXR/RXR in late atherogenesis). The top upstream regulatory network in mice (sc-58125, a COX2 inhibitor) linked 64.0% of the genes into a single network. The pathways and networks identified in mice were interrogated by testing for associations between the genetically predicted gene expression of each mouse pathway-identified human homolog with clinical atherosclerosis in a cohort of 88 660 human subjects. Homologous human pathways and networks were significantly enriched for gene-atherosclerosis associations (empirical P<0.01 for TREM1 and LXR/RXR pathways and COX2 network). This included 12(60.0%) TREM1 pathway genes, 15(53.6%) LXR/RXR pathway genes, and 67(49.3%) COX2 network genes. Mouse analyses predicted, and human study validated, the strong association of COX2 expression (PTGS2) with increased likelihood of atherosclerosis (odds ratio, 1.68 per SD of genetically predicted gene expression; P=1.07×10-6). CONCLUSIONS PRESCIANT (Preclinical Science Integration and Translation) leverages published preclinical investigations to identify high-confidence pathways, networks, and regulators of human disease.
Collapse
Affiliation(s)
| | | | - M. Elizabeth Moss
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Brigett V. Carvajal
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Yihua Wang
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Nicholas Camarda
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Daniel Fabbri
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Protiva Rahman
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Jacob Ramsey
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Alec Stepanian
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | - Paola Sebastiani
- Department of Medicine (M.M.S., J.R., Q.S.W., J.A.B.) and Department of Biomedical Informatics (D.F., P.R.), Vanderbilt University Medical Center, Nashville, TN. Molecular Cardiology Research Institute (R.R.X., M.E.M., B.V.C., Y.W., N.C., A.S., I.Z.J.) and Institute for Clinical Research and Health Policy Studies (P.S.), Tufts Medical Center, Boston, MA
| | | | | | | |
Collapse
|
5
|
Tripathi S, Kushwaha R, Mishra J, Gupta MK, Kumar H, Sanyal S, Singh D, Sanyal S, Sahasrabuddhe AA, Kamthan M, Mudiam MKR, Bandyopadhyay S. Docosahexaenoic acid up-regulates both PI3K/AKT-dependent FABP7-PPARγ interaction and MKP3 that enhance GFAP in developing rat brain astrocytes. J Neurochem 2016; 140:96-113. [DOI: 10.1111/jnc.13879] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/24/2016] [Accepted: 10/20/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Sachin Tripathi
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Amity Institute of Biotechnology; Amity University (Lucknow campus); Lucknow India
| | - Rajesh Kushwaha
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IITR campus; Lucknow India
| | - Juhi Mishra
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Babu Banarasi Das University; Lucknow India
| | - Manoj Kumar Gupta
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IITR campus; Lucknow India
- Analytical Chemistry Laboratory and Regulatory Toxicology group; CSIR-IITR; Lucknow India
| | - Harish Kumar
- Division of Biochemistry; CSIR-Central Drug Research Institute (CDRI); Lucknow India
| | - Somali Sanyal
- Amity Institute of Biotechnology; Amity University (Lucknow campus); Lucknow India
| | | | - Sabyasachi Sanyal
- Division of Biochemistry; CSIR-Central Drug Research Institute (CDRI); Lucknow India
| | | | - Mohan Kamthan
- Environmental Biotechnology Laboratory; Environmental Toxicology Group; CSIR-IITR; Lucknow India
| | | | - Sanghamitra Bandyopadhyay
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IITR campus; Lucknow India
| |
Collapse
|
6
|
McCowan C, Munro AJ, Donnan PT, Steele RJC. Use of aspirin post-diagnosis in a cohort of patients with colorectal cancer and its association with all-cause and colorectal cancer specific mortality. Eur J Cancer 2012. [PMID: 23182687 DOI: 10.1016/j.ejca.2012.10.024] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Aspirin is associated with a reduced risk of developing colorectal cancer. This study examined whether patients with colorectal cancer prescribed aspirin had improved survival. DESIGN An observational population cohort study was undertaken using data linkage of cancer registry, dispensed prescriptions and death certificate records in Tayside, Scotland. All community prescribed aspirin pre- and post-diagnosis was extracted and periods of aspirin use post-diagnosis for each individual were analysed using Cox proportional hazard models. Main outcome measures were all-cause and colorectal mortality from death certificates. RESULTS Two thousand nine hundred ninety patients were identified with colorectal cancer between 1st January 1997 and 30th December 2006 and followed up until 28th February 2010. Median age at diagnosis was 73 (interquartile range [IQR] 65-80) with 52% male. One thousand nine hundred ninety-eight (67%) deaths were recorded with 1021 (34%) attributed to colorectal cancer. One thousand three hundred forty (45%) patients used aspirin at some stage of the study period. Aspirin use post-diagnosis was associated with lower risk of all cause mortality (hazard ratio [HR]=0.67, 95% confidence interval [CI]=0.57-0.79, p<0.001) and colorectal cancer specific mortality after allowing for age, Dukes' stage, gender, socio-economic status and aspirin use pre-diagnosis. Increasing age and stage at diagnosis were associated with increased risk, with more affluent patients at reduced risk. CONCLUSIONS Our study suggests that aspirin use post-diagnosis of colorectal cancer may reduce both all cause and colorectal cancer specific mortality. However further work is required to ensure this is a causal relationship and to identify whether it is best used in specific groups of patients.
Collapse
Affiliation(s)
- C McCowan
- Division of Population Health Sciences, Medical Research Institute, University of Dundee, Dundee, United Kingdom.
| | | | | | | |
Collapse
|
7
|
Abstract
Celecoxib is a multifaceted drug with promising anticancer properties. A number of studies have been conducted that implicate the compound in modulating the expression of Bcl-2 family members and mitochondria-mediated apoptosis. The growing data surrounding the role of celecoxib in the regulation of the mitochondrial death pathway provides a platform for ongoing debate. Studies that describe celecoxib's properties as a BH3 mimic or as a direct inhibitor of Bcl-2 are not available. The motivations for this review are: to provide the basis for the development of novel compounds that modulate Bcl-2 expression using celecoxib as a structural starting point and to encourage additional biological studies (such as binding and enzymatic assays) that would provide information regarding celecoxib's role as a Bcl-2 antagonist. The current review summarizes work that identifies the role of celecoxib in blocking the activity of Bcl-2.
Collapse
|
8
|
Abstract
Background: The preventive role of non-steroid anti-inflammatory drugs (NSAIDs) and aspirin, in particular, on colorectal cancer is well established. More recently, it has been suggested that aspirin may also have a therapeutic role. Aim of the present observational population-based study was to assess the therapeutic effect on overall survival of aspirin/NSAIDs as adjuvant treatment used after the diagnosis of colorectal cancer patients. Methods: Data concerning prescriptions were obtained from PHARMO record linkage systems and all patients diagnosed with colorectal cancer (1998–2007) were selected from the Eindhoven Cancer Registry (population-based cancer registry). Aspirin/NSAID use was classified as none, prediagnosis and postdiagnosis and only postdiagnosis. Patients were defined as non-user of aspirin/NSAIDs from the date of diagnosis of the colorectal cancer to the date of first use of aspirin or NSAIDs and user from first use to the end of follow-up. Poisson regression was performed with user status as time-varying exposure. Results: In total, 1176 (26%) patients were non-users, 2086 (47%) were prediagnosis and postdiagnosis users and 1219 (27%) were only postdiagnosis users (total n=4481). Compared with non-users, a survival gain was observed for aspirin users; the adjusted rate ratio (RR) was 0.77 (95% confidence interval (CI) 0.63–0.95; P=0.015). Stratified for colon and rectal, the survival gain was only present in colon cancer (adjusted RR 0.65 (95%CI 0.50–0.84; P=0.001)). For frequent users survival gain was larger (adjusted RR 0.61 (95%CI 0.46–0.81; P=0.001). In rectal cancer, aspirin use was not associated with survival (adjusted RR 1.10 (95%CI 0.79–1.54; P=0.6). The NSAIDs use was associated with decreased survival (adjusted RR 1.93 (95%CI 1.70–2.20; P<0.001). Conclusion: Aspirin use initiated or continued after diagnosis of colon cancer is associated with a lower risk of overall mortality. These findings strongly support initiation of a placebo-controlled trial that investigates the role of aspirin as adjuvant treatment in colon cancer patients.
Collapse
|
9
|
Identifying inhibitors of epithelial-mesenchymal transition by connectivity map-based systems approach. J Thorac Oncol 2012; 6:1784-92. [PMID: 21964532 DOI: 10.1097/jto.0b013e31822adfb0] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Acquisition of mesenchymal phenotype by epithelial cells by means of epithelial-mesenchymal transition (EMT) is considered as an early event in the multistep process of tumor metastasis. Therefore, inhibition of EMT might be a rational strategy to prevent metastasis. METHODS Using the global gene expression profile from a cell culture model of transforming growth factor-β (TGF-β)-induced EMT, we identified potential EMT inhibitors. We used a publicly available database (www.broad.mit.edu/cmap) comprising gene expression profiles obtained from multiple different cell lines in response to various drugs to derive negative correlations to EMT gene expression profile using Connectivity Map, a pattern matching tool. RESULTS Experimental validation of the identified compounds showed rapamycin as a novel inhibitor of TGF-β signaling along with 17-AAG, a known modulator of TGF-β pathway. Both of these compounds completely blocked EMT and the associated migratory and invasive phenotype. The other identified compound, LY294002, demonstrated a selective inhibition of mesenchymal markers, cell migration and invasion, without affecting the loss of E-cadherin expression or Smad phosphorylation. CONCLUSIONS Our data reveal that rapamycin is a novel modulator of TGF-β signaling, and along with 17-AAG and LY294002, could be used as therapeutic agent for inhibiting EMT. This study demonstrates the potential of a systems approach in identifying novel modulators of a complex biological process.
Collapse
|
10
|
James CG, Stanton LA, Agoston H, Ulici V, Underhill TM, Beier F. Genome-wide analyses of gene expression during mouse endochondral ossification. PLoS One 2010; 5:e8693. [PMID: 20084171 PMCID: PMC2805713 DOI: 10.1371/journal.pone.0008693] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 12/13/2009] [Indexed: 12/24/2022] Open
Abstract
Background Endochondral ossification is a complex process involving a series of events that are initiated by the establishment of a chondrogenic template and culminate in its replacement through the coordinated activity of osteoblasts, osteoclasts and endothelial cells. Comprehensive analyses of in vivo gene expression profiles during these processes are essential to obtain a complete understanding of the regulatory mechanisms involved. Methodology/Principal Findings To address these issues, we completed a microarray screen of three zones derived from manually segmented embryonic mouse tibiae. Classification of genes differentially expressed between each respective zone, functional categorization as well as characterization of gene expression patterns, cytogenetic loci, signaling pathways and functional motifs both confirmed reported data and provided novel insights into endochondral ossification. Parallel comparisons of the microdissected tibiae data set with our previously completed micromass culture screen further corroborated the suitability of micromass cultures for modeling gene expression in chondrocyte development. The micromass culture system demonstrated striking similarities to the in vivo microdissected tibiae screen; however, the micromass system was unable to accurately distinguish gene expression differences in the hypertrophic and mineralized zones of the tibia. Conclusions/Significance These studies allow us to better understand gene expression patterns in the growth plate and endochondral bones and provide an important technical resource for comparison of gene expression in diseased or experimentally-manipulated cartilages. Ultimately, this work will help to define the genomic context in which genes are expressed in long bones and to understand physiological and pathological ossification.
Collapse
Affiliation(s)
- Claudine G. James
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Lee-Anne Stanton
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Hanga Agoston
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Veronica Ulici
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- * E-mail: (VU); (FB)
| | - T. Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank Beier
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- * E-mail: (VU); (FB)
| |
Collapse
|
11
|
Abstract
CONTEXT Aspirin reduces risk of colorectal neoplasia in randomized trials and inhibits tumor growth and metastases in animal models. However, the influence of aspirin on survival after diagnosis of colorectal cancer is unknown. OBJECTIVE To examine the association between aspirin use after colorectal cancer diagnosis on colorectal cancer-specific and overall survival. DESIGN, SETTING, AND PARTICIPANTS Prospective cohort study of 1279 men and women diagnosed with stage I, II, or III colorectal cancer. Participants were enrolled in 2 nationwide health professional cohorts in 1980 and 1986 prior to diagnosis and followed up through June 1, 2008. MAIN OUTCOME MEASURE Colorectal cancer-specific and overall mortality. RESULTS After a median follow-up of 11.8 years, there were 193 total deaths (35%) and 81 colorectal cancer-specific deaths (15%) among 549 participants who regularly used aspirin after colorectal cancer diagnosis, compared with 287 total deaths (39%) and 141 colorectal cancer-specific deaths (19%) among 730 participants who did not use aspirin. Compared with nonusers, participants who regularly used aspirin after diagnosis experienced a multivariate hazard ratio (HR) for colorectal cancer-specific mortality of 0.71 (95% confidence interval [CI], 0.53-0.95) and for overall mortality of 0.79 (95% CI, 0.65-0.97). Among 719 participants who did not use aspirin before diagnosis, aspirin use initiated after diagnosis was associated with a multivariate HR for colorectal cancer-specific mortality of 0.53 (95% CI, 0.33-0.86). Among 459 participants with colorectal cancers that were accessible for immunohistochemical assessment, the effect of aspirin differed significantly according to cyclooxygenase 2 (COX-2) expression (P for interaction = .04). Regular aspirin use after diagnosis was associated with a lower risk of colorectal cancer-specific mortality among participants in whom primary tumors overexpressed COX-2 (multivariate HR, 0.39; 95% CI, 0.20-0.76), whereas aspirin use was not associated with lower risk among those with primary tumors with weak or absent expression (multivariate HR, 1.22; 95% CI, 0.36-4.18). CONCLUSION Regular aspirin use after the diagnosis of colorectal cancer is associated with lower risk of colorectal cancer-specific and overall mortality, especially among individuals with tumors that overexpress COX-2.
Collapse
Affiliation(s)
- Andrew T Chan
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, GRJ 722 Boston, MA 02114, USA.
| | | | | |
Collapse
|
12
|
Stoeltzing O, Liu W, Fan F, Wagner C, Stengel K, Somcio RJ, Reinmuth N, Parikh AA, Hicklin DJ, Ellis LM. Regulation of cyclooxygenase-2 (COX-2) expression in human pancreatic carcinoma cells by the insulin-like growth factor-I receptor (IGF-IR) system. Cancer Lett 2007; 258:291-300. [PMID: 17950526 PMCID: PMC2147684 DOI: 10.1016/j.canlet.2007.09.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 09/15/2007] [Accepted: 09/17/2007] [Indexed: 01/18/2023]
Abstract
Both the insulin-like growth factor-I receptor (IGF-IR) and cyclooxygenase-2 (COX-2) are frequently overexpressed in pancreatic cancer. We hypothesized that IGF-IR is directly involved in induction of COX-2 and sought to investigate signaling pathways mediating this effect. Pancreatic cancer cells (L3.6pl) were stably transfected with a dominant-negative receptor (IGF-IR DN) construct or empty vector (pcDNA). Cells were stimulated with IGF-I to determine activated signaling intermediates and induction of COX-2. Signaling pathways mediating COX-2 induction were identified using signaling inhibitors. IGF-I up-regulated COX-2 selectively via the MAPK/(Erk-1/2) pathway. In addition, IGF-IR DN cells showed a marked decrease in constitutive COX-2 and a blunted response to IGF-I. Similarly, treatment with an anti-IGF-IR antibody effectively inhibited IGF-IR and MAPK/Erk activation and decreased COX-2 in parental cells. In conclusion, activation of IGF-IR mediates COX-2 expression in human pancreatic cancer cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Blotting, Northern
- Blotting, Western
- Cell Line, Tumor
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Insulin Receptor Substrate Proteins
- Insulin-Like Growth Factor I/pharmacology
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/metabolism
- Models, Biological
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Phosphorylation/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/physiology
- Signal Transduction/drug effects
- Transfection
Collapse
Affiliation(s)
- Oliver Stoeltzing
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Departments of Surgery and Surgical Oncology, University of Regensburg Medical Center, Regensburg, Germany
| | - Wenbiao Liu
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Fan Fan
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Christine Wagner
- Departments of Surgery and Surgical Oncology, University of Regensburg Medical Center, Regensburg, Germany
| | - Kathrin Stengel
- Departments of Surgery and Surgical Oncology, University of Regensburg Medical Center, Regensburg, Germany
| | - Ray J. Somcio
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Niels Reinmuth
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Alexander A. Parikh
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Lee M. Ellis
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
13
|
|
14
|
Marshall JC, Caissie AL, Cruess SR, Cools-Lartigue J, Burnier MN. The effects of a cyclooxygenase-2 (COX-2) expression and inhibition on human uveal melanoma cell proliferation and macrophage nitric oxide production. J Carcinog 2007; 6:17. [PMID: 18042295 PMCID: PMC2222223 DOI: 10.1186/1477-3163-6-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Accepted: 11/27/2007] [Indexed: 01/11/2023] Open
Abstract
Background Cyclooxygenase-2 (COX-2) expression has previously been identified in uveal melanoma although the biological role of COX-2 in this intraocular malignancy has not been elucidated. This study aimed to investigate the effect of a COX-2 inhibitor on the proliferation rate of human uveal melanoma cells, as well as its effect on the cytotoxic response of macrophages. Methods Human uveal melanoma cell lines were transfected to constitutively express COX-2 and the proliferative rate of these cells using two different methods, with and without the addition of Amfenac, was measured. Nitric oxide production by macrophages was measured after exposure to melanoma-conditioned medium from both groups of cells as well as with and without Amfenac, the active metabolite of Nepafenac. Results Cells transfected to express COX-2 had a higher proliferation rate than those that did not. The addition of Amfenac significantly decreased the proliferation rate of all cell lines. Nitric oxide production by macrophages was inhibited by the addition of melanoma conditioned medium, the addition of Amfenac partially overcame this inhibition. Conclusion Amfenac affected both COX-2 transfected and non-transfected uveal melanoma cells in terms of their proliferation rates as well as their suppressive effects on macrophage cytotoxic activity.
Collapse
Affiliation(s)
- Jean-Claude Marshall
- The Henry C, Witelson Ocular Pathology Laboratory and Registry, McGill University, 3775 University Street, Lyman Duff Building, Room 216, Montreal, Quebec H3A 2B4, Canada.
| | | | | | | | | |
Collapse
|
15
|
Buchanan FG, Holla V, Katkuri S, Matta P, DuBois RN. Targeting cyclooxygenase-2 and the epidermal growth factor receptor for the prevention and treatment of intestinal cancer. Cancer Res 2007; 67:9380-8. [PMID: 17909047 DOI: 10.1158/0008-5472.can-07-0710] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical and animal studies indicate a role for cyclooxygenase-2 (COX-2) and the epidermal growth factor receptor (EGFR) in the development and progression of intestinal polyps and cancers. Although this combination of enzyme inhibition has shown synergy in intestinal polyp and tumor models, the exact mechanism for these effects remains undefined. Therefore, we sought to define the molecular mechanisms through which this process occurs. We observed a significant reduction in the number and size of small intestinal polyps in APC(min+/-) mice treated with either celecoxib (a selective COX-2 inhibitor) or erlotinib (Tarceva, an EGFR inhibitor). However, in combination, there was an overall prevention in the formation of polyps by over 96%. Furthermore, we observed a 70% reduction of colorectal xenograft tumors in mice treated with the combination and microarray analysis revealed genes involved in cell cycle progression were negatively regulated. Although we did not observe significant changes in mRNAs of genes with known apoptotic function, there was a significant increase of apoptosis in tumors from animals treated with the combination. The inhibition of EGFR also induced the down-regulation of COX-2 and further inhibited prostaglandin E2 formation. We observed similar effects on the prevention of intestinal adenomas and reduction of xenograft tumor volume when nonselective COX inhibitors were used in combination with erlotinib. Together, these findings suggest that the inhibition of both COX-2 and EGFR may provide a better therapeutic strategy than either single agent through a combination of decreased cellular proliferation and prostaglandin signaling as well as increased apoptosis.
Collapse
Affiliation(s)
- F Gregory Buchanan
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 27232-6838, USA
| | | | | | | | | |
Collapse
|
16
|
Ali MA, Choy H, Habib AA, Saha D. SNS-032 prevents tumor cell-induced angiogenesis by inhibiting vascular endothelial growth factor. Neoplasia 2007; 9:370-81. [PMID: 17534442 PMCID: PMC1877978 DOI: 10.1593/neo.07136] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 03/22/2007] [Accepted: 03/23/2007] [Indexed: 12/28/2022] Open
Abstract
Cell proliferation, migration, and capillary network formation of endothelial cells are the fundamental steps for angiogenesis, which involves the formation of new blood vessels. The purpose of this study is to investigate the effect of a novel aminothiazole SNS-032 on these critical steps for in vitro angiogenesis using a coculture system consisting of human umbilical vein endothelial cells (HUVECs) and human glioblastoma cells (U87MG). SNS-032 is a potent selective inhibitor of cyclin-dependent kinases 2, 7, and 9, and inhibits both transcription and cell cycle. In this study, we examined the proliferation and viability of HUVECs and U87MG cells in the presence of SNS-032 and observed a dose-dependent inhibition of cellular proliferation in both cell lines. SNS-032 inhibited threedimensional capillary network formations of endothelial cells. In a coculture study, SNS-032 completely prevented U87MG cell-mediated capillary formation of HUVECs. This inhibitor also prevented the migration of HUVECs when cultured alone or cocultured with U87MG cells. In addition, SNS-032 significantly prevented the production of vascular endothelial growth factor (VEGF) in both cell lines, whereas SNS-032 was less effective in preventing capillary network formation and migration of endothelial cells when an active recombinant VEGF was added to the medium. In conclusion, SNS-032 prevents in vitro angiogenesis, and this action is attributable to blocking of VEGF.
Collapse
Affiliation(s)
- M. Aktar Ali
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9187, USA
| | - Hak Choy
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9187, USA
| | - Amyn A Habib
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9187, USA
| | - Debabrata Saha
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9187, USA
| |
Collapse
|
17
|
Bottone FG, Moon Y, Kim JS, Alston-Mills B, Ishibashi M, Eling TE. The anti-invasive activity of cyclooxygenase inhibitors is regulated by the transcription factor ATF3 (activating transcription factor 3). Mol Cancer Ther 2005; 4:693-703. [PMID: 15897233 DOI: 10.1158/1535-7163.mct-04-0337] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We previously showed that nonsteroidal anti-inflammatory drugs (NSAID) such as sulindac sulfide, which has chemopreventive activity, modulate the expression of several genes detected by microarray analysis. Activating transcription factor 3 (ATF3) was selected for further study because it is a transcription factor involved in cell proliferation, apoptosis, and invasion, and its expression is repressed in human colorectal tumors as compared with normal adjacent tissue. In this report, we show that ATF3 mRNA and protein expression are up-regulated in HCT-116 human colorectal cancer cells following treatment with NSAIDs, troglitazone, diallyl disulfide, and resveratrol. To ascertain the biological significance of ATF3, we overexpressed full-length ATF3 protein in the sense and antisense orientations. Overexpression of ATF3 in the sense orientation decreased focus formation in vitro and reduced the size of mouse tumor xenografts by 54% in vivo. Conversely, overexpression of antisense ATF3 was protumorigenic in vitro, however, not in vivo. ATF3 in the sense orientation did not modulate apoptosis, indicating another mechanism is involved. With microarray analysis, several genes relating to invasion and metastasis were identified by ATF3 overexpression and were confirmed by real-time reverse transcription-PCR, and several of these genes were modulated by sulindac sulfide, which inhibited invasion in these cells. Furthermore, overexpression of ATF3 inhibited invasion to a similar degree as sulindac sulfide treatment, whereas antisense ATF3 increased invasion. In conclusion, ATF3 represents a novel mechanism in which NSAIDs exert their anti-invasive activity, thereby linking ATF3 and its gene regulatory activity to the biological activity of these compounds.
Collapse
Affiliation(s)
- Frank G Bottone
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, NIH, P.O. Box 12233, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|
18
|
Ding J, Chang Q, Gong S. Inhibitory effects of Celecoxib and Sc-58125 on proliferation of human carcinoma of larynx Hep-2 in vitro. Curr Med Sci 2005; 25:202-5. [PMID: 16116973 DOI: 10.1007/bf02873577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Indexed: 10/19/2022]
Abstract
The inhibitory effects of two kinds of selective cyclooxygenase-2 inhibitors on the proliferation of human carcinoma of larynx Hep-2 in vitro and their corresponding mechanisms were investigated. Hep-2 cells were cultured with two kinds of selective cyclooxygenase-2 inhibitors (Sc-58125 and Celecoxib) at various concentrations for 24 h. Morphological changes were observed under the phase microscopy and the growth suppression was detected by using MTT colorimetric assay. Apoptotic DNA fragments were observed by agarose gel electrophoresis, and the cell cycle and apoptotic rate were detected by flow cytometry (FCM) respectively. Hep-2 cells became rounded and detached from the culture dish after being treated with Celecoxib for 24 h, however, they remained morphologically unchanged with Sc-58125. Sc-58125 could increase G2 phase cells, whereas, Celecoxib rose G1 phase cells. Both of the two effects were dose-dependent. Moreover, the Hep-2 cells cultured with 50 micromol/L and 100 micromol/L Celecoxib showed obvious apoptosis, with the nuclear DNA of cells exhibiting characteristic DNA ladder. So Sc-58125 could inhibit the proliferation of Hep-2 cells by altering the G2 phase cells. However, Celecoxib had the same effect by changing the G1 phase cells and inducing apoptosis at higher concentration.
Collapse
Affiliation(s)
- Juan Ding
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | |
Collapse
|
19
|
Hull ML, Prentice A, Wang DY, Butt RP, Phillips SC, Smith SK, Charnock-Jones DS. Nimesulide, a COX-2 inhibitor, does not reduce lesion size or number in a nude mouse model of endometriosis. Hum Reprod 2004; 20:350-8. [PMID: 15567877 DOI: 10.1093/humrep/deh611] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Women with endometriosis have elevated levels of cyclooxygenase-2 (COX-2) in peritoneal macrophages and endometriotic tissue. Inhibition of COX-2 has been shown to reduce inflammation, angiogenesis and cellular proliferation. It may also downregulate aromatase activity in ectopic endometrial lesions. Ectopic endometrial establishment and growth are therefore likely to be suppressed in the presence of COX-2 inhibitors. We hypothesized that COX-2 inhibition would reduce the size and number of ectopic human endometrial lesions in a nude mouse model of endometriosis. METHODS The selective COX-2 inhibitor, nimesulide, was administered to estrogen-supplemented nude mice implanted with human endometrial tissue. Ten days after implantation, the number and size of ectopic endometrial lesions were evaluated and compared with lesions from a control group. Immunohistochemical assessment of vascular development and macrophage and myofibroblast infiltration in control and treated lesions was performed. RESULTS There was no difference in the number or size of ectopic endometrial lesions in control and nimesulide-treated nude mice. Nimesulide did not induce a visually identifiable difference in blood vessel development or macrophage or myofibroblast infiltration in nude mouse explants. CONCLUSION The hypothesized biological properties of COX-2 inhibition did not influence lesion number or size in the nude mouse model of endometriosis.
Collapse
Affiliation(s)
- M L Hull
- Reproductive Molecular Research Group, Department of Pathology Tennis Court Road, Cambridge CB2 1QP, UK.
| | | | | | | | | | | | | |
Collapse
|
20
|
Buchanan FG, Chang W, Sheng H, Shao J, Morrow JD, DuBois RN. Up-regulation of the enzymes involved in prostacyclin synthesis via Ras induces vascular endothelial growth factor. Gastroenterology 2004; 127:1391-400. [PMID: 15521009 DOI: 10.1053/j.gastro.2004.07.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS The constitutive activation of Ras is an important step in the development and progression of several different cancers and is known to increase the level of cyclooxygenase 2 (COX-2). Prostaglandins are the downstream bioactive lipid mediators produced by the COX-2 enzyme. We sought to determine the role of Ras-induced up-regulation of the enzymes involved in prostacyclin biosynthesis in nontransformed rat intestinal epithelial cells (IECs). METHODS Messenger RNA (mRNA) and protein expression were analyzed by Northern and Western analysis, respectively, to determine the level of enzymes induced by Ras. In vitro assays were used to determine the production of vascular endothelial growth factor (VEGF) and prostaglandins as well as the promoter and enzymatic activation of the rate-limiting enzyme in prostaglandin production (phospholipase A(2) [cPLA(2)]). RESULTS The inducible expression of Ha-Ras(V12) increased the production of prostaglandin (PG)F(2alpha) and prostacyclin by 2- and 13-fold, respectively. The induction of Ha-Ras(V12) also up-regulated the mRNA and protein levels of cPLA(2), COX-2, and prostacyclin synthase, as well as the promoter and enzyme activity of cPLA(2). Furthermore, oncogenic Ras increased the production of the pro-angiogenic factor VEGF. The increase of VEGF was abolished after treatment with celecoxib, a selective COX-2 inhibitor. The addition of PGI 2 alone also induced the expression of VEGF. CONCLUSIONS Inducible Ha-Ras(V12) increases the production of PGI(2) through the coordinate up-regulation of cPLA(2), COX-2, and prostacyclin synthase (PGIS). The production of PGI(2) leads to an increase in the level of the pro-angiogenic factor VEGF, which is known to play a crucial role in the regulation of tumor-associated angiogenesis.
Collapse
Affiliation(s)
- F Gregory Buchanan
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
21
|
Chen WS, Liu JH, Liu JM, Lin JK. Sequence-dependent effect of a cyclooxygenase-2 inhibitor on topoisomerase I inhibitor and 5-fluorouracil-induced cytotoxicity of colon cancer cells. Anticancer Drugs 2004; 15:287-94. [PMID: 15014363 DOI: 10.1097/00001813-200403000-00014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Selective cyclooxygenase-2 (COX-2) inhibitors have been found to induce anti-proliferative and apoptotic activity in many cancer cells. However, interaction between COX-2 inhibitors and other chemotherapeutic agents remains to be determined. We investigated the interactive effects of a selective COX-2 inhibitor, etodolac, in combination with 5-fluorouracil (5-FU) or SN-38 (active metabolite of irinotecan) on colon cancer cell lines, HT29 and SW620, in simultaneous and sequential administration schedules. Isobologram analysis demonstrated that etodolac in combination with 5-FU or SN-38 according to a simultaneous schedule resulted in only an additive effect; however, synergism was achieved in a sequential schedule. Apoptosis induction in both cell lines was also significantly increased after sequential treatment with etodolac followed by either 5-FU or SN-38 compared to that after simultaneous treatment with etodolac and either 5-FU or SN-38. Our study suggests apoptosis-inducing synergism resulted from administration of etodolac and either 5-FU or SN-38 sequentially, but not simultaneously.
Collapse
Affiliation(s)
- Wei-Shone Chen
- Division of Colorectal Surgery, Taipei Veterans General Hospital, National Yang-Ming University, Taipei, Taiwan.
| | | | | | | |
Collapse
|
22
|
Abstract
Breast cancer is a worldwide epidemic among women, and one of the most rapidly increasing cancers. Not only the incidence rate but also the death rate is increasing. Despite enthusiastic efforts in early diagnosis, aggressive surgical treatment and application of additional non-operative modalities, its prognosis is still dismal. This emphasizes the necessity to develop new measures and strategies for its prevention. The understanding of the biology of angiogenesis is improving rapidly, offering the hope for more specific vascular targeting of tumor neovasculature. Anti-angiogenic therapy is a promising, relatively new form of cancer treatment using drugs called angiogenesis inhibitors that specifically inhibit new blood vessel growth. Extensive studies conducted over the past few years have recognized that overexpression of COX-2, VEGF in the cancer might be the leading factors, can induce angiogenesis via induction of multiple pro-angiogenic regulators. Breast tumor growth and metastasization are both hormone-sensitive and angiogenesis-dependent. A single angiogenic inhibitor is not capable to inhibit angiogenesis. Therefore, we should select a combination of angiogenesis inhibitors targeting COX-2, VEGF, and bFGF pathway. This article reviews the background and implementation of the current use of angiogenesis inhibitors and discusses the likely therapeutic roles in the early and advanced breast cancer together with its potential for chemoprevention.
Collapse
Affiliation(s)
- Mohammad Atiqur Rahman
- Breast Cancer Research Program, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, 3-18-22, Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | | |
Collapse
|
23
|
Teh SH, Hill AK, Foley DA, McDermott EW, O'Higgins NJ, Young LS. COX inhibitors modulate bFGF-induced cell survival in MCF-7 breast cancer cells. J Cell Biochem 2004; 91:796-807. [PMID: 14991771 DOI: 10.1002/jcb.10767] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Basic fibroblast growth factor (bFGF) serves as a modulator of survival in breast cancer cells. The mechanisms by which bFGF transduces the anti-apoptotic signal and interacts with COX inhibitors were investigated. bFGF reduced apoptosis in MCF-7 breast cancer cells and up-regulated the expression of mitocondrial Bcl-2, whereas COX inhibitors meloxicam (selective COX-2) and aspirin (non-selective), induced apoptosis. bFGF up-regulated survivin protein expression and induced cdc-2 phosphorylation moderately at early (2-6 h), and substantially at late (24 h), time-points. Survivin mRNA expression was up-regulated only at the later time-point. COX inhibitors prevented up-regulation of survivin protein expression at both 2 and 24 h and prevented early modest increases in cdc-2 phosphorylation. Up-regulation of survivin mRNA was not found to be modulated by the COX-2 inhibitor meloxicam. bFGF regulation of survivin expression was found to be ERK1/2 kinase dependent and bFGF-induced phosphorylation of c-raf was prevented by the COX-2 inhibitor. bFGF was, however, unable to induce COX-2 protein expression or modulate COX-2 activity in MCF-7 cells as evidenced by unaltered PGE(2) production. These results indicate that bFGF regulates survivin expression in MCF-7 breast cancer cells by signaling through an ERK1/2 dependent pathway. COX-2 inhibitors can modulate bFGF-induced survivin expression in a COX-2 independent manner.
Collapse
Affiliation(s)
- Swee H Teh
- Department of Surgery, St. Vincent's University Hospital and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
24
|
Buchanan FG, Wang D, Bargiacchi F, DuBois RN. Prostaglandin E2 regulates cell migration via the intracellular activation of the epidermal growth factor receptor. J Biol Chem 2003; 278:35451-7. [PMID: 12824187 DOI: 10.1074/jbc.m302474200] [Citation(s) in RCA: 364] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Over the past decade cyclooxygenase-2-derived prostaglandins have been implicated in the development and progression of many types of cancer. Recently our laboratory has shown that treatment with prostaglandin E2 (PGE2) induces increased proliferation, migration, and invasiveness of colorectal carcinoma cells (Sheng, H., Shao, J., Washington, M. K., and DuBois, R. N. (2001) J. Biol. Chem. 276, 18075-18081). The stimulatory effects of PGE2 were dependent upon the activation of the phosphatidylinositol 3-kinase/Akt pathway. However, the exact signaling cascade responsible for phosphatidylinositol 3-kinase/Akt activation by PGE2 remains poorly defined. In the present study, we demonstrate that the PGE2-induced migration and invasion occurs via rapid transactivation and phosphorylation of the epidermal growth factor receptor (EGFR). Within minutes following treatment, PGE2 induces the activation of Akt. This effect was completely abolished by EGFR-specific tyrosine kinase inhibitors providing evidence for the role of the EGFR in this response. The rapid transactivation of the EGFR occurs via an intracellular Src-mediated event but not through the release of an extracellular epidermal growth factor-like ligand. EGFR transactivation was also observed in vivo by the direct comparison of normal and malignant human colorectal samples. These results suggest that in developing colonic carcinomas, the early effects of cyclooxygenase-2-derived PGE2 are in part mediated by the EGFR, and this transactivation is responsible for subsequent down-stream effects including the stimulation of cell migration and invasion.
Collapse
Affiliation(s)
- F Gregory Buchanan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | |
Collapse
|
25
|
Ma BBY, Bristow RG, Kim J, Siu LL. Combined-modality treatment of solid tumors using radiotherapy and molecular targeted agents. J Clin Oncol 2003; 21:2760-76. [PMID: 12860956 DOI: 10.1200/jco.2003.10.044] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Molecular targeted agents have been combined with radiotherapy (RT) in recent clinical trials in an effort to optimize the therapeutic index of RT. The appeal of this strategy lies in their potential target specificity and clinically acceptable toxicity. DESIGN This article integrates the salient, published research findings into the underlying molecular mechanisms, preclinical efficacy, and clinical applicability of combining RT with molecular targeted agents. These agents include inhibitors of intracellular signal transduction molecules, modulators of apoptosis, inhibitors of cell cycle checkpoints control, antiangiogenic agents, and cyclo-oxygenase-2 inhibitors. RESULTS Molecular targeted agents can have direct effects on the cytoprotective and cytotoxic pathways implicated in the cellular response to ionizing radiation (IR). These pathways involve cellular proliferation, DNA repair, cell cycle progression, nuclear transcription, tumor angiogenesis, and prostanoid-associated inflammation. These pathways can also converge to alter RT-induced apoptosis, terminal growth arrest, and reproductive cell death. Pharmacologic modulation of these pathways may potentially enhance tumor response to RT though inhibition of tumor repopulation, improvement of tumor oxygenation, redistribution during the cell cycle, and alteration of intrinsic tumor radiosensitivity. CONCLUSION Combining RT and molecular targeted agents is a rational approach in the treatment of solid tumors. Translation of this approach from promising preclinical data to clinical trials is actively underway.
Collapse
Affiliation(s)
- Brigette B Y Ma
- Department of Medical Oncology and Hematology, Precess Margaret Hospital, University Health Network, Suite 5-210, 610 University Ave, Toronto, Ontario, Canada M5G 2M9
| | | | | | | |
Collapse
|
26
|
Rüegg C, Zaric J, Stupp R. Non steroidal anti-inflammatory drugs and COX-2 inhibitors as anti-cancer therapeutics: hypes, hopes and reality. Ann Med 2003; 35:476-87. [PMID: 14649330 DOI: 10.1080/07853890310017053] [Citation(s) in RCA: 41] [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] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) and specific inhibitors of cyclooxygenase (COX)-2, are therapeutic groups widely used for the treatment of pain, inflammation and fever. There is growing experimental and clinical evidence indicating NSAIDs and COX-2 inhibitors also have anti-cancer activity. Epidemiological studies have shown that regular use of Aspirin and other NSAIDs reduces the risk of developing cancer, in particular of the colon. Molecular pathology studies have revealed that COX-2 is expressed by cancer cells and cells of the tumor stroma during tumor progression and in response to chemotherapy or radiotherapy. Experimental studies have demonstrated that COX-2 over expression promotes tumorigenesis, and that NSAIDs and COX-2 inhibitors suppress tumorigenesis and tumor progression. Clinical trials have shown that NSAIDs and COX-2 inhibitors suppress colon polyp formation and malignant progression in patients with familial adenomatous polyposis (FAP) syndrome. Recent advances in the understanding of the cellular and molecular mechanisms of the anti-cancer effects of NSAIDs and COX-2 inhibitors have demonstrated that these drugs target both tumor cells and the tumor vasculature. The therapeutic benefits of COX-2 inhibitors in the treatment of human cancer in combination with chemotherapy or radiotherapy are currently being tested in clinical trials. In this article we will review recent advances in the understanding of the anti-tumor mechanisms of these drugs and discuss their potential application in clinical oncology.
Collapse
Affiliation(s)
- Curzio Rüegg
- Centre Pluridisciplinaire d'Oncologie, University of Lausanne Medical School, CH-1011 Lausanne, Switzerland.
| | | | | |
Collapse
|
27
|
Gonzalez-Angulo AM, Fuloria J, Prakash O. Cyclooxygenase 2 inhibitors and colon cancer. Ochsner J 2002; 4:176-179. [PMID: 22822342 PMCID: PMC3399281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
|