1
|
Verdina A, Garufi A, D’Orazi V, D’Orazi G. HIPK2 in Colon Cancer: A Potential Biomarker for Tumor Progression and Response to Therapies. Int J Mol Sci 2024; 25:7678. [PMID: 39062921 PMCID: PMC11277226 DOI: 10.3390/ijms25147678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Colon cancer, one of the most common and fatal cancers worldwide, is characterized by stepwise accumulation of specific genetic alterations in tumor suppressor genes or oncogenes, leading to tumor growth and metastasis. HIPK2 (homeodomain-interacting protein kinase 2) is a serine/threonine protein kinase and a "bona fide" oncosuppressor protein. Its activation inhibits tumor growth mainly by promoting apoptosis, while its inactivation increases tumorigenicity and resistance to therapies of many different cancer types, including colon cancer. HIPK2 interacts with many molecular pathways by means of its kinase activity or transcriptional co-repressor function modulating cell growth and apoptosis, invasion, angiogenesis, inflammation and hypoxia. HIPK2 has been shown to participate in several molecular pathways involved in colon cancer including p53, Wnt/β-catenin and the newly identified nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2). HIPK2 also plays a role in tumor-host interaction in the tumor microenvironment (TME) by inducing angiogenesis and cancer-associated fibroblast (CAF) differentiation. The aim of this review is to assess the role of HIPK2 in colon cancer and the underlying molecular pathways for a better understanding of its involvement in colon cancer carcinogenesis and response to therapies, which will likely pave the way for novel colon cancer therapies.
Collapse
Affiliation(s)
- Alessandra Verdina
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.V.); (A.G.)
| | - Alessia Garufi
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.V.); (A.G.)
| | - Valerio D’Orazi
- Department of Surgery, Sapienza University, 00185 Rome, Italy;
| | - Gabriella D’Orazi
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.V.); (A.G.)
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
2
|
Willis JA, Reyes-Uribe L, Chang K, Lipkin SM, Vilar E. Immune Activation in Mismatch Repair-Deficient Carcinogenesis: More Than Just Mutational Rate. Clin Cancer Res 2020; 26:11-17. [PMID: 31383734 PMCID: PMC6942620 DOI: 10.1158/1078-0432.ccr-18-0856] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/26/2019] [Accepted: 07/31/2019] [Indexed: 01/08/2023]
Abstract
Mismatch repair (MMR)-deficient colorectal cancers (dMMR colorectal cancer) are characterized by the expression of highly immunogenic neoantigen peptides, which stimulate lymphocytic infiltration as well as upregulation of inflammatory cytokines. These features are key to understanding why immunotherapy (specifically PD-1 and/or CTLA-4 checkpoint blockade) has proved to be highly effective for the treatment of patients with advanced dMMR colorectal cancer. Importantly, preclinical studies also suggest that this correlation between potent tumor neoantigens and the immune microenvironment is present in early (premalignant) stages of dMMR colorectal tumorigenesis as well, even in the absence of a high somatic mutation burden. Here, we discuss recent efforts to characterize how neoantigens and the tumor immune microenvironment coevolve throughout the dMMR adenoma-to-carcinoma pathway. We further highlight how this preclinical evidence forms the rational basis for developing novel immunotherapy-based colorectal cancer prevention strategies for patients with Lynch syndrome.
Collapse
Affiliation(s)
- Jason A Willis
- Hematology and Oncology Fellowship Program, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kyle Chang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Steven M Lipkin
- Department of Medicine, Weill-Cornell Medical College, Cornell University, New York, New York
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
3
|
Han Y, Cai H, Ma L, Ding Y, Tan X, Liu Y, Su T, Yu Y, Chang W, Zhang H, Fu C, Cao G. Nuclear orphan receptor NR4A2 confers chemoresistance and predicts unfavorable prognosis of colorectal carcinoma patients who received postoperative chemotherapy. Eur J Cancer 2013; 49:3420-30. [PMID: 23809767 DOI: 10.1016/j.ejca.2013.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/25/2013] [Accepted: 06/05/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND NR4A2, an orphan nuclear receptor essential in neuron generation, has been recently linked to inflammatory and metabolic pathways of colorectal carcinoma (CRC). However, the effects of NR4A2 on chemo-resistance and postoperative prognosis of CRC remain unknown. METHODS NR4A2 was transfected into CRC cells to investigate its effects on chemo-resistance to 5-fluorouracil and oxaliplatin and chemotherapeutics-induced apoptosis. We also investigated prostaglandin E2 (PGE2)-induced NR4A2 expression and its effect on chemo-resistance. Tissue microarrays including 51 adenoma, 14 familial adenomatous polyposis with CRC, 17 stage IV CRC with adjacent mucosa and 682 stage I-III CRC specimens were examined immunohistochemically for NR4A2 expression. Median follow-up time for stage I-III CRC patients was 53 months. RESULTS Ectopic expression of NR4A2 increased the chemo-resistance, and attenuated the chemotherapeutics-induced apoptosis. Transient treatment of PGE2 significantly up-regulated NR4A2 expression via protein kinase A pathway and increased the chemo-resistance. NR4A2 expression in epithelials consecutively increased from adenoma, adjacent mucosa to CRC (P(trend)<0.001). In multivariate Cox regression analyses, high NR4A2 expression in cancer nuclei (immunoreactive score ≥ 4) significantly predicted a shorter disease-specific survival (DSS) of CRC patients (hazard ratio [HR]=1.88, P=0.024). High NR4A2 expression specifically predicted a shorter DSS of colon cancer patients (dichotomisation, HR=2.55, log-rank test P=0.011), especially for those who received postoperative 5-fluorouracil/leucovorin plus oxaliplatin (FOLFOX) chemotherapy (3-score range, HR=1.86, log-rank test P=0.020). CONCLUSION High expression of NR4A2 in CRC cells confers chemo-resistance, attenuates chemotherapeutics-induced apoptosis, and predicts unfavorable prognosis of colon cancer patients, especially for those who received postoperative chemotherapy. NR4A2 may be prognostic and predictive for colon cancer.
Collapse
Affiliation(s)
- Yifang Han
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Han YF, Cao GW. Role of nuclear receptor NR4A2 in gastrointestinal inflammation and cancers. World J Gastroenterol 2012; 18:6865-73. [PMID: 23322982 PMCID: PMC3531668 DOI: 10.3748/wjg.v18.i47.6865] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/27/2012] [Accepted: 09/12/2012] [Indexed: 02/06/2023] Open
Abstract
NR4A2 is a transcription factor belonging to the steroid orphan nuclear receptor superfamily. It was originally considered to be essential in the generation and maintenance of dopaminergic neurons, and associated with neurological disorders such as Parkinson’s disease. Recently, NR4A2 has been found to play a critical role in some inflammatory diseases and cancer. NR4A2 can be efficiently trans-activated by some proinflammatory cytokines, such as tumor necrosis factor-α, interleukin-1β, and vascular endothelial growth factor (VEGF). The nuclear factor-κB signaling pathway serves as a principal regulator of inducible NR4A expression in immune cells. NR4A2 can trans-activate Foxp3, a hallmark specifically expressed in regulatory T (Treg) cells, and plays a critical role in the differentiation, maintenance, and function of Treg cells. NR4A2 in T lymphocytes is pivotal for Treg cell induction and suppression of aberrant induction of Th1 under physiological and pathological conditions. High density of Foxp3+ Treg cells is significantly associated with gastrointestinal inflammation, tumor immune escape, and disease progression. NR4A2 is produced at high levels in CD133+ colorectal carcinoma (CRC) cells and significantly upregulated by cyclooxygenase-2-derived prostaglandin E2 in a cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)-dependent manner in CRC cells. The cAMP/PKA signaling pathway is the common pathway of NR4A2-related inflammation and cancer. NR4A2 trans-activates osteopontin, a direct target of the Wnt/β-catenin pathway associated with CRC invasion, metastasis, and poor prognosis. Knockdown of endogenous NR4A2 expression attenuates VEGF-induced endothelial cell proliferation, migration and in vivo angiogenesis. Taken together, NR4A2 emerges as an important nuclear factor linking gastrointestinal inflammation and cancer, especially CRC, and should serve as a candidate therapeutic target for inflammation-related gastrointestinal cancer.
Collapse
|
5
|
Garufi A, Pistritto G, Ceci C, Di Renzo L, Santarelli R, Faggioni A, Cirone M, D’Orazi G. Targeting COX-2/PGE(2) pathway in HIPK2 knockdown cancer cells: impact on dendritic cell maturation. PLoS One 2012; 7:e48342. [PMID: 23144866 PMCID: PMC3492329 DOI: 10.1371/journal.pone.0048342] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/24/2012] [Indexed: 02/04/2023] Open
Abstract
Background Homeodomain-interacting protein kinase 2 (HIPK2) is a multifunctional protein that exploits its kinase activity to modulate key molecular pathways in cancer to restrain tumor growth and induce response to therapies. For instance, HIPK2 knockdown induces upregulation of oncogenic hypoxia-inducible factor-1 (HIF-1) activity leading to a constitutive hypoxic and angiogenic phenotype with increased tumor growth in vivo. HIPK2 inhibition, therefore, releases pathways leading to production of pro-inflammatory molecules such as vascular endothelial growth factor (VEGF) or prostaglandin E2 (PGE2). Tumor-produced inflammatory mediators other than promote tumour growth and vascular development may permit evasion of anti-tumour immune responses. Thus, dendritic cells (DCs) dysfunction induced by tumor-produced molecules, may allow tumor cells to escape immunosurveillance. Here we evaluated the molecular mechanism of PGE2 production after HIPK2 depletion and how to modulate it. Methodology/Principal findings We show that HIPK2 knockdown in colon cancer cells resulted in cyclooxygenase-2 (COX-2) upregulation and COX-2-derived PGE2 generation. At molecular level, COX-2 upregulation depended on HIF-1 activity. We previously reported that zinc treatment inhibits HIF-1 activity. Here, zinc supplementation to HIPK2 depleted cells inhibited HIF-1-induced COX-2 expression and PGE2/VEGF production. At translational level, while conditioned media of both siRNA control and HIPK2 depleted cells inhibited DCs maturation, conditioned media of only zinc-treated HIPK2 depleted cells efficiently restored DCs maturation, seen as the expression of co-stimulatory molecules CD80 and CD86, cytokine IL-10 release, and STAT3 phosphorylation. Conclusion/Significance These findings show that: 1) HIPK2 knockdown induced COX-2 upregulation, mostly depending on HIF-1 activity; 2) zinc treatment downregulated HIF-1-induced COX-2 and inhibited PGE2/VEGF production; and 3) zinc treatment of HIPK2 depleted cells restored DCs maturation.
Collapse
Affiliation(s)
- Alessia Garufi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppa Pistritto
- Department of Neuroscience, Section of Pharmacology, University “Tor Vergata”, Rome, Italy
| | - Claudia Ceci
- Department of Neuroscience, Section of Pharmacology, University “Tor Vergata”, Rome, Italy
| | - Livia Di Renzo
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Institute Pasteur-Foundation Cenci Bolognetti, “Sapienza” University, Rome, Italy
- * E-mail: (GD); (MC)
| | - Gabriella D’Orazi
- Department of Experimental Oncology, Molecular Oncogenesis Laboratory, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio”, Chieti, Italy
- * E-mail: (GD); (MC)
| |
Collapse
|
6
|
Coghill AE, Newcomb PA, Poole EM, Hutter CM, Makar KW, Duggan D, Potter JD, Ulrich CM. Genetic variation in inflammatory pathways is related to colorectal cancer survival. Clin Cancer Res 2011; 17:7139-47. [PMID: 21976545 PMCID: PMC3218294 DOI: 10.1158/1078-0432.ccr-11-1134] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE Prognosis of patients with colorectal cancer (CRC) is associated with systemic inflammation, and anti-inflammatory drugs can reduce both CRC incidence and mortality. Genetic variation in proinflammatory pathways can affect an individual's CRC risk. However, few studies have investigated the prognostic importance of this genetic variation in CRC patients. EXPERIMENTAL DESIGN We investigated the association between CRC survival and genetic variation in proinflammatory pathways among patients from the Puget Sound Surveillance Epidemiology and End Results registry. Single-nucleotide polymorphisms were genotyped in five genes (PTGS-1, PTGS-2, MRP4, NFκB, and IκBKβ). Vital status was ascertained through linkage to the National Death Index. Cox proportional hazards regression was used to calculate HRs and 95% confidence intervals (CI). The false discovery rate method of Benjamini and Hochberg was applied to address multiple testing. RESULTS Four PTGS-1 variants were associated with CRC survival. One, G>A intron 9 (rs1213266), was associated with approximately 50% lower CRC mortality (HR(AA/AG vs. GG) = 0.48; 95% CI, 0.25-0.93). Three variants, including L237M, resulted in significantly elevated CRC mortality risk, with HRs ranging from approximately 1.5 to 2.0. Two variants in IκBKβ, including R526Q, were significantly associated with CRC survival. Correction for multiple testing indicated that variants in both PTGS-1 and IκBKβ are reproducibly associated with CRC survival. CONCLUSION Our findings suggest that genetic variation in proinflammatory pathways may be important for CRC prognosis. This investigation represents one of the first descriptions of the relationship between inherited polymorphisms and mortality in CRC patients and provides a starting point for further research.
Collapse
Affiliation(s)
- Anna E Coghill
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Pink RC, Bailey TA, Iputo JE, Sammon AM, Woodman AC, Carter DRF. Molecular basis for maize as a risk factor for esophageal cancer in a South African population via a prostaglandin E2 positive feedback mechanism. Nutr Cancer 2011; 63:714-21. [PMID: 21667399 DOI: 10.1080/01635581.2011.570893] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The incidence of squamous cancer of the esophagus varies up to a hundredfold in different regions of the world. In Transkei, South Africa, a particularly high incidence of the disease is observed. We have previously proposed an association between a maize-rich diet and elevated levels of intragastric prostaglandin E2 production (PGE(2)). Here we investigate the molecular mechanisms by which a high-maize diet could lead to increased incidence of squamous cancer of the esophagus. We confirm that levels of PGE(2) are high (606.8 pg/ml) in the gastric fluid of individuals from Transkei. We also show that treatment of esophageal cells with linoleic acid, which is found at high levels in maize and is a precursor to PGE(2), leads to increased cell proliferation. Similarly, treatment of cells with PGE(2) or with gastric fluid from Transkeians also leads to increased proliferation. Our data suggest that the high levels of PGE(2) associated with a maize-rich diet stimulate cell division and induce the enzyme COX 2, resulting in a positive feedback mechanism that predisposes the esophagus to carcinoma.
Collapse
Affiliation(s)
- Ryan C Pink
- Cranfield Health, Cranfield University, Cranfield, Bedfordshire, United Kingdom.
| | | | | | | | | | | |
Collapse
|
8
|
Coghill AE, Newcomb PA, Chia VM, Zheng Y, Wernli KJ, Passarelli MN, Potter JD. Pre-diagnostic NSAID use but not hormone therapy is associated with improved colorectal cancer survival in women. Br J Cancer 2011; 104:763-8. [PMID: 21304527 PMCID: PMC3048198 DOI: 10.1038/sj.bjc.6606041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Non-steroidal anti-inflammatory drugs (NSAIDs) and hormone therapy (HT) independently decrease the risk of colorectal cancer. However, their role in altering survival after a colorectal cancer diagnosis is not well established. Methods: We examined the association between the use of these common medications before diagnosis and colorectal cancer survival among women in western Washington State diagnosed with incident colorectal cancer from 1997 to 2002. Cases were ascertained using the Surveillance, Epidemiology and End Results cancer registry; mortality follow-up was completed through linkages to the National Death Index. Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Results: We observed no overall association between colorectal cancer survival and pre-diagnostic NSAID use. However, when stratified by tumour sub-site, NSAID use was associated with a reduced risk of colorectal cancer mortality for women diagnosed with proximal (HR=0.55; 95% CI: 0.32–0.92), but not distal or rectal (HR=1.32; 95% CI: 0.83–2.10) tumours. The usage of HT was not associated with colorectal cancer survival overall or by tumour sub-site. Conclusion: Usage of NSAIDs before diagnosis may be associated with improved colorectal cancer survival among women diagnosed with proximal tumours. The usage of HT does not appear to have a function in altering colorectal cancer mortality.
Collapse
Affiliation(s)
- A E Coghill
- Fred Hutchinson Cancer Research Center, Cancer Prevention Program, 1100 Fairview Ave N, M4-B402, Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Knopfová L, Smarda J. The use of Cox-2 and PPARγ signaling in anti-cancer therapies. Exp Ther Med 2010; 1:257-264. [PMID: 22993537 DOI: 10.3892/etm_00000040] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/02/2009] [Indexed: 02/06/2023] Open
Abstract
Increased production of the pro-inflammatory enzyme cyclooxygenase-2 (Cox-2) and altered expression and activity of peroxisome proliferator-activated receptor γ (PPARγ) have been observed in many malignancies. Both the PPARγ ligands and the Cox-2 inhibitors possess anti-inflammatory and anti-neoplastic effects in vitro and have been assessed for their therapeutic potential in several pre-clinical and clinical studies. Recently, multiple interactions between PPARγ and Cox-2 signaling pathways have been revealed. Understanding of the cross-talk between PPARγ and Cox-2 might provide important novel strategies for the effective treatment and/or prevention of cancer. This article summarizes recent achievements involving the functional interactions between the PPARγ and Cox-2 signaling pathways and discusses the implications of such interplay for clinical use.
Collapse
Affiliation(s)
- Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
| | | |
Collapse
|
10
|
Grau MV, Sandler RS, McKeown-Eyssen G, Bresalier RS, Haile RW, Barry EL, Ahnen DJ, Gui J, Summers RW, Baron JA. Nonsteroidal anti-inflammatory drug use after 3 years of aspirin use and colorectal adenoma risk: observational follow-up of a randomized study. J Natl Cancer Inst 2009; 101:267-76. [PMID: 19211442 DOI: 10.1093/jnci/djn484] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Frequent use of nonsteroidal anti-inflammatory drugs (NSAIDs) has been shown to reduce the risk of colorectal adenomas in randomized trials. We examined the persistence of the protective effect after the cessation of randomized aspirin treatment and whether it is affected by the duration and frequency of subsequent NSAID use. METHODS We used data from the Aspirin/Folate Polyp Prevention Study (AFPPS), in which 1121 subjects were randomly assigned to receive placebo or aspirin (81 or 325 mg/d) for 3 years. After the end of treatment and a follow-up colonoscopy, AFPPS participants were invited to remain under follow-up until their next surveillance colonoscopies, scheduled 3-5 years later. Information regarding use of NSAIDs during posttreatment follow-up was gathered periodically via questionnaires. Average weekly NSAID use was classified as sporadic (<2 days per week), moderate (2 to <4 days per week), or frequent (>or=4 days per week). The analysis was stratified according to randomized aspirin groups and posttreatment NSAID use; placebo subjects who later were sporadic NSAID users formed the reference group. The primary outcomes were all adenomas and advanced lesions. Adjusted relative risks and 95% confidence intervals were computed with generalized linear models. All statistical tests were two-sided. RESULTS A total of 850 subjects underwent a posttreatment colonoscopy, on average 4 years after the end of study treatment. The protective effect of 81 mg of aspirin for colorectal adenomas persisted with continued posttreatment NSAID use. The risk of any adenoma among frequent NSAID users was 26.8% vs 39.9% among placebo subjects who later used NSAIDs sporadically (adjusted relative risk = 0.62, 95% confidence interval [CI] = 0.39 to 0.98; P(trend) with NSAID use frequency = .03). The unadjusted absolute risk reduction was 13.1 percentage points (95% CI = -0.3 to 26.5 percentage points) (P = .07). Results for 325 mg of aspirin were similar, although not statistically significant. For advanced lesions, small numbers of endpoints limited the analysis, but findings among subjects randomly assigned to 81 mg of aspirin suggested a protective association regardless of posttreatment NSAID use. CONCLUSION Long-term and frequent use of NSAIDs may enhance the chemopreventive effect of aspirin against colorectal neoplasia.
Collapse
Affiliation(s)
- Maria V Grau
- Departments of Community and Family Medicine, Dartmouth Medical School, Hanover, NH, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Trojian TH, Mody K, Chain P. Exercise and colon cancer: primary and secondary prevention. Curr Sports Med Rep 2007; 6:120-4. [PMID: 17376341 DOI: 10.1007/bf02941153] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Many studies have demonstrated the effects of exercise on both primary and secondary prevention of colon cancer. Exercise appears to have a dose-response reduction in the rate of colon cancer. The mechanism by which exercise provides this benefit is not known, but increase in insulin-like growth factor-binding protein and reduction of prostaglandins appear to be the likely cause. Once a person develops colon cancer the benefits of exercise appear to continue both by increasing quality of life and reducing cancer-specific and overall mortality.
Collapse
Affiliation(s)
- Thomas H Trojian
- University of Connecticut Health Center/Saint Francis Hospital and Medical Center, Department of Family Medicine, Hartford, CT 06105, USA.
| | | | | |
Collapse
|
12
|
|