Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Eur J Gynaecol Oncol ; 37(1): 36-40, 2016.
Article in English | MEDLINE | ID: mdl-27048107

ABSTRACT

PURPOSE OF INVESTIGATION: To investigate the presence of 15-lipoxygenase-1 (15-LOX-1) expression and its potential role in the pathogenesis of endometrial hyperplasia and endometrial adenocarcinomas. MATERIALS AND METHODS: The authors investigated the presence of 15-LOX-1 expression in samples from patients diagnosed with normal endometrium (n = 12), endometrial hyperplasia (n = 12), and endometrial cancer (n = 12). The immunohistochemical stainings were scored by three independent pathologists. A Western blot of 15- LOX-1 determined the presence of protein expression in normal endometrium. A Kolmogorov-Smirnov test was used to evaluate the data's distribution pattern. For pairwise comparisons of the combined scores between groups, the Mann-Whitney U test was used. RESULTS: Based on the combined scores for 15-LOX-1 expression, strong immunochemistry staining was observed in samples diagnosed with normal endometrium. There was a significant difference in 15-LOX-1 expression between normal endometrium and endometrial adenocarcinoma (p = 0.03). Comparing tissues from normal endometrium and endometrial hyperplasia, there was a decline in the expression from normal endometrium to endometrial hyperplasia. However, the difference was not statistically significant. CONCLUSION: The present results show that a decrease of 15-LOX-1 expression in the endometrial tumorigenesis process, starting from normal endometrium to hyperplasia and endometrial cancer, might be a trigger. Further studies are required to determine its potential use as a marker in a larger randomized multicenter study.


Subject(s)
Arachidonate 15-Lipoxygenase/physiology , Endometrial Hyperplasia/etiology , Endometrial Neoplasms/etiology , Adult , Aged , Arachidonate 15-Lipoxygenase/analysis , Endometrial Hyperplasia/enzymology , Endometrial Neoplasms/enzymology , Female , Humans , Immunohistochemistry , Middle Aged
2.
Ann Oncol ; 27(6): 1068-1074, 2016 06.
Article in English | MEDLINE | ID: mdl-27045102

ABSTRACT

BACKGROUND: Incorporation of multiple enrichment biomarkers into prospective clinical trials is an active area of investigation, but the factors that determine clinical trial enrollment following a molecular prescreening program have not been assessed. PATIENTS AND METHODS: Patients with 5-fluorouracil-refractory metastatic colorectal cancer at the MD Anderson Cancer Center were offered screening in the Assessment of Targeted Therapies Against Colorectal Cancer (ATTACC) program to identify eligibility for companion phase I or II clinical trials with a therapy targeted to an aberration detected in the patient, based on testing by immunohistochemistry, targeted gene sequencing panels, and CpG island methylation phenotype assays. RESULTS: Between August 2010 and December 2013, 484 patients were enrolled, 458 (95%) had a biomarker result, and 157 (32%) were enrolled on a clinical trial (92 on biomarker-selected and 65 on nonbiomarker selected). Of the 458 patients with a biomarker result, enrollment on biomarker-selected clinical trials was ninefold higher for predefined ATTACC-companion clinical trials as opposed to nonpredefined biomarker-selected clinical trials, 17.9% versus 2%, P < 0.001. Factors that correlated positively with trial enrollment in multivariate analysis were higher performance status, older age, lack of standard of care therapy, established patient at MD Anderson, and the presence of an eligible biomarker for an ATTACC-companion study. Early molecular screening did result in a higher rate of patients with remaining standard of care therapy enrolling on ATTACC-companion clinical trials, 45.1%, in contrast to nonpredefined clinical trials, 22.7%; odds ratio 3.1, P = 0.002. CONCLUSIONS: Though early molecular prescreening for predefined clinical trials resulted in an increase rate of trial enrollment of nonrefractory patients, the majority of patients enrolled on clinical trials were refractory to standard of care therapy. Within molecular prescreening programs, tailoring screening for preidentified and open clinical trials, temporally linking screening to treatment and optimizing both patient and physician engagement are efforts likely to improve enrollment on biomarker-selected clinical trials. CLINICAL TRIALS NUMBER: The study NCT number is NCT01196130.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , CpG Islands/genetics , Eligibility Determination , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Staging , Patient Selection
3.
Invest New Drugs ; 33(4): 977-84, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26062928

ABSTRACT

BACKGROUND: Src has a critical role in tumor cell migration and invasion. Increased Src activity has been shown to correlate with disease progression and poor prognosis, suggesting Src could serve as a therapeutic target for kinase inhibition. Saracatinib (AZD0530) is a novel selective oral Src kinase inhibitor. METHODS: Metastatic colorectal cancer patients who had received one prior treatment and had measurable disease were enrolled in this phase 2 study. Saracatinib was administered at 175 mg by mouth daily for 28 day cycles until dose-limiting toxicity or progression as determined by staging every 2 cycles. The primary endpoint was improvement in 4 month progression-free survival. Design of Thall, Simon, and Estey was used to monitor proportion of patients that were progression free at 4 months. The trial was opened with plan to enroll maximum of 35 patients, with futility assessment every 10 patients. RESULTS: A total of 10 patients were enrolled between January and November 2007. Further enrollment was stopped due to futility. Median progression-free survival was 7.9 weeks, with all 10 patients showing disease progression following radiographic imaging. Median overall survival was 13.5 months. All patients were deceased by time of analysis. Observed adverse events were notable for a higher than expected number of patients with grade 3 hypophosphatemia (n = 5). CONCLUSION: Saracatinib is a novel oral Src kinase inhibitor that was well tolerated but failed to meet its primary endpoint of improvement in 4 month progression-free survival as a single agent in previously treated metastatic colorectal cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Benzodioxoles/therapeutic use , Colorectal Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Adenocarcinoma/blood , Adenocarcinoma/pathology , Aged , Antineoplastic Agents/adverse effects , Benzodioxoles/adverse effects , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinazolines/adverse effects , Vascular Endothelial Growth Factor A/blood , src-Family Kinases/antagonists & inhibitors
4.
Oncogene ; 28(12): 1496-505, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19198625

ABSTRACT

15-Lipoxygenase-1 (15-LOX-1) is transcriptionally silenced in cancer cells, and its transcription reactivation (for example, through histone deacetylase inhibitors (HDACIs)) restores apoptosis to cancer cells. However, the exact mechanism underlying 15-LOX-1 transcription reactivation in cancer cells is still undefined. Therefore, we evaluated the critical mechanisms required for 15-LOX-1 transcription reactivation in colon cancer cells. Specific HDAC1 and HDAC2 inhibition activated 15-LOX-1 transcription. 15-LOX-1 transcription was repressed through transcription repressor complex recruitment in the region of -120 to -391 of the 15-LOX-1 promoter. The nucleosome remodeling and histone deacetylase (NuRD) repression complex was recruited to this region. Depsipeptide significantly reduced the recruitment of NuRD key components (for example, metastasis-associated protein 1 (MTA1) and HDAC1) to the 15-LOX-1 promoter before 15-LOX-1 transcriptional activation. Knock down of NuRD key components (for example, MTA1 and HDAC1) by small interfering RNA (siRNA) activated 15-LOX-1 transcription, as measured by luciferase reporter assays in stably transfected SW480 cells with the 15-LOX-1 promoter construct of the -391, but not the -120 region. Relative to expression in normal tissue, MTA1 expression in colorectal cancer mucosa from colorectal cancer patients was negatively related to 15-LOX-1 expression. Thus, our results show that NuRD contributes to 15-LOX-1 transcription suppression in colon cancer cells and that HDACIs can inhibit NuRD recruitment to a promoter to activate gene transcription, as in the case of 15-LOX-1.


Subject(s)
Arachidonate 15-Lipoxygenase/genetics , Colonic Neoplasms/enzymology , Histone Deacetylases/physiology , Repressor Proteins/physiology , Aged , Aged, 80 and over , Cell Line, Tumor , Colonic Neoplasms/pathology , Female , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Humans , Male , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Middle Aged , Promoter Regions, Genetic , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Trans-Activators , Transcription, Genetic
5.
Oncogene ; 25(8): 1225-41, 2006 Feb 23.
Article in English | MEDLINE | ID: mdl-16288226

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are transcription factors that strongly influence molecular events in normal and cancer cells. PPAR-beta/delta (PPAR-b/d) overexpression suppresses the activity of PPAR-gamma (PPAR-g) and PPAR-alpha. This interaction has been questioned, however, by studies with synthetic ligands of PPARs in PPAR-b/d-null cells, and it is not known whether an interaction between PPAR-b/d and PPAR-g exists, especially in relation to the signaling by natural PPAR ligands. Oxidative metabolites of linoleic and arachidonic acids are natural ligands of PPARs. 13-S-hydroxyoctadecadienoic acid (13-S-HODE), the main product of 15-lipoxygenase-1 (15-LOX-1) metabolism of linoleic acid, downregulates PPAR-b/d. We tested (a) whether PPAR-b/d expression modulates PPAR-g activity in experimental models of the loss and gain of PPAR-b/d function in colon cancer cells and (b) whether 15-LOX-1 formation of 13-S-HODE influences the interaction between PPAR-b/d and PPAR-g. We found that (a) 15-LOX-1 formation of 13-S-HODE promoted PPAR-g activity, (b) PPAR-b/d expression suppressed PPAR-g activity in models of both loss and gain of PPAR-b/d function, (c) 15-LOX-1 activated PPAR-g by downregulating PPAR-b/d, and (d) 15-LOX-1 expression induced apoptosis in colon cancer cells via modulating PPAR-b/d suppression of PPAR-g. These findings elucidate a novel mechanism of the signaling by natural ligands of PPARs, which involves modulating the interaction between PPAR-b/d and PPAR-g.


Subject(s)
Colorectal Neoplasms/drug therapy , Linoleic Acid/pharmacology , PPAR delta/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Adenoviridae/genetics , Arachidonate 15-Lipoxygenase/metabolism , Colorectal Neoplasms/metabolism , Down-Regulation , Humans , Linoleic Acids/metabolism , Oxidation-Reduction , PPAR delta/antagonists & inhibitors , PPAR gamma/antagonists & inhibitors , PPAR-beta/antagonists & inhibitors
6.
Melanoma Res ; 12(5): 429-34, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12394183

ABSTRACT

12-Lipoxygenase (12-LOX), through its metabolite 12( )-hydroxyeicosatetraenoic acid [12( )-HETE], has been demonstrated to play a pivotal role in experimental melanoma invasion and metastasis, and 12-LOX expression may be important in early human melanoma carcinogenesis. We have studied the differences in 12-LOX protein expression during the progression of melanoma from human melanocytic cells to benign and dysplastic naevi to malignant metastatic disease. 12-LOX expression was determined in normal human skin melanocytes and in melanocytes found in compound naevi, dysplastic naevi and melanomas using a platelet-type 12-LOX antibody with a diaminobenzidine immunoperoxidase system detection system and was quantified using the analysis software NIH Image 1.62. Mean cellular pixel densities for 12-LOX staining ( = 50 cells/histological type) were unchanged in compound naevi ( = 0.14) and were increased in dysplastic naevi and melanomas compared with normal skin melanocytes ( = 0.03 and = 0.01, respectively). Similarly, melanomas had higher levels of expression compared with dysplastic naevi ( = 0.03). 12-LOX expression was significantly different between compound naevus and dysplastic naevus melanocytes ( = 0.01). These data suggest that 12-LOX may be an important novel marker for cancer progression within the melanoma system, and therefore could be a useful biomarker and therapeutic target for melanoma chemoprevention.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Melanoma/diagnosis , Melanoma/enzymology , Skin Neoplasms/diagnosis , Skin Neoplasms/enzymology , Blood Platelets/enzymology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Melanocytes/enzymology , Nevus/enzymology , Precancerous Conditions
7.
Br J Cancer ; 86(2): 226-32, 2002 Jan 21.
Article in English | MEDLINE | ID: mdl-11870511

ABSTRACT

The clinical and economic impacts of monitoring cardiac function in patients given doxorubicin have yet to be determined, especially in relation to patient age, cumulative doxorubicin dose, and the relative efficacies of doxorubicin-based vs alternative regimens. We developed a decision analysis model that includes these factors to estimate the incremental survival benefit and cost-effectiveness of using multiple gated acquisition scans to measure left-ventricular ejection fraction before and during doxorubicin chemotherapy. Probability distributions for the incidences of abnormal left-ventricular ejection fraction findings and congestive heart failure were derived from a retrospective review of 227 consecutive cases at The University of Michigan Medical Center and published findings. Multiple gated acquisition-scan monitoring minimally improved the probability of 5-year survival (<1.5% in the base--case scenario). For patients who received up to 350 mg m(-2) of doxorubicin, multiple gated acquisition-scan screening had an incremental cost of $425 402 per life saved for patients between the ages of 15--39. This incremental cost markedly decreased to $138 191, for patients between the ages of 40--59, and to $86 829 for patients older than 60 years. The small gain in 5-year survival probability secondary to multiple gated acquisition scan monitoring doubled for all age groups when the average cumulative dose for doxorubicin reached 500 mg m(-2). Variations in the cure rate differences between the doxorubicin and alternative regimens had insignificant effects on the improvement in 5-year survival rates from multiple gated acquisition-scan screening. The use of multiple gated acquisition scans for pretreatment screening appears to be more cost-effective for patients who are 40 years or older, when cumulative doxorubicin dose is 350 mg m(-2) or less.


Subject(s)
Antineoplastic Agents/adverse effects , Doxorubicin/adverse effects , Gated Blood-Pool Imaging/economics , Health Care Costs/statistics & numerical data , Heart Failure/diagnostic imaging , Stroke Volume , Adolescent , Adult , Aged , Cost-Benefit Analysis , Decision Support Techniques , Female , Heart Failure/chemically induced , Humans , Male , Middle Aged , Neoplasms/drug therapy , Predictive Value of Tests , Retrospective Studies , Survival Analysis , Ventricular Function, Left
8.
J Clin Oncol ; 19(22): 4202-8, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11709563

ABSTRACT

PURPOSE: The primary objective of this phase I trial was to determine the maximum-tolerated dose of radiation that could be delivered to the primary tumor concurrent with full-dose gemcitabine in patients with advanced pancreatic cancer. PATIENTS AND METHODS: Thirty seven patients with unresectable (n = 34) or incompletely resected pancreatic cancer (n = 3) were treated. Gemcitabine was administered as a 30-minute intravenous infusion at a dose of 1,000 mg/m(2) on days 1, 8, and 15 of a 28-day cycle. Radiation therapy was initiated on day 1 and directed at the primary tumor alone, without prophylactic nodal coverage. The starting radiation dose was 24 Gy in 1.6-Gy fractions. Escalation was achieved by increasing the fraction size in increments of 0.2 Gy, keeping the duration of radiation constant at 3 weeks. A second cycle of gemcitabine alone was intended after a 1-week rest. RESULTS: Two of six assessable patients experienced dose-limiting toxicity at the final planned dose level of the trial (42 Gy in 2.8-Gy fractions), one with grade 4 vomiting and one with gastric/duodenal ulceration. Two additional patients at this dose level experienced late gastrointestinal toxicity that required surgical management. CONCLUSION: The final dose investigated (42 Gy) is not recommended for further study considering the occurrence of both acute and late toxicity. However, a phase II trial of this novel gemcitabine-based chemoradiotherapy approach, at a radiation dose of 36 Gy in 2.4-Gy fractions, is recommended on the basis of tolerance, patterns of failure, and survival data.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Follow-Up Studies , Humans , Infusions, Intravenous , Maximum Tolerated Dose , Middle Aged , Pancreatic Neoplasms/pathology , Radiation Dosage , Tomography, X-Ray Computed , Treatment Outcome , Gemcitabine
10.
Cancer Res ; 61(17): 6307-12, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522616

ABSTRACT

New studies of the relationship between polyunsaturated fatty acid metabolismand carcinogenesis have led to novel molecular targets for cancer chemoprevention research. These targets include procarcinogenic lipoxygenases (LOXs), including 5-, 8-, and 12-LOX, and anticarcinogenic LOXs, including 15-LOX-1 and possibly 15-LOX-2. Recent studies indicate that 15-LOX-1 is down-regulated in colorectal cancer cells and that the ability of nonsteroidal anti-inflammatory drugs, a class of clinically active cancer chemopreventive agents, to induce apoptosis and growth inhibition in these cells was dependent on the induction of 15-LOX-1 and its metabolic product 13-S-hydroxyoctadecadienoic acid. Consistent with the colorectal studies, 15-LOX very recently has shown anticarcinogenic activity in esophageal and prostatic carcinogenesis. Inhibitors of other LOXs (e.g., 5-LOX) have preclinical anticarcinogenic activity and are being developed for clinical chemoprevention study. These and other LOX data led us to propose that the various LOX pathways exist in a dynamic balance that shifts during carcinogenesis toward 5-, 8-, and 12-LOX (and cyclooxygenase-2) and away from 15-LOX. A novel approach for cancer chemoprevention would involve LOX modulators, i.e., agents that can induce the anticarcinogenic and/or inhibit the procarcinogenic LOXs, thereby shifting the balance of LOX activities from procarcinogenic to anticarcinogenic metabolism of polyunsaturated fatty acids.


Subject(s)
Lipoxygenase/metabolism , Neoplasms/enzymology , Neoplasms/prevention & control , Animals , Chemoprevention/methods , Humans , Isoenzymes/metabolism
11.
Cancer Res ; 61(12): 4879-84, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406566

ABSTRACT

In previous studies, we have found that expression of 15-lipoxygenase-1 (15-LOX-1) and its main product, 13-S-hydroxyoctadecadienoic acid, are decreased in human colorectal cancers and that nonsteroidal anti-inflammatory drugs (NSAIDs) can therapeutically induce 15-LOX-1 expression to trigger apoptosis in human colorectal cancer cells. NSAIDs similarly induce apoptosis in esophageal cancer cells, although the mechanisms of these effects remain to be defined. In the present study, we tested whether 15-LOX-1 is down-regulated in human esophageal cancers using paired normal and tumor human surgical samples and whether NSAIDs can up-regulate 15-LOX-1 to restore apoptosis in esophageal cancer cells. We found that: (a) 15-LOX-1 was down-regulated in human esophageal carcinomas; (b) NSAIDs induced 15-LOX-1 expression during apoptosis in esophageal cancer cells; and (c) 15-LOX-1 inhibition suppressed NSAID-induced apoptosis, which was restored by 13-S-hydroxyoctadecadienoic acid but not by its parent compound, linoleic acid. These findings demonstrate that 15-LOX-1 is down-regulated in human esophageal carcinomas and that NSAIDs induce apoptosis in esophageal cancer cells via up-regulation of 15-LOX-1. They also support the concept that the loss of the proapoptotic role of 15-LOX-1 in epithelial cancers is not limited to human colorectal cancers.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Arachidonate 15-Lipoxygenase/biosynthesis , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Apoptosis/physiology , Arachidonate 15-Lipoxygenase/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Enzyme Induction/drug effects , Esophageal Neoplasms/drug therapy , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Linoleic Acids/pharmacology , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Sulindac/pharmacology , Up-Regulation/drug effects
12.
Cancer Epidemiol Biomarkers Prev ; 10(5): 447-53, 2001 May.
Article in English | MEDLINE | ID: mdl-11352853

ABSTRACT

UNLABELLED: Development of potential cancer chemopreventive drugs involves the systematic evaluation of these drugs in preliminary Phase I and II studies in human beings to identify the optimal drug dose, drug toxicity, and surrogate end point biomarker modulation. OBJECTIVES: We tested the hypothesis that aspirin, at a single, once-daily 81-mg dose, will reduce colonic mucosal concentration of prostaglandin estradiol (E2) in individuals at high risk for colorectal cancer development similar to our prior observations in a young normal-risk population. METHODS: Aspirin was administered at a dose of 81 mg once daily for 28 days in a cohort of 92 matched high-risk and normal-risk colorectal cancer subjects. Prostaglandin E2 and cyclooxygenase expression were assayed from distal sigmoid biopsies from all of the subjects before and after treatment. RESULTS: The mean prostaglandin E2 for normal-risk subjects before aspirin treatment was 11.3 +/- 1.7 pg/microg (mean +/- SE) tissue protein and after aspirin treatment was 4.9 +/- 0.91 pg/microg tissue protein (P < 0.0001). In high-risk subjects, mean pretreatment prostaglandin E2 was 14.4 +/- 1.7 pg/microg tissue protein and after aspirin treatment was 4.7 +/- 0.70 pg/microg tissue protein (P < 0.0001). Aspirin treatment did not alter cyclooxygenase-1 protein expression. CONCLUSIONS: Aspirin treatment at a dose of 81 mg reduces colorectal mucosal prostaglandin E2 concentration after 28 daily doses. Risk for colorectal carcinoma did not modify colorectal mucosal baseline or post-aspirin prostaglandin E2, or cyclooxygenase expression. Colorectal mucosal prostaglandin concentration may be used as a "drug-effect surrogate biomarker," that is, a surrogate to assess sufficient delivery and tissue effect of a chemopreventive agent.


Subject(s)
Aspirin/administration & dosage , Biomarkers, Tumor/analysis , Carcinoma/prevention & control , Colorectal Neoplasms/prevention & control , Dinoprostone/analysis , Intestinal Mucosa/chemistry , Intestinal Mucosa/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Adult , Aged , Analysis of Variance , Biopsy, Needle , Carcinoma/epidemiology , Carcinoma/pathology , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Proportional Hazards Models , Prostaglandin-Endoperoxide Synthases/drug effects , Reference Values , Risk Assessment , Sensitivity and Specificity
13.
J Natl Cancer Inst ; 92(14): 1136-42, 2000 Jul 19.
Article in English | MEDLINE | ID: mdl-10904086

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAIDs) appear to act via induction of apoptosis-programmed cell death-as potential colorectal cancer chemopreventive agents. NSAIDs can alter the production of different metabolites of polyunsaturated fatty acids (linoleic and arachidonic acids) through effects on lipoxygenases (LOXs) and cyclooxygenases. 15-LOX-1 is the main enzyme for metabolizing colonic linoleic acid to 13-S-hydroxyoctadecadienoic acid (13-S-HODE), which induces apoptosis. In human colorectal cancers, the expression of this enzyme is reduced. NSAIDs can increase 15-LOX enzymatic activity in normal leukocytes, but their effects on 15-LOX in neoplastic cells have been unknown. We tested the hypothesis that NSAIDs induce apoptosis in colorectal cancer cells by increasing the protein expression and enzymatic activity of 15-LOX-1. METHODS: We assessed 15-LOX-1 protein expression and enzymatic activity, 13-S-HODE levels, and 15-LOX-1 inhibition in association with cellular growth inhibition and apoptosis induced by NSAIDs (primarily sulindac and NS-398) in two colorectal cancer cell lines (RKO and HT-29). All P values are two-sided. RESULTS: Sulindac and NS-398 progressively increased 15-LOX-1 protein expression in RKO cells (at 24, 48, and 72 hours) in association with subsequent growth inhibition and apoptosis. Increased 13-S-HODE levels and the formation of 15-hydroxyeicosatetraenoic acid on incubation of the cells with the substrate arachidonic acid confirmed the enzymatic activity of 15-LOX-1. Inhibition of 15-LOX-1 in RKO cells by treatment with caffeic acid blocked NS-398-induced 13-S-HODE production, cellular growth inhibition, and apoptosis (P =. 007, P<.0001, and P<.0001, respectively); growth inhibition and apoptosis were restored by adding exogenous 13-S-HODE (P<.0001 for each) but not its parent compound, linoleic acid (P = 1.0 for each). Similar results occurred with other NSAIDs and in HT-29 cells. CONCLUSIONS: These data identify 15-LOX-1 as a novel molecular target of NSAIDs for inducing apoptosis in colorectal carcinogenesis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Arachidonate 15-Lipoxygenase/biosynthesis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Cyclooxygenase Inhibitors/pharmacology , Linoleic Acids/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/pharmacology , Arachidonate 15-Lipoxygenase/drug effects , Blotting, Western , Caffeic Acids/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Gas Chromatography-Mass Spectrometry , Gene Expression Regulation, Neoplastic , Humans , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Sulindac/pharmacology , Tumor Cells, Cultured , Up-Regulation
14.
Crit Rev Oncol Hematol ; 33(3): 157-67, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10789490

ABSTRACT

Chemoprevention is the use of natural or synthetic compounds to block, reverse, or prevent the development of invasive cancers. Cellular carcinogenesis forms the biologic basis for the identification of chemopreventives, assessment of their activity, and ultimately the success or failure of a chemopreventive. Chemopreventive agents undergo multistep evaluations to assess efficacy that are similar in concept but vastly different in practice to standard ablative oncologic therapeutics. In vitro assessments of potential anticarcinogenesis efficacy include measurements of an agent's antioxidant activity, induction of phase II metabolizing enzymes and effects upon cellular proliferation and apoptotic control pathways. In vivo efficacy is assessed primarily in rodent models of carcinogenesis that are specific for a given organ target. The role of genetically modified animal models in the in vivo assessment of chemoprevention agents remains unclear. Clinical assessment of chemopreventive agent efficacy consists of a multistep process of identification of an optimal chemopreventive agent (phase 1), demonstration of efficacy in humans through the modulation of reversal of a tissue, biochemical, and molecular surrogates for neoplastic transformation and invasion (phase 2) and cancer risk reduction in large cohort trials (phase 3). Opportunities and future needs include the development of reliable, predictive in vivo models of carcinogenesis, careful exploration of the preventive pharmacology of therapeutic agents being used for non-cancer prevention indications, and the incorporation of genetic risk cohorts to define cancer chemopreventive efficacy.


Subject(s)
Neoplasms/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/therapeutic use , Antioxidants/therapeutic use , Apoptosis/drug effects , Biomarkers , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/drug effects , Clinical Trials as Topic , Drug Screening Assays, Antitumor , Enzyme Inhibitors/therapeutic use , Epithelial Cells/drug effects , Epithelial Cells/pathology , Forecasting , Genetic Predisposition to Disease , Hormone Antagonists/therapeutic use , Humans , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Neoplasms, Experimental/prevention & control , Patient Compliance , Patient Selection , Rats , Rats, Inbred F344 , Risk Factors , Safety , Treatment Outcome
15.
Cancer Res ; 60(24): 6846-50, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11156377

ABSTRACT

We previously found (I. Shureiqi et al., Carcinogenesis (Lond.), 20: 1985-1995, 1999; I. Shureiqi et al, J. Natl. Cancer Inst., 92: 1136-1142, 2000) that (a) 15-lipoxygenase-1 (15-LOX-1) protein and its product 13-S-hydroxyoctadecadienoic acid (13-S-HODE) are decreased; and (b) nonsteroidal anti-inflammatory drug (NSAID)-induced 15-LOX-1 expression is critical to NSAID-induced apoptosis in colorectal cancer cells expressing cyclooxygenase-2 (COX-2). We used the NSAIDs sulindac sulfone (COX-2-independent) and NS-398 (a COX-2 inhibitor) to assess NSAID upregulation of 15-LOX-1 in relation to COX-2 inhibition during NSAID-induced apoptosis in the DLD-1 (COX-2-negative) colon cancer cell line. We found that: (a) NSAIDs up-regulated 15-LOX-1, which preceded apoptosis; and (b) 15-LOX-1 inhibition blocked NSAID-induced apoptosis, which was restored by 13-S-HODE but not by its parent, linoleic acid. NSAIDs can induce apoptosis in colon cancer cells via up-regulation of 15-LOX-1 in the absence of COX-2.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Arachidonate 15-Lipoxygenase/pharmacology , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Antithrombins/pharmacology , Arachidonic Acid/pharmacology , Blotting, Western , Caffeic Acids/pharmacology , Cell Line , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Linoleic Acid/pharmacology , Linoleic Acids/pharmacology , Membrane Proteins , Nitrobenzenes/pharmacology , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , Sulindac/analogs & derivatives , Sulindac/pharmacology , Time Factors , Tumor Cells, Cultured , Up-Regulation
16.
Curr Opin Oncol ; 11(5): 408-13, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10505781

ABSTRACT

Over the past 2 years, new insights into mutator and stromal epithelial cell interactions have enhanced the understanding of the carcinogenesis process and have identified potential new approaches to chemoprevention in diverse epithelial sites. Data testing the efficacy of chemopreventive agents in genetically mutated animal carcinogenesis models as screening tools for chemopreventive agents remain immature and point to a continued need for chemical carcinogenesis models to screen for the potential efficacy of chemopreventive agents. The Breast Cancer Prevention Trial is a published, risk-reduction trial that demonstrated a tamoxifen-induced reduction of the risk for breast cancer and focused attention on the clinical use of chemopreventive agents in healthy women. This trial highlighted the potential chemopreventive activity of selective estrogen-receptor antagonists as chemopreventives for breast cancer. New data from animal and human models continue to support the development of nonsteroidal anti-inflammatory agents as chemopreventives for colorectal cancer. Micronutrient- and diet-intervention trials for colorectal chemoprevention present a mixed picture. Although calcium and vitamin supplements, including folate, reduce the recurrence of adenomatous polyps in humans, the effect is small. Fiber supplementation does not reduce the incidence of polyps or colorectal cancer. New approaches for the chemoprevention of esophageal adenocarcinomas, hepatomas, and squamous cell skin cancers represent promising new approaches to the chemoprevention of epithelial cancers.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Neoplasms/prevention & control , Animals , Chemoprevention , Disease Models, Animal , Humans
17.
Carcinogenesis ; 20(10): 1985-95, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506115

ABSTRACT

13-S-Hydroxyoctadecadienoic acid (13-S-HODE), the product of 15-lipoxygenase (15-LOX) metabolism of linoleic acid, enhances cellular mitogenic responses to certain growth factors. Other observations have questioned whether 13-S-HODE has tumorigenic effects. Our study evaluated the hypothesis that 15-LOX-1 is overexpressed in colon cancers resulting in an increase in intracellular 13-S-HODE. 15-LOX-1 and 13-S-HODE were quantified using western blots, ELISA and immunohistochemistry in 18 human colon cancers with paired normal colonic mucosa. Additionally, 15-LOX-1 expression was measured by western blots in three transformed colonic cell lines and in a human umbilical vein endothelial cell line. Next, we evaluated 13-S-HODE effects on cellular proliferation, cell cycle distribution and apoptosis in a transformed colonic cell line (RKO). Cell cycle distributions were measured by flow cytometry and apoptosis was assessed by phase contrast microscopy, electron microscopy, flow cytometry and DNA fragmentation assay. 15-LOX-1 immunohistochemistry staining scores were reduced in tumor tissues (P

Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Colonic Neoplasms/metabolism , Linoleic Acids/metabolism , Apoptosis , Blotting, Western , Cell Cycle , Cell Division , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...