Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Epidemiol ; 32(1): 60-3, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12690010

ABSTRACT

BACKGROUND: A genetic component of early-onset lung cancer has been suggested. The role of metabolic gene polymorphisms has never been studied in young lung cancer cases. Phase 1 and Phase 2 gene polymorphisms are involved in tobacco carcinogens' metabolism and therefore in lung cancer risk. METHODS: The effect of metabolic gene polymorphisms on lung cancer at young ages was studied by pooling data from the Genetic Susceptibility to Environmental Carcinogens (GSEC) database. All primary lung cancer cases of both sexes who were Caucasian and

Subject(s)
Cytochrome P-450 CYP1A1/genetics , Genetic Predisposition to Disease , Lung Neoplasms/genetics , Polymorphism, Genetic , Adult , Age of Onset , Case-Control Studies , Chi-Square Distribution , Databases, Factual , Factor Analysis, Statistical , Female , Glutathione Transferase/genetics , Humans , Male , Risk Factors , Smoking/adverse effects
2.
Gastroenterology ; 121(6): 1407-16, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11729120

ABSTRACT

BACKGROUND & AIMS: Differences in genetic background may play a role in the development of ulcerative colitis (UC)-related neoplasia. Loss of heterozygosity (LOH) of APC has been reported in human UC-associated neoplasia. To investigate the role of genetic differences in UC-associated neoplasia, we compared differences in dextran sodium sulfate (DSS) colitis-associated neoplasia between wild-type C57BL/6J mice (WT-DSS) and C57BL/6J mice with a germline mutation in Apc (Min-DSS). METHODS: DSS colitis was induced in female wild-type and Min mice. Age- and sex-matched non-DSS-treated Mins were also studied. Animals were sacrificed after 1 and 2 cycles of DSS. The cecums and large intestines were studied for numbers of dysplasias/cancers. Dysplasias were studied for LOH of Apc. RESULTS: No WT-DSS, 100% of Min-DSS, and 50% of non-DSS-treated Mins had dysplasia. The mean numbers of lesions per mouse were 0 (WT-DSS), 15.6 and 29.3 (1 and 2 cycles Min-DSS, respectively), 1.2 and 1.9 (age-matched control Min, 1 and 2 cycle equivalents, respectively; P < 0.0002, Min-DSS vs. WT-DSS and non-DSS-treated Min; P = 0.03, Min-DSS 2 cycle vs. Min-DSS 1 cycle). Cancers were seen in 0%, 22%, and 40% of non-DSS Min, Min-DSS-1 cycle, and Min-DSS-2 cycle animals, respectively. LOH of Apc was observed in 90.6% of dysplasias and 6% of nondysplastic mucosa. CONCLUSIONS: A germline mutation in Apc contributes significantly to the development of colitis-associated neoplasia. Colitis markedly accelerates the development of dysplasia and cancer in the Min mouse. Dysplasia in Min-DSS occurs through LOH of Apc.


Subject(s)
Colitis/chemically induced , Colitis/genetics , Colonic Diseases/genetics , Colonic Neoplasms/genetics , Dextran Sulfate , Genes, APC , Germ-Line Mutation/physiology , Adenoma/epidemiology , Adenoma/genetics , Adenoma/pathology , Animals , Colitis/pathology , Colonic Diseases/epidemiology , Colonic Diseases/pathology , Colonic Neoplasms/epidemiology , Colonic Neoplasms/pathology , Female , Incidence , Loss of Heterozygosity , Mice , Mice, Inbred C57BL/genetics , Ulcer/genetics , Ulcer/pathology
3.
Cancer Epidemiol Biomarkers Prev ; 10(12): 1239-48, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751440

ABSTRACT

Using the International Project on Genetic Susceptibility to Environmental Carcinogens (GSEC) database containing information on over 15,000 control (noncancer) subjects, the allele and genotype frequencies for many of the more commonly studied metabolic genes (CYP1A1, CYP2E1, CYP2D6, GSTM1, GSTT1, NAT2, GSTP, and EPHX) in the human population were determined. Major and significant differences in these frequencies were observed between Caucasians (n = 12,525), Asians (n = 2,136), and Africans and African Americans (n = 996), and some, but much less, heterogeneity was observed within Caucasian populations from different countries. No differences in allele frequencies were seen by age, sex, or type of controls (hospital patients versus population controls). No examples of linkage disequilibrium between the different loci were detected based on comparison of observed and expected frequencies for combinations of specific alleles.


Subject(s)
Black People/genetics , Gene Frequency , Genetic Predisposition to Disease , Neoplasms/genetics , Polymorphism, Genetic , White People/genetics , Cytochrome P-450 Enzyme System/genetics , Databases, Factual , Genetic Linkage , Humans
4.
Cancer Detect Prev ; 25(4): 352-61, 2001.
Article in English | MEDLINE | ID: mdl-11531012

ABSTRACT

The dithiolethione oltipraz is being developed as a chemopreventive agent for many malignancies, including colorectal cancer, on the basis of its in vivo protective activity against chemically induced tumors in a variety of animal models. This protection has been associated with an enhanced capacity to detoxify reactive carcinogens and, more recently, with increased DNA repair. In a previous single-dose study, elevated detoxification gene expression was observed in the days after oltipraz dosing. Now, in this clinical study, we evaluated the effects of oltipraz when given over a 3-month period. Fourteen individuals with increased risk for colorectal cancer were randomly assigned to one of two oral doses (125 or 250 mg/m2) of oltipraz twice weekly for 12 weeks. Two of seven subjects at the 250 mg/m2 dosage required dose reductions, owing to significant fatigue. The 125 mg/m2 dose level was well tolerated by all patients. Blood or colon tissue (or both) for evaluation of glutathione, glutathione S-transferase, DT-diaphorase activity, and DT-diaphorase mRNA expression were obtained prior to treatment and at weeks 6, 12, and 16. No significant modulation of phase II detoxification enzymes was seen at either dose studied during this period. Phase II trials evaluating a tolerable regimen of oltipraz (as demonstrated in this study) and other possible mechanisms that may be responsible for the protective activity of oltipraz should be pursued.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Pyrazines/therapeutic use , Aged , Aged, 80 and over , Anticarcinogenic Agents/administration & dosage , Biomarkers, Tumor/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Enzyme Induction/drug effects , Female , Glutathione/blood , Glutathione Transferase/biosynthesis , Humans , Male , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Pyrazines/administration & dosage , Thiones , Thiophenes
5.
Curr Opin Oncol ; 13(4): 307-13, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429490

ABSTRACT

Chemopreventive strategies hold substantial promise for reducing the incidence of colorectal cancer, the second leading cause of cancer-related mortality in the United States. This review focuses on recent advances in the identification of molecular targets and novel strategies for chemopreventive intervention. Many clinical trials are now in progress to assess the ability of synthetic agents or nutritional supplements to alter either the number of colorectal adenomas or biomarkers associated with colorectal tumorigenesis. Populations under study include genetically defined high-risk people and those with increased risk based on a personal history of colorectal neoplasia. A recent study showing that celecoxib, a cyclooxygenase-2 inhibitor, can alter the natural history of polyp formation in patients with familial adenomatous polyposis has provided a benchmark for the clinical development of other chemopreventive agents and heightened awareness that colorectal cancer is a preventable disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colorectal Neoplasms/prevention & control , Chemoprevention , Colorectal Neoplasms/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Diet Therapy , Humans , Vitamins/therapeutic use
6.
Curr Oncol Rep ; 2(3): 251-6, 2000 May.
Article in English | MEDLINE | ID: mdl-11122850

ABSTRACT

Inter-individual variability in carcinogen metabolism has been attributed in part to the polymorphic expression of several phase I and II detoxification enzymes. The role of these genetic polymorphisms in cancer susceptibility has been most extensively evaluated for isozymes of cytochrome P450 (CYP1A1, CYP2D6, and CYP2E1), N-acetyltransferase (NAT1 and NAT2), glutathione S-transferase (GSTM1, GSTT1, and GSTP1), microsomal epoxide hydrolase, and NAD(P)H:quinone oxidoreductase. Our understanding of the genetic basis of cancer risk has been enhanced most recently by establishment of genotype-phenotype correlations in humans and identification of numerous diverse factors, both genetic and environmental, that can modify risk.


Subject(s)
Genetic Testing , Neoplasms/genetics , Polymorphism, Genetic , Female , Genetic Predisposition to Disease/epidemiology , Humans , Male , Neoplasms/epidemiology , Risk Assessment , Risk Factors , Sensitivity and Specificity
7.
Lung Cancer ; 30(3): 153-60, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11137199

ABSTRACT

In contrast to men, the incidence of lung cancer among women has increased over the past decade. The basis for this increase among female smokers remains unknown. Surgical patients with a diagnosis of lung cancer and control subjects without a history of malignancy completed a smoking questionnaire and donated a blood sample. DNA was extracted from peripheral mononuclear cells and genotyped for polymorphisms in cytochrome P450 1A1 (CYP1A1) (exon 7) and glutathione S-transferase M1 (GSTM1) (null). No gender differences in either age at diagnosis or histological subtype were observed among lung cancer patients. In both patients (n = 180) and controls (n = 163), females smoked significantly less than males. The pack-year history associated with adenocarcinoma was smaller than that for squamous cell carcinoma. No significant association was observed between the GSTM1 null genotype and cancer risk. However, women had a larger cancer risk than men (odds ratio 4.98 vs. 1.37) if they possessed the mutant CYP1A1 genotype. Female cancer patients were significantly more likely than female controls to have both the CYP1A1 mutation and GSTM1 null genotype. The combined variant genotypes conferred an odds ratio of 6.54 for lung cancer in women versus 2.36 for men, independent of age or smoking history. These data suggest that polymorphisms in CYP1A1 and GSTM1 contribute to the increased risk of females for lung cancer.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Lung Neoplasms/genetics , Polymorphism, Genetic , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Genotype , Humans , Male , Middle Aged , Risk Factors , Sex Factors , Smoking/adverse effects
8.
Anal Quant Cytol Histol ; 21(2): 166-73, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10560487

ABSTRACT

OBJECTIVE: The expression of p53 and cyclin D1 proteins was analyzed by image analysis in oral premalignant lesions and normal oral mucosa. STUDY DESIGN: Punch biopsies from the normal oral mucosa were obtained from 20 normal donors and 41 patients with oral dysplastic leukoplakias. After controlled formaldehyde fixation and paraffin embedding, immunohistochemistry was used to detect cyclin D1 and p53. Image analysis was performed using stain intensity levels established by determining color thresholds (nuclear score) in the basal and parabasal layers. RESULTS: Analysis of sections showed a similar pattern: only two normal donors had a few intensely positive p53 cells in the basal layer of the floor of the mouth and the tongue epithelia. Similarly, only three donors had intensely positive cyclin D1 cells in the normal epithelia of the same sites. Most cells fell in the range of negative or marginal stain (lower quartiles or terciles of nuclear score). These data on normal mucosa were compared with low grade oral leukoplakias (LGD) with mild to moderate dysplasia and with high grade leukoplakias (HGD) with severe dysplasia. Both markers were differentially expressed in precursor lesions versus normal epithelia. Statistical analysis of our data shows that the intensity of the immunohistochemical stain, as reflected in the nuclear scores of p53, is a reliable parameter that can differentiate between LGD and HGD of the oral mucosa. This was especially true when higher nuclear scores were compared. In contrast, low nuclear scores are more effective in differentiating normal epithelia from dysplastic epithelia. Although cyclin D1 immunohistochemistry does not stain as intensely as p53 stain, similar conclusions can be derived from those data. CONCLUSION: Image analysis of these two markers proved useful in distinguishing normal oral epithelia from low grade and high grade leukoplakias. With further developments in this field it is hoped that image analysis procedures could be used in different types of studies in which variations of protein expression in tissue sections could have prognostic implications or could be useful to determine subtle effects of curative or preventive treatment.


Subject(s)
Cyclin D1/metabolism , Leukoplakia, Oral/metabolism , Mouth Mucosa/metabolism , Precancerous Conditions/metabolism , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Biopsy , Cell Nucleus/pathology , Humans , Image Cytometry , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Leukoplakia, Oral/pathology , Middle Aged , Mouth Mucosa/pathology , Precancerous Conditions/pathology
9.
Aliment Pharmacol Ther ; 13(3): 389-96, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10102973

ABSTRACT

BACKGROUND: The increased risk of colonic malignancies in individuals with ulcerative colitis has prompted a search for early biomarkers of disease progression. AIM: To characterize Phase II detoxication enzyme expression during acute and chronic colitis. The mouse model of dextran sulphate sodium (DSS)-induced colitis represents a relevant system with which to sequentially evaluate the spectrum of biochemical changes associated with colorectal cancer risk. METHODS: Acute and chronic colitis were induced in Swiss Webster mice by administering DSS in the drinking water (5%) for 1-4 cycles. Each cycle consisted of 7 days DSS and 14 days of water. The glutathione S-transferase (GST) activity, gamma-glutamylcysteine synthetase (gamma-GCS) activity and glutathione content of the colonic tissues were determined at various time points throughout the experiment. Alterations in GST isozyme expression were confirmed by Western and Northern blot. RESULTS: GST activity was reduced significantly in the colon by the end of Cycle 1 (84% of control values). Specific activities continued to decrease with subsequent cycles of DSS exposure. By the end of Cycle 4, glutathione levels and gamma-GCS activity had reached 29% and 56% of control, respectively. CONCLUSIONS: These data suggest that detoxication enzyme depletion is associated with both acute and chronic colitis and may be an important event in the progression of ulcerative colitis to colon cancer.


Subject(s)
Colitis/enzymology , Colon/enzymology , Dextran Sulfate , Animals , Biomarkers , Blotting, Northern , Blotting, Western , Colitis/chemically induced , Colonic Neoplasms/enzymology , Female , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Immunohistochemistry , Isoenzymes/metabolism , Mice
11.
Cancer Epidemiol Biomarkers Prev ; 7(7): 597-603, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9681528

ABSTRACT

The expression of several markers of epithelial cell proliferation was analyzed to establish baseline data for future chemoprevention studies of oral premalignant lesions. Punch biopsies (n = 60) from three different sites of oral mucosa (bucca, lateral tongue, and the floor of the mouth) were obtained from 20 normal donors of both sexes. After formaldehyde fixation and paraffin embedding, immunohistochemistry was used to detect the proliferation markers Mib-1, cyclin D1, and centromere-associated protein CENP-F. Analysis of sections stained for the three markers showed similar patterns, i.e., a low labeling index (LI) in the basal layer and a high LI in the parabasal layer at all three intraoral sites. No proliferative activity was seen above the parabasal layer (superficial layer). All sites showed similar Mib-1 LI values for the proliferative markers. The tongue epithelium exhibited higher parabasal LIs of cyclin D1 and CENP-F than did the other two sites. No significant differences were detected between smokers and nonsmokers. The data from normal mucosa were compared with those from low (n = 30)- and high (n = 17)-grade dysplastic leukoplakias. The Mib-1 LI showed a very significant change, with a 9-fold increase in the basal layer LI in dysplastic leukoplakias. Cyclin D1 and CENP-F showed similar trends with increments of up to 7-fold in the basal layer of high-grade dysplasia. Although the proliferative activity of the parabasal layer was similar in normal and leukoplakic epithelia, the superficial layer showed a significant increment in proliferative activity mainly in high-grade leukoplakia. These studies suggest that proliferation markers in the basal and superficial cells of premalignant lesions may serve as surrogate end point biomarkers for chemoprevention trials.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma in Situ/chemistry , Chromosomal Proteins, Non-Histone/analysis , Cyclin D1/analysis , Epithelial Cells/chemistry , Leukoplakia, Oral/chemistry , Mouth Mucosa/chemistry , Nuclear Proteins/analysis , Antigens, Nuclear , Carcinoma in Situ/pathology , Cell Division , Epithelial Cells/cytology , Epithelial Cells/pathology , Female , Humans , Ki-67 Antigen , Leukoplakia, Oral/pathology , Male , Microfilament Proteins , Mouth Mucosa/cytology , Mouth Mucosa/pathology
12.
Chem Biol Interact ; 111-112: 377-88, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9679568

ABSTRACT

The critical role of the glutathione S-transferase (GST) multigene family in cellular protection in combination with the large interindividual variability in the expression of these enzymes has prompted an investigation of their importance in cancer prevention and susceptibility. Previous preclinical and clinical studies from this laboratory have established an association between decreased GST activity and increased risk for colorectal cancer. Based upon the increased incidence of colon malignancies among patients with ulcerative colitis, GST activity has been examined in a mouse model of induced colitis. Significant decreases (50% of controls) in the GST activity of colon tissue were observed during the establishment and progression of colitis. These data suggested that depletion of cellular protection may be an important event in the carcinogenic progression of ulcerative colitis. The ability of the dithiolthione oltipraz to induce GST expression within the murine colon has been demonstrated. Use of chemopreventive regimens to induce phase 2 detoxication enzyme expression represents a promising strategy for the prevention of cancer. Clinical studies revealed that the GST activity of blood lymphocytes from individuals with either a personal or family history of colorectal cancer or a personal history of colon polyps was decreased significantly when compared to that of healthy controls. Phase 1 clinical evaluation of oltipraz has demonstrated its ability to induce GST activity as well as the level of transcripts encoding gamma-glutamylcysteine synthetase (gamma-GCS) and DT-diaphorase in the colon mucosa of individuals at increased risk for colorectal cancer. The observed correlation between the posttreatment response in blood lymphocytes and colon mucosa suggested that blood lymphocytes may be used in future trials as a surrogate biomarker of the responsiveness of colon tissue to chemopreventive regimens.


Subject(s)
Glutathione Transferase/metabolism , Neoplasms/etiology , Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/pharmacology , Biomarkers , Brassica , Colitis, Ulcerative/complications , Colitis, Ulcerative/enzymology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/etiology , Disease Models, Animal , Enzyme Induction , Gene Expression , Glutathione Transferase/deficiency , Glutathione Transferase/genetics , Humans , Inactivation, Metabolic , Mice , Neoplasms/enzymology , Pyrazines/pharmacology , Risk Factors , Thiones , Thiophenes
13.
Pharmacol Ther ; 78(1): 17-27, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9593327

ABSTRACT

The antischistosomal agent oltipraz [5-(2-pyrazinyl)-4-methyl-1,2-dithiol-3-thione] has been shown to inhibit chemically induced carcinogenesis in a variety of animal models. Of greatest interest is its unique ability to protect several target organs from structurally diverse carcinogens. Molecular and biochemical studies suggest that oltipraz affords cellular protection by inducing the expression of a battery of Phase II detoxification enzymes. Induction of glutathione S-transferase, gamma-glutamylcysteine synthetase and DT-diaphorase has been observed in human tissues following the administration of a single oral dosage of oltipraz. Preclinical and clinical data continue to support the development of oltipraz as a chemopreventive agent for clinical usage.


Subject(s)
Anticarcinogenic Agents/pharmacology , Pyrazines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Schistosomicides/pharmacology , Administration, Oral , Animals , Anticarcinogenic Agents/therapeutic use , Biological Availability , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Enzyme Induction/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Glutathione Transferase/biosynthesis , Humans , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Pyrazines/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Schistosomicides/therapeutic use , Thiones , Thiophenes , Tissue Distribution
14.
Biochem Biophys Res Commun ; 253(3): 855-8, 1998 Dec 30.
Article in English | MEDLINE | ID: mdl-9918819

ABSTRACT

Regulation of the basal and induced expression of detoxifying enzymes such as NAD(P)H:quinone oxidoreductasel (NQO1) and glutathione S-transferase (GST) by the antioxidant response element (ARE) is important for cellular protection against oxidative stress. The ARE contains AP1 and AP1-like elements and is known to bind to several leucine zipper proteins including c-Fos. Previous studies (Venugopal, R., and Jaiswal, A.K. (1996) Proc. NatL Acad. Sci. USA 93, 14960-14965) have shown that overexpression of c-Fos in transfected cells leads to repression of ARE-mediated gene expression. In the present report, we used c-Fos-/- mice and investigated the physiological (in vivo) role of c-Fos in repression of the NQO1 and GST genes expression. The analysis of enzyme activity levels showed significant increases in NQO1 and GST activities in several tissues of c-Fos-/- mice, as compared with wild type (c-Fos+/+) mice. The increases in enzyme activities were supported by Wetern analysis of respective proteins. Western analyses showed significant increases in the expression of NQO1 in kidney, liver and skin tissues of c-Fos-/- mice, as compared with wild type (c-Fos+/+) controls. Western analyses also demonstrated an increased expression of the GST Ya gene in kidney and liver tissues of the c-Fos-/-mice. These results confirm a negative (repressive) role for c-Fos in the expression of NQO1, GST Ya, and other detoxifying enzyme genes.


Subject(s)
Glutathione Transferase/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Proto-Oncogene Proteins c-fos/deficiency , Animals , Antioxidants/metabolism , Dicumarol/pharmacology , Gene Expression Regulation, Enzymologic , Glutathione Transferase/genetics , Inactivation, Metabolic , Mice , Mice, Mutant Strains , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/genetics , Proto-Oncogene Proteins c-fos/genetics , Response Elements , Tissue Distribution
15.
Cancer Res ; 57(13): 2749-53, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9205086

ABSTRACT

Hepatitis B virus (HBV) and aflatoxin B1 represent the main risk factors for the development of hepatocellular carcinoma (HCC) in areas endemic for liver cancer. The glutathione S-transferases (GSTs) are a family of Phase II detoxification enzymes that catalyze the conjugation of a wide variety of endogenous and exogenous toxins, including aflatoxin B1, with glutathione. This study characterizes the GST isoenzyme composition (alpha, mu, and pi) of both HBV-infected normal hepatic tissues and HCCs. Analysis of matched pairs of hepatic tissue (normal and tumor) from 32 HCC patients indicated that total GST activity was significantly higher in normal tissues than in tumor tissues, although the percentage of samples expressing GST alpha and pi was equivalent. GST mu was detected by Western blot in the normal tissue from 87.5% of the subjects possessing the GST M1 gene but only 28.6% of the corresponding tumor tissues. The GST activity of normal tissue from GST M1 null patients was significantly decreased as compared to that of subjects possessing the GST M1 gene (264.6 and 422.2 nmol/min/mg, respectively; P = 0.005). GST pi appeared to be overexpressed in the normal tissue of GST M1 null patients, a potential compensatory effect. Patients positive for HBV DNA had significantly lower GST activity than those who were HBV negative (302.1 versus 450.0 nmol/min/mg, respectively; P = 0.02). These results suggest that cellular protection within the human liver is compromised by HBV infection and further decreased during hepatocellular tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Glutathione Transferase/metabolism , Hepatitis B/metabolism , Liver Neoplasms/enzymology , Adult , Age Factors , Aged , Carcinoma, Hepatocellular/genetics , Female , Glutathione Transferase/genetics , Humans , Liver Neoplasms/genetics , Male , Middle Aged
16.
Clin Cancer Res ; 3(1): 25-30, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9815533

ABSTRACT

Previous studies suggest that cruciferous vegetables may provide protection against carcinogen exposure by inducing detoxification enzymes. ICR(Ha) mice were gavaged with broccoli tablets (1 g/kg), and colon tissues were collected after treatment. Glutathione S-transferase (GST) activity was assayed and peaked on days 1 and 2 after treatment, respectively (P = 0.03). Elevations in GST activity were attributed to the increased expression of mu and pi. These data supported a clinical assessment of broccoli supplements. Twenty-nine subjects at increased risk for colorectal cancer were randomized to group 1 (no cruciferous vegetables) or group 2 (broccoli supplements, 3 g/day) for 14 days. Blood samples and colon biopsies were obtained pre- and postintervention. No significant difference was observed between the GST activities of the control and broccoli supplementation groups posttreatment. Mean lymphocyte GST activity was 107% of baseline in the broccoli supplementation group (range, 79-158%) and 102% of baseline in the control group (range, 75-158 percent;). Correlation of the GST activities of blood lymphocytes and colon mucosa taken simultaneously suggested that the GST activity of blood lymphocytes may be used as a biomarker of the responsiveness of colon tissue to chemopreventive regimens. Future clinical studies evaluating cruciferous vegetables should consider using concentrated dietary supplements in subjects with a previous history of colorectal cancer.


Subject(s)
Brassica , Dietary Supplements , Glutathione Transferase/biosynthesis , Adult , Aged , Animals , Chemoprevention , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/prevention & control , Enzyme Induction , Female , Gastric Mucosa/enzymology , Humans , Lymphocytes/enzymology , Male , Mice , Mice, Inbred ICR , Middle Aged , Risk Factors
17.
Proc Soc Exp Biol Med ; 216(2): 192-200, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9349688

ABSTRACT

Detoxication enzymes protect cells from a wide variety of xenobiotics and endogenous toxins. Current data suggest that the balance between the Phase I carcinogen-activating enzymes and the Phase II detoxifying enzymes is critical to determining an individual's risk for cancer. Human deficiencies in Phase II enzyme activity, specifically glutathione-S-transferase (GST), have been identified and associated with increased risk for colon cancer. The increased frequency of the GST M1 null genotype among individuals with primarily smoking-related cancers has been documented. Induction of Phase II enzymes by naturally occurring or synthetic agents represents a promising strategy for cancer prevention. Both the required characteristics of potential chemopreventive agents and the role of the antioxidant response element in the monofunctional induction of Phase II enzymes have been discussed. The synthetic dithiolthione oltipraz induces a battery of Phase II enzymes and inhibits chemically induced tumors in a variety of target organs. Its ability to induce Phase II enzymes in human colon tissue and blood lymphocytes has been reported. Other promising inducers with chemopreventive activity include the isothiocyanates and polyphenols. These data collectively support the future development of Phase II enzyme inducers as clinical chemopreventive agents.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Chemoprevention , Inactivation, Metabolic , Neoplasms/prevention & control , Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Carcinogens/metabolism , Enzyme Induction , Enzymes/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Risk Factors
18.
J Clin Invest ; 98(5): 1210-7, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8787684

ABSTRACT

Prolonged exposure to mutagenic substances is strongly associated with an individual's risk of developing colorectal cancer. Clinical investigation of oltipraz as a chemopreventive agent is supported by its induction of the expression of detoxication enzymes in various tissues, and its protective activity against the formation of chemically induced colorectal tumors in animals. The goals of the present study were: to determine if oltipraz could induce detoxicating gene expression in human tissues; to identify effective non-toxic doses for more extensive clinical testing; and to establish a relationship between effects in the colon mucosa and those in a more readily available tissue, the peripheral mononuclear cell. 24 evaluable patients at high risk for colorectal cancer were treated in a dose-finding study with oltipraz 125, 250, 500, or 1,000 mg/m2 as a single oral dose. Biochemical analysis of sequential blood samples and colon mucosal biopsies revealed increases in glutathione transferase activity at the lower dose levels. These effects were not observed at the higher doses. More pronounced changes were observed in detoxicating enzyme gene expression in both tissues at all doses. Peripheral mononuclear cell and colon mRNA content for gamma-glutamylcysteine synthetase (gamma-GCS) and DT-diaphorase increased after dosing to reach a peak on day 2-4 after treatment, and declined to baseline in the subsequent 7-10 d. The extent of induction of gene expression in colon mucosa reached a peak of 5.75-fold for gamma-GCS, and a peak of 4.14-fold for DT-diaphorase at 250 mg/m2 ; higher doses were not more effective. Levels of gamma-GCS and DT-diaphorase correlated closely (P < or = 0.001) between peripheral mononuclear cells and colon mucosa both at baseline and at peak. These findings demonstrate that the administration of minimally toxic agents at low doses may modulate the expression of detoxicating genes in the tissues of individuals at high risk for cancer. Furthermore, peripheral mononuclear cells may be used as a noninvasive surrogate endpoint biomarker for the transcriptional response of normal colon mucosa to drug administration.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Gene Expression Regulation, Neoplastic , Pyrazines/therapeutic use , Adult , Aged , Aged, 80 and over , Chemoprevention , Colon/drug effects , Colon/enzymology , Female , Glutamate-Cysteine Ligase/analysis , Humans , Inactivation, Metabolic , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Male , Middle Aged , Mutagenesis/drug effects , NAD(P)H Dehydrogenase (Quinone)/analysis , Risk , Thiones , Thiophenes
19.
Carcinogenesis ; 16(9): 2159-65, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7554069

ABSTRACT

The synthetic dithiolethione Oltipraz has marked cancer chemopreventive and phase II enzyme inducing activity in various animal carcinogenesis models, but has not been examined in any animal models of ductal pancreatic cancer relevant to the human disease. The chemopreventive potential of Oltipraz on pancreatic tumor incidence and multiplicity was examined in the N-nitrosobis(2-oxopropyl)-amine (BOP)-induced ductal pancreatic adenocarcinoma model in Syrian hamsters. Animals were maintained on control semipurified diets or semipurified diets containing 300 and 600 mg/kg Oltipraz beginning 2 weeks prior to BOP initiation and throughout the 26 week study. Oltipraz at 300 mg/kg had no effect on the incidence or multiplicity of preneoplastic, neoplastic or metastatic lesions, while at 600 mg/kg dietary Oltipraz the incidence of pancreatic adenocarcinomas was reduced significantly (P < or = 0.05) compared to BOP-treated controls. Dietary Oltipraz at both doses had a significant influence on reducing mortality and morbidity in tumor-bearing animals with metastatic disease. At 26 weeks, total hepatic glutathione-S transferase (GST) activity and GST mu activity were elevated significantly in Oltipraz-treated animals, while total pancreatic GST activity was reduced, albeit not significantly. Serum lipase activity, a marker for pancreatic damage, exhibited a progressive decline in BOP-treated animals administered Oltipraz compared to BOP-treated controls at 12 weeks of the study; by week 26, lipase activity was comparable in all groups and reduced compared to activity at week 12. Positive nuclear immunostaining for the p53 tumor suppressor protein, a hallmark of human pancreatic cancer and a transient response to DNA damage, was observed in only a small percentage of BOP-induced pancreatic lesions and was not influenced Oltipraz administration. Further chemoprevention and pharmacologic studies of Oltipraz in relevant animal models of ductal pancreatic cancer could provide a foundation for future studies in human populations at potential risk for pancreatic cancer.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Ductal, Breast/prevention & control , Pancreatic Neoplasms/prevention & control , Pyrazines/therapeutic use , Animals , Body Weight/drug effects , Carcinogens , Carcinoma, Ductal, Breast/chemically induced , Cricetinae , Female , Glutathione Transferase/metabolism , Immunohistochemistry , Liver/enzymology , Mesocricetus , Nitrosamines , Pancreas/enzymology , Pancreatic Neoplasms/chemically induced , Thiones , Thiophenes , Tumor Suppressor Protein p53/analysis
20.
Proc Natl Acad Sci U S A ; 92(6): 2384-7, 1995 Mar 14.
Article in English | MEDLINE | ID: mdl-7892276

ABSTRACT

Aflatoxin B1 (AFB1) has been postulated to be a hepatocarcinogen in humans, possibly by causing p53 mutations at codon 249. AFB1 is metabolized via the phase I and II detoxification pathways; hence, genetic variation at those loci may predict susceptibility to the effects of AFB1. To test this hypothesis, genetic variation in two AFB1 detoxification genes, epoxide hydrolase (EPHX) and glutathione S-transferase M1 (GSTM1), was contrasted with the presence of serum AFB1-albumin adducts, the presence of hepatocellular carcinoma (HCC), and with p53 codon 249 mutations. Mutant alleles at both loci were significantly overrepresented in individuals with serum AFB1-albumin adducts in a cross-sectional study. Mutant alleles of EPHX were significantly overrepresented in persons with HCC, also in a case-control study. The relationship of EPHX to HCC varied by hepatitis B surface antigen status and indicated that a synergistic effect may exist. p53 codon 249 mutations were observed only among HCC patients with one or both high-risk genotypes. These results indicate that individuals with mutant genotypes at EPHX and GSTM1 may be at greater risk of developing AFB1 adducts, p53 mutations, and HCC when exposed to AFB1. Hepatitis B carriers with the high-risk genotypes may be an even greater risk than carriers with low-risk genotypes. These findings support the existence of genetic susceptibility in humans to the environmental carcinogen AFB1 and indicate that there is a synergistic increase in risk of HCC with the combination of hepatitis B virus infection and susceptible genotype.


Subject(s)
Aflatoxin B1/metabolism , Carcinoma, Hepatocellular/genetics , Epoxide Hydrolases/genetics , Genes, p53 , Genetic Variation , Glutathione Transferase/genetics , Liver Neoplasms/genetics , Aflatoxin B1/analysis , Aflatoxin B1/blood , Carcinoma, Hepatocellular/epidemiology , Case-Control Studies , China , Codon , DNA/analysis , Genetic Predisposition to Disease , Genotype , Hepatitis B Surface Antigens/blood , Humans , Inactivation, Metabolic/genetics , Liver Neoplasms/epidemiology , Point Mutation , Polymerase Chain Reaction , Reference Values , Risk Factors , Serum Albumin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL