Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Oncogene ; 25(8): 1277-80, 2006 Feb 23.
Article in English | MEDLINE | ID: mdl-16247444

ABSTRACT

Although ras and p53 are the most commonly found oncogene and tumor suppressor gene, respectively, in human cancers, their collective roles in tumor progression have yet to be defined in animal models. Here, we demonstrated the synergistic effect between ras and p53 in promoting tumor progression during lung tumorigenesis using bitransgenic mice. Mice with a heterozygous knockout of K-ras (K-ras(wt/ko)) were mated to p53 transgenic mice (p53(val135/wt)) in lung tumorigenesis (K-ras(wt/ko) x p53(val135/wt)). F(1) mice exhibited a significant increase in lung tumor load (tumor multiplicity x tumor volume) when compared to those seen in either K-ras(wt/ko) mice or p53(val135/wt) mice alone. Furthermore, over 50% of the lung tumors were lung adenocarcinomas in bitransgenic mice compared to only 3% in wild-type mice. Alterations of ras and p53 appear to promote the development of lung adenocarcinomas. These results provide the in vivo experimental evidence of synergistic interactions of ras and p53 in lung tumor progression.


Subject(s)
Adenocarcinoma/genetics , Disease Models, Animal , Genes, ras/physiology , Lung Neoplasms/genetics , Tumor Suppressor Protein p53/physiology , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Cocarcinogenesis , Disease Progression , Genes, ras/genetics , Heterozygote , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mice, Transgenic , Tumor Suppressor Protein p53/genetics , Urethane/analogs & derivatives , Urethane/toxicity
2.
Oncogene ; 25(9): 1320-9, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16247446

ABSTRACT

Bexarotene (Targretin), is a synthetic high-affinity RXR receptor agonist with limited affinity for RAR receptors. Bexarotene has shown efficacy in a phase I/II trial of non-small-cell lung cancers. However, the chemopreventive efficacy of bexarotene has not been determined in mouse lung cancer models. In this study, we have investigated the ability of bexarotene to inhibit lung tumor progression in the mutant A/J mouse models with genetic alterations in p53 or K-ras, two of the most commonly altered genes in human lung tumorigenesis. Mice were administered vinyl carbamate (VC), a carcinogen, by a single intraperitoneal injection (i.p.) at 6 weeks of age. Bexarotene was given by gavage starting at 16 weeks after VC and was continued for 12 weeks. Although all mice developed lung tumors, only 7% of lung tumors were adenocarcinomas in wild-type mice, whereas 22 and 26% of lung tumors were adenocarcinomas in p53 transgenic or K-ras heterozygous deficient mice. Bexarotene inhibited both tumor multiplicity and tumor volume in mice of all three genotypes. Furthermore, bexarotene reduced the progression of adenoma to adenocarcinoma by approximately 50% in both p53(wt/wt)K-ras(ko/wt) and p53(wt/wt)K-ras(wt/wt) mice. Thus, bexarotene appears to be an effective preventive agent against lung tumor growth and progression.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/prevention & control , Lung Neoplasms/prevention & control , Tetrahydronaphthalenes/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Animals , Bexarotene , Carcinogens/administration & dosage , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Disease Progression , Genes, p53 , Genes, ras , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Urethane/administration & dosage , Urethane/analogs & derivatives
3.
J Natl Cancer Inst ; 61(3): 897-903, 1978 Sep.
Article in English | MEDLINE | ID: mdl-278867

ABSTRACT

The murine plasmacytoma MOPC 104E was susceptible to cytotoxic therapy in female inbred BALB/c mice. Palpable subcutaneous tumors (0.6--1.2 X 10(8) cells) could be cured with a single administration of cyclophosphamide (5--250 mg/kg) or localized irradiation (800--2,400 R). Clonogenic assay showed that, following minimal curative doses of cyclophosphamide or radiation, 0.5--1.5 X 10(6) tumor cells should remain viable. Control animals succumbed to progressive, invariably lethal tumor growth after they were given sc injections of 2--3 X 10(3) tumor cells. Minimal doses of cyclophosphamide, which were curative in control tumor-bearing animals, were ineffective in treating tumor-bearing animals immunosuppressed by 450 R whole-body irradiation. Subsequent experiments measured the ability of animals cured of the murine plasmacytoma to reject secondary challenge with the same tumor. These experiments demonstrated and partially quantitated the substantial role of the immune response in effecting tumor cure following radiotherapy or chemotherapy.


Subject(s)
Cyclophosphamide/therapeutic use , Immunity , Plasmacytoma/therapy , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , Female , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/therapy , Plasmacytoma/immunology , Plasmacytoma/pathology , Radiotherapy Dosage , Remission, Spontaneous
4.
J Natl Cancer Inst ; 60(5): 1107-11, 1978 May.
Article in English | MEDLINE | ID: mdl-642029

ABSTRACT

BALB/c mice with the plasmacytoma MOPC 104E were cured of palpable tumors (6-15x10(7) cells) with a single injection of cyclophosphamide (10 mg/kg). Animals cured of tumor showed a considerable increase in their ability to reject secondary challenge with graded numbers of viable tumor cells. Mice with palpable subcutaneous tumors were cured therapeutically and rechallenged 22, 44, or 120 days post therapy. The ability of such animals to reject secondary tumor cell challenge was similar in all groups, which implied that in vivo tumor immunity remained relatively constant for at least 4 months post therapy. A second group of animals was treated therapeutically (10 mg cyclophosphamide/kg) 4, 11, or 20 days post tumor cell injection. These therapeutically treated animals were then rechallenged with various numbers of viable tumor cells 30 days post therapy. Mice given cyclophosphamide 4, 11, or 20 days post tumor injection rejected 6, 60, or 400 times as many tumor cells, respectively, as did control animals. These results implied that, over the range of tumor sizes investigated, exposure to greater amounts of tumor antigen resulted in increasing amounts of residual tumor immunity following cure.


Subject(s)
Immunity , Plasmacytoma/immunology , Animals , Antigens, Neoplasm/administration & dosage , Cell Count , Cyclophosphamide/therapeutic use , Female , Graft Rejection , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Plasmacytoma/drug therapy , Plasmacytoma/pathology , Time Factors , Transplantation, Isogeneic
5.
J Natl Cancer Inst ; 79(3): 487-98, 1987 Sep.
Article in English | MEDLINE | ID: mdl-3114533

ABSTRACT

A method has been developed by which to amplify expression of phenotypic transformation of C3H/10T1/2 clone 8 mouse embryo cells not otherwise observed in the standard transformation assay. The expression of transformed foci was amplified by subcultivating chemically treated target cells after they had reached confluence and replating them at subconfluent cell densities. Conditions leading to the expression of the highest numbers of transformed foci include a) a cell seeding density for chemical treatment of 1 X 10(4) cells/dish, b) subculture 4 weeks after treatment, and c) replating cells at a density of 2 X 10(5) cells/-dish. Agents capable of inducing transformation in the standard assay (e.g., 4,4'-bis(dimethylamino)benzophenone, benzo[a]pyrene, 7,12-dimethylbenz[a]anthracene, and others) also yielded transformation in the replating assay. The more marginal transforming activities of chemicals such as ethyl methanesulfonate, 7-(bromomethyl)-12-methylbenz[a]anthracene, and N-methyl-N'-nitro-N-nitrosoguanidine were enhanced by the amplification procedure. Compounds that failed to elicit focal transformation in the standard assay (e.g., dibenz[a,h]anthracene, Tris(2,3-dibromopropyl) phosphate, lead acetate, benzidine, propyleneimine, N-hydroxy-2-fluorenylacetamide, and numerous other compounds of various chemical classes) induced significant levels of phenotypic transformation upon amplification. Noncarcinogens (e.g., phenanthrene, anthracene, 2-aminobiphenyl, cycloheximide, and others) failed to cause significant phenotypic transformation even when cells were replated. To further enhance the applicability of this new replating system, an exogenous source of metabolic activation was added: a 9,000 X g supernatant from Aroclor 1254-induced rat hepatic S-9. This activation system was found a) to be only minimally cytotoxic by itself and b) to be able to mediate NADPH-dependent, dose-dependent toxicity, and transformation by activating the procarcinogens dimethylnitrosamine, 2-naphthylamine, 2-aminoanthracene, and aflatoxin B1. With the use of this revised assay, 14 coded and 23 model compounds were tested. Agreement with in vivo results was observed to be over 85%. The marked sensitivity and discriminatory ability of this revised assay procedure suggest its usefulness as a screen for potential carcinogens of diverse chemical structure.


Subject(s)
Carcinogens , Cell Transformation, Neoplastic/drug effects , 2-Naphthylamine , Aflatoxin B1 , Aflatoxins , Animals , Biotransformation , Carcinogens/metabolism , Clone Cells , Diethylnitrosamine , Dimethylnitrosamine , Male , Methylcholanthrene , Methylnitronitrosoguanidine , Rats , Rats, Inbred F344
6.
J Natl Cancer Inst ; 71(5): 991-7, 1983 Nov.
Article in English | MEDLINE | ID: mdl-6316014

ABSTRACT

The standard C3H/10T1/2 clone 8 (C3H/10T1/2 CL8) cell transformation assay was tested for its ability to identify a variety of polycyclic hydrocarbons and alkylating agents. Dose-dependent morphologic transformation occurred with benzo[a]pyrene (BaP), 3-methylcholanthrene (MCA), 7,12-dimethylbenz[a]anthracene, BaP-7,8-dihydroxy-7,8-dihydrodiol (BaP-7,8-diol), as well as with the relatively weak in vivo carcinogen benzo[e]pyrene. Dibenz[a,h]anthracene yielded a relatively weak response, whereas anthracene and phenanthrene were negative. In contrast, treatment of C3H/10T1/2 CL8 cells with two directly acting alkylating agents, N-nitroso-N-methylnitroguanidine (MNNG) and styrene oxide, gave no transformation, whereas a third alkylating agent, ethyl methanesulfonate (EMS), gave a weak response. Treatment with MCA (2.5 micrograms/ml) yielded a reproducible positive response and, therefore, served as a positive control for routine use of the C3H/10T1/2 CL8 assay. When cells treated with the hydrocarbons BaP, BaP-7,8-diol, or MCA were analyzed for nonspecific DNA damage (single-strand breaks or alkaline-labile sites) by alkaline elution techniques, little if any DNA damage was observed. In contrast, the alkylating agents MNNG, styrene oxide, and EMS yielded substantial numbers of single-strand breaks.


Subject(s)
Alkylating Agents/toxicity , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Drug Evaluation, Preclinical/methods , Polycyclic Compounds/toxicity , Animals , Benzo(a)pyrene , Benzopyrenes/toxicity , Clone Cells , DNA Repair , Ethyl Methanesulfonate/toxicity , Methylcholanthrene/toxicity , Methylnitronitrosoguanidine/toxicity , Mice
7.
J Natl Cancer Inst ; 82(16): 1333-9, 1990 Aug 15.
Article in English | MEDLINE | ID: mdl-2380990

ABSTRACT

The major polycyclic aromatic hydrocarbon inducible-cytochrome P4501A1 gene (CYP1A1) is presumed to be important in pulmonary carcinogenesis and toxicology because its product, the cytochrome P4501A1-dependent (CYP1A1-dependent) monooxygenase, transforms selected xenobiotics (including polycyclic aromatic hydrocarbon procarcinogens in cigarette smoke) to potent carcinogenic metabolites. CYP1A1 messenger RNA (mRNA) expression has not, however, been previously demonstrated in human pulmonary tissue. This report defines CYP1A1 gene expression in normal lung tissue and primary pulmonary carcinoma tissue obtained at thoracotomy from 56 patients with lung cancer. When Northern blot hybridization analyses were performed, 17 of 19 (89%) and zero of five (0%) samples of normal lung tissue from active cigarette smokers and nonsmokers, respectively, expressed the normal 2.8-kilobase CYP1A1 mRNA. In addition, a time-dependent decrease in expression of the CYP1A1 gene was noted in normal lung tissue from individuals who were former smokers, with a decrease in expression occurring as early as 2 weeks following cessation of cigarette smoking. Expression became undetectable in all patients who had stopped smoking more than 6 weeks prior to study. When CYP1A1 gene expression was evaluated in lung cancers, mRNA levels were detectable in one of four (25%) tumors from nonsmokers; two of 24 (8%) tumors from former smokers; and seven of 15 (47%) tumors from cigarette smokers. In addition, an approximately 10-kilobase CYP1A1 RNA species, which was not detectable in normal lung tissue, was observed in five of ten (50%) of the lung cancers that expressed the CYP1A1 gene.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Neoplastic , Isoenzymes/biosynthesis , Lung Neoplasms/genetics , Lung/metabolism , Smoking/adverse effects , Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Female , Humans , Isoenzymes/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Oxidoreductases/metabolism , RNA, Messenger/analysis , RNA, Neoplasm/analysis
8.
Cancer Res ; 48(9): 2492-7, 1988 May 01.
Article in English | MEDLINE | ID: mdl-3356011

ABSTRACT

Male F344/NCr rats, 6 wk old, were fed 500 ppm of phenobarbital (PB) or equimolar doses of either 5-ethyl-5-phenylhydantoin (EPH) or 5,5-diethylhydantoin (EEH) in diet for 2 wk and hepatic cytochrome P-450-mediated alkoxyresorufin O-dealkylase and aminopyrine N-demethylase activities were determined. Both PB and EPH greatly increased P-450-mediated enzyme activities in rat liver while EEH was ineffective. To evaluate the hydantoins as tumor promoters, 5-wk-old male F344 rats were given a single i.p. injection of 75 mg N-nitrosodiethylamine/kg body weight. Beginning 2 wk later, they were placed either on normal diet or diet containing 500 ppm of PB or equimolar doses of EPH or EEH for the remaining experimental period. Control groups received an i.p. injection of saline followed by each of the test diets. Animals were sacrificed at either 52 or 78 wk. PB and EPH significantly enhanced the development of hepatocellular foci and hepatocellular adenomas at 52 wk and hepatocellular carcinomas at 78 wk in N-nitrosodiethylamine-initiated rats. Neither the incidence of hepatocellular neoplasms nor the number and size of hepatocellular foci was significantly increased by EEH. At 78 wk, both PB and EPH enhanced the development of thyroid follicular cell neoplasms in N-nitrosodiethylamine-initiated rats while no such enhancement was observed with EEH. Thus, EPH, a long-acting sedative/anticonvulsant, like the structurally similar PB, promoted hepatocellular and thyroid follicular cell carcinogenesis and induced the PB-inducible form(s) of cytochrome P-450 (P-450b) in rats. In contrast, EEH unlike barbital failed to promote hepatocellular and thyroid follicular cell carcinogenesis and also failed to induce PB-inducible form(s) of cytochrome P-450 in rats.


Subject(s)
Barbital/toxicity , Barbiturates/toxicity , Cytochrome P-450 Enzyme System/biosynthesis , Hydantoins/toxicity , Liver Neoplasms, Experimental/chemically induced , Mephenytoin/toxicity , Phenobarbital/toxicity , Thyroid Neoplasms/chemically induced , Animals , Body Weight/drug effects , Diethylnitrosamine , Enzyme Induction/drug effects , Liver/drug effects , Liver Neoplasms, Experimental/enzymology , Male , Mephenytoin/analogs & derivatives , Organ Size/drug effects , Rats , Rats, Inbred F344 , Thyroid Neoplasms/enzymology
9.
Cancer Res ; 60(18): 5040-4, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016626

ABSTRACT

Epidemiological and animal studies suggest that nonsteroidal anti-inflammatory drugs (NSAIDs) may reduce colon cancer risk. NSAIDs nonselectively inhibit both the constitutive cyclooxygenase (COX) 1 associated with side effects and the desired therapeutic target COX-2, which is induced in inflammation and neoplasia. We used the adenomatous polyposis coli (Apc) mutant Min mouse model to determine whether the selective COX-2 inhibitor celecoxib is effective for adenoma prevention and/or regression, and whether it might be safer than the nonselective NSAID previously shown to be most effective in this model, piroxicam. Min mice (n = 120) were randomized to treatment with celecoxib (0, 150, 500, or 1500 ppm celecoxib mixed in the diet) or piroxicam. To distinguish prevention from regression effects, groups were treated either "early" (before adenomas develop) or "late" (after most adenomas are established). Celecoxib caused dramatic reductions in both the multiplicity and size of tumors in a dose-dependent manner (P < 0.01). Early treatment with 1500 ppm of celecoxib was effective for prevention, decreasing tumor multiplicity to 29% and tumor size to only 17% of controls (P < 0.01). Late treatment demonstrated regression effects, reducing tumor multiplicity and size by about half. In contrast to the significant toxicity of piroxicam, which caused ulcers complicated by perforation and bleeding, celecoxib caused no gastrointestinal side effects and did not inhibit platelet thromboxane B2 at plasma drug levels similar to those obtained in early clinical trials in humans. These results provide the first evidence that selective inhibitors of COX-2 are safe and effective for the prevention and regression of adenomas in a mouse model of adenomatous polyposis and strongly support ongoing clinical trials in humans with the same syndrome. The broader population of patients with common sporadic adenomas that have somatic mutations of the same gene (APC) may also benefit from this treatment approach.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Anticarcinogenic Agents/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Sulfonamides/therapeutic use , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/pharmacology , Celecoxib , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Disease Models, Animal , Female , Isoenzymes/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Prostaglandin-Endoperoxide Synthases/metabolism , Pyrazoles , Substrate Specificity , Sulfonamides/pharmacology , Thromboxane B2/metabolism
10.
Cancer Res ; 59(14): 3387-91, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10416599

ABSTRACT

Epidemiological and model studies with laboratory animals have provided evidence that nonsteroidal anti-inflammatory drugs reduce the risk of colon cancer. Sulindac, a nonsteroidal anti-inflammatory drug, has been shown to inhibit azoxymethane (AOM)-induced colon carcinogenesis in rats when administered continuously before, during, and after carcinogen treatment (initiation and postinitiation periods) or when given continuously beginning 14 weeks after carcinogen administration (promotion/ progression stage). The present study was designed to investigate the chemopreventive efficacy of sulindac sulfone (exisulind), the sulfone metabolite of sulindac, when administered during the promotion/progression stage of colon carcinogenesis in comparison to the effect during the initiation and postinitiation periods. We have also studied the modulating effect of exisulind on colonic tumor apoptosis. At 5 weeks of age, groups of male F344 rats were fed diets containing 0%, 0.06%, and 0.12% exisulind. At 7 weeks of age, groups of animals were injected s.c. with AOM (15 mg/kg body weight, once weekly for 2 weeks). Animals intended for the promotion/progression study and receiving 0% exisulind were switched to an experimental diet containing 0.12% exisulind at 14 weeks after the second AOM treatment. All rats remained on their respective dietary regimens until the termination of the study, 50 weeks after the second AOM injection. Colon tumors were evaluated histopathologically for tumor type. Administration of 0.06% and 0.12% exisulind during the initiation and postinitiation periods significantly inhibited the incidence and multiplicity of invasive and/or noninvasive adenocarcinomas of the colon. The inhibition of colon tumorigenesis by exisulind was associated with a significant retardation of body weight gain shortly after sulfone administration and increased apoptosis in the colon tumors. In contrast, administration of the higher dose (0.12%) of exisulind during the promotion/progression stage had only minimal effects on colon tumorigenesis and apoptosis in the colon tumors, suggesting that early administration, but not late administration, may be required for chemopreventive efficacy of this drug.


Subject(s)
Adenocarcinoma/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anticarcinogenic Agents/administration & dosage , Colonic Neoplasms/prevention & control , Sulindac/analogs & derivatives , Adenocarcinoma/chemically induced , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Azoxymethane/administration & dosage , Carcinogens/administration & dosage , Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/chemically induced , Cyclooxygenase Inhibitors/pharmacology , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Neoplasm Invasiveness , Rats , Rats, Inbred F344 , Sulindac/administration & dosage , Sulindac/pharmacokinetics , Sulindac/pharmacology , Sulindac/therapeutic use , Weight Gain/drug effects
11.
Cancer Res ; 54(7 Suppl): 2015s-2024s, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8137331

ABSTRACT

The basic cancer-related chemical and biological sciences, pathology, and epidemiology have contributed to the understanding that antimutagenesis and antiproliferation are the important general mechanisms of chemoprevention and to the development of antimutagenic and anti-proliferative agents as potential chemopreventive drugs. These disciplines have also provided the biochemical and histopathological bases for identifying intermediate biomarkers that can be used as surrogate end points for cancer incidence in clinical chemoprevention trials and for selecting cohorts for these trials. Particularly important as histological biomarkers of cancer are the cytonuclear morphological and densitometric changes that define intraepithelial neoplasia (IEN). IEN changes are on the causal pathway to cancer. They may serve as target lesions in Phase II chemoprevention trials and as standards against which other earlier cellular and molecular biomarkers can be evaluated. Strategies for the clinical evaluation of chemopreventive agents have been defined for seven targets--colorectal, prostate, lung, breast, bladder, oral, and cervical cancers. Cohorts have been identified for short-term Phase II trials that investigate the effects of chemopreventive agents on IEN and on earlier biomarkers. Patients with adenomas serve as a cohort for trials in colon. One cohort for Phase II trials in prostate is patients with early stage cancers scheduled for prostatectomy; another is patients with prostatic intraepithelial neoplasia (without prostatic carcinoma). Patients treated for lung cancer are at high risk for bronchial dysplasia and second cancers; such patients are a cohort for Phase II trials in lung cancer. Presurgical breast cancer patients and patients with ductal or lobular carcinoma in situ are cohorts for studies in breast. Patients with superficial bladder cancers (Ta/T1 with or without carcinoma in situ) are cohorts for studies of chemoprevention in bladder, and patients with dysplastic oral leukoplakia are evaluated for chemoprevention of oral cancers. Cervical intraepithelial neoplasia is a prototype IEN, and patients with cervical intraepithelial neoplasia are a cohort for studies of cervical cancer.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Clinical Trials as Topic/methods , Neoplasms/prevention & control , Adenoma/epidemiology , Adenoma/prevention & control , Biomarkers, Tumor/analysis , Breast Neoplasms/epidemiology , Breast Neoplasms/prevention & control , Clinical Trials, Phase II as Topic/methods , Cohort Studies , Colonic Neoplasms/epidemiology , Colonic Neoplasms/prevention & control , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/prevention & control , Female , Humans , Male , Mouth Neoplasms/epidemiology , Mouth Neoplasms/prevention & control , Neoplasms/epidemiology , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/prevention & control , Research Design , United States/epidemiology , Urinary Bladder Neoplasms/epidemiology , Urinary Bladder Neoplasms/prevention & control , Uterine Neoplasms/epidemiology , Uterine Neoplasms/prevention & control
12.
Cancer Res ; 57(17): 3717-22, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9288778

ABSTRACT

Several phytochemicals and micronutrients that are present in fruits and vegetables are known to exert cancer chemopreventive effects in several organs, including the colon. Among them, the soybean isoflavonoid genistein received much attention due to its potential anticarcinogenic, antiproliferative effects and its potential role in several signal transduction pathways. The present study was designed to investigate the effect of genistein on azoxymethane (AOM)-induced colon carcinogenesis and to study its modulatory role on the levels of activity of 8-isoprostane, cyclooxygenase (COX), and 15-hydroxyprostaglandin F2alpha dehydrogenase (15-PGDH) in the colonic mucosa and colon tumors of male F344 rats. At 5 weeks of age, groups of male F344 rats were fed control (AIN-76A) diet or a diet containing 250 ppm genistein. Beginning 2 weeks later, all animals except those in the vehicle-treated groups were given weekly s.c. injections of AOM (15 mg/kg body weight) for 2 successive weeks. All rats were continued on their respective dietary regimen for 52 weeks after AOM treatment and were then sacrificed. Colon tumors were evaluated histopathologically. Colonic mucosae and tumors were analyzed for COX, 15-PGDH, and 8-isoprostane levels. Administration of genistein significantly increased noninvasive and total adenocarcinoma multiplicity (P < 0.01) in the colon, compared to the control diet, but it had no effect on the colon adenocarcinoma incidence nor on the multiplicity of invasive adenocarcinoma (P > 0.05). Also, genistein significantly inhibited the 15-PGDH activity (>35%) and levels of 8-iosoprostane (50%) in colonic mucosa and in tumors. In contrast, genistein had no significant effect on the COX synthetic activity, as measured by the rate of formation of prostaglandins and thromboxane B2 from [14C]arachidonic acid. The results of this investigation emphasize that the biological effects of genistein may be organ specific, inhibiting cancer development in some sites yet showing no effect or an enhancing effect on the tumorigenesis at other sites, such as the colon. The inhibition of 8-isoprostane levels by genistein indicates its possible antioxidant potential, which is independent of the observed colon tumor enhancement, yet this agent may also possess several biological effects that overshadow its antioxidant potential. The exact mechanism(s) of colon tumor enhancement by genistein remain to be elucidated; it is likely that its colon tumor-enhancing effects may, at least in part, be related to inhibition of prostaglandin catabolic enzyme activities.


Subject(s)
Adenocarcinoma/chemically induced , Anticarcinogenic Agents/pharmacology , Colonic Neoplasms/chemically induced , Isoflavones/pharmacology , Animals , Azoxymethane , Carcinogens , Colon/drug effects , Colon/metabolism , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Drug Synergism , F2-Isoprostanes , Genistein , Hydroxyprostaglandin Dehydrogenases/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Neoplasms, Experimental/chemically induced , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Inbred F344
13.
Cancer Res ; 60(2): 293-7, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667579

ABSTRACT

Epidemiological observations and laboratory research have suggested that nonsteroidal anti-inflammatory drugs (NSAIDs) reduce the risk of colon cancer and that the inhibition of colon carcinogenesis by NSAIDs is mediated through the modulation of prostaglandin production by rate-limiting enzymes known as cyclooxygenases (COXs). Because traditional NSAIDs inhibit both COX-1 and COX-2, these drugs induce side effects, such as gastrointestinal ulceration and renal toxicity, through the inhibition of the constitutive COX-1. Overexpression of COX-2 has been observed in colon tumors; therefore, specific inhibitors of COX-2 could serve as chemopreventive agents. Our previous study has shown that celecoxib, an inhibitor of COX-2, while sparing COX-1, inhibited azoxymethane (AOM)-induced colon tumorigenesis when administered during both initiation and postinitiation stages, ie., celecoxib administered continuously before, during, and after carcinogen treatment. This study examined the dose-response effect of celecoxib when administered during the initiation and postinitiation stages. In addition, the chemopreventive effects of high-dose celecoxib administered during the promotion/progression stage of colon carcinogenesis, ie., continuous celecoxib administration beginning 14 weeks after the carcinogen treatment, was determined in male F344 rats. We also measured the steady-state levels of celecoxib in the plasma of animals given this inhibitor. Groups of 5-week-old male F344 rats were fed either a control diet or experimental diets containing 500, 1000, or 1500 ppm celecoxib. At 7 and 8 weeks of age, rats scheduled for carcinogen treatment were injected s.c. with AOM at a dose rate of 15 mg/kg body weight/week. Groups of animals destined for the promotion/ progression study and initially receiving the control diet were switched to a diet containing 1500 ppm celecoxib beginning 14 weeks after the second AOM treatment. All rats remained on their respective dietary regimens until the termination of the study, ie., 52 weeks, and were then sacrificed. Colon tumors were evaluated histopathologically. Administration of 500, 1000, or 1500 ppm celecoxib during the initiation and postinitiation stages significantly inhibited the incidence (P < 0.01 to P < 0.0001) as well as the multiplicity (P < 0.01 to P < 0.0001) of adenocarcinomas of the colon in a dose-dependent manner. Importantly, administration of 1500 ppm celecoxib during the promotion/progression stage also significantly suppressed the incidence and multiplicity of adenocarcinomas of the colon (P < 0.01). Also, administration of celecoxib to the rats during the initiation and postinitiation periods and throughout the promotion/progression stage strongly suppressed colon tumor volume (P < 0.0002 to P < 0.001). The steady-state plasma concentration of celecoxib increases somewhat with the dose. Thus, in this model system, the chemopreventive efficacy of celecoxib is dose-dependent when this COX-2 inhibitor is administered during the initiation and postinitiation periods. This study provides the first evidence that celecoxib is also very effective when it is given during the promotion/progression stage of colon carcinogenesis, indicating that the chemopreventive efficacy is achieved during the later stages of colon tumor development. This suggests that celecoxib may potentially be an effective chemopreventive agent for the secondary prevention of colon cancer in patients with familial adenomatous polyposis and sporadic polyps.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Colonic Neoplasms/prevention & control , Cyclooxygenase Inhibitors/therapeutic use , Sulfonamides/therapeutic use , Administration, Oral , Animals , Azoxymethane/toxicity , Celecoxib , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/administration & dosage , Disease Progression , Isoenzymes/metabolism , Male , Prostaglandin-Endoperoxide Synthases/metabolism , Pyrazoles , Rats , Rats, Inbred F344 , Sulfonamides/administration & dosage , Time Factors
14.
Cancer Res ; 60(4): 901-7, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706103

ABSTRACT

Recent evidence indicates that individuals with a p53 germ-line mutation (Li-Fraumeni syndrome) have a 50% risk of developing lung cancer by age 60. In this study, p53 heterozygous knockout mice and p53 transgenic mice carrying a dominant negative mutant were crossed with the A/J mouse, which is highly susceptible to lung tumor induction, to investigate whether a p53 germ-line mutation is a predisposing gene for carcinogen-induced pulmonary adenomas in mice. The number of lung tumors was not significantly increased in (TSG-p53 x A/J)F1 p53 heterozygous knockout mice as compared with that in (TSG-p53 x A/J)F1 wt mice 16 weeks after exposure to N-nitrosomethylurea (MNU). In contrast, an average of 22 lung tumors were observed in (UL53-3 x A/J)F1 mice carrying a mutant p53 transgene (135Valp53) compared with an average of 7 lung tumors seen in (UL53-3 x A/J)F1 wt mice after treatment with N-nitrosomethylurea. Similar enhancement of lung tumor multiplicity (approximately 3-fold) was seen when mutant versus wt mice were treated with the tobacco-related carcinogens benzo[a]pyrene or 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. These results suggest that the mutant p53 transgene may have a dominant negative effect on the wt p53. The potential usefulness of this new mouse model in lung cancer chemoprevention and chemotherapy was examined. The chemopreventive efficacy of the green tea or a combination of dietary dexamethasone and myoinositol and the chemotherapeutic efficacy of Taxol or Adriamycin was examined in wt mice or mice with a mutation in the p53 gene. Mice treated with dexamethasone/myo-inositol and green tea displayed an average of 70 and 50% inhibition of lung tumors, respectively, regardless of p53 status. Similarly, when mice bearing established lung adenomas were treated with Taxol or Adriamycin, a decrease in tumor volume of approximately 70% was observed independent of p53 mutation status. Thus, the (UL53-3 x A/J)F1 p53 transgenic mouse seems to be an excellent model for human carriers of p53 germ-line mutations (Li-Fraumeni syndrome). Furthermore, the lung adenomas generated in this model possess mutations in both the K-ras proto-oncogene and the p53 tumor suppressor gene. This model should prove directly useful for chemoprevention and chemotherapy studies.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Dexamethasone/therapeutic use , Doxorubicin/therapeutic use , Genes, p53/physiology , Germ-Line Mutation , Inositol/therapeutic use , Lung Neoplasms/prevention & control , Paclitaxel/therapeutic use , Tea , Alleles , Animals , Humans , Lung Neoplasms/etiology , Methylnitrosourea , Mice , Mice, Transgenic , Nitrosamines , Proto-Oncogene Mas
15.
Cancer Res ; 61(20): 7449-55, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11606379

ABSTRACT

Female transgenic mice that express SV40 T/t antigens under the regulatory control of the rat C3(1) gene spontaneously develop multifocal mammary lesions that predictably evolve into invasive, hormone-independent carcinomas, whereas male mice are prone to develop prostate cancer. Chemopreventive agents were administered to female C3(1)/SV40 large T-antigen mice from 7 to 19 weeks of age, during which time the mammary lesions developed and progressed to invasive carcinomas. No significant differences in the numbers of preinvasive mammary intraepithelial neoplasia lesions (histologically similar to human ductal carcinoma in situ) were observed after 2 or 8 weeks of treatment between mice receiving either vehicle alone, dehydroepiandrosterone (DHEA), or 2-difluoromethylornithine (DFMO). However, a dose-response reduction in invasive carcinoma growth was observed for both DFMO, an inhibitor of ornithine decarboxylase, and DHEA, the primary steroid precursor to both androgens and estrogens in primates. Despite unaltered expression of the transgene, tumor incidence was reduced approximately 20% by DFMO (8000 mg/kg) and 30% by DHEA (4000 mg/kg; P < 0.05). Tumor multiplicity was reduced by approximately 50% by both DFMO and DHEA (P < 0.05). DFMO had a dose-dependent effect on total tumor burden, which was reduced by 25% at low doses (4000 mg/kg) and 70% at high doses (8000 mg/kg). DHEA reduced tumor burden by 50% and 66% at low (2000 mg/kg) and high (4000 mg/kg) doses, respectively. Interestingly, despite its inhibitory effects on tumor development, DHEA caused a dose-dependent increase of serum estradiol levels that we have previously shown to increase mammary tumor formation in this model. No effect on the development of the prostate cancer precursor lesions (prostate intraepithelial neoplasia) was observed when mice were treated with DHEA, DFMO, tocopherol acetate, selenomethionine, or 9-cis-retinoic acid, although the effects on late-stage prostate cancer development were not determined. These results demonstrate that despite the expression of the highly transforming C3(1)/SV40 large T-antigen transgene, this transgenic model can be used to study the effects of chemopreventive agents on mammary cancer progression. The tumor-inhibitory effects of DHEA and DFMO on mammary cancer growth appear to occur after the development of preinvasive lesions, suggesting that these agents inhibit tumor progression but not initiation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Dehydroepiandrosterone/pharmacology , Eflornithine/pharmacology , Mammary Neoplasms, Experimental/prevention & control , Prostatic Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/toxicity , Antigens, Polyomavirus Transforming/biosynthesis , Antigens, Polyomavirus Transforming/genetics , Apoptosis/drug effects , Cell Division/drug effects , Dehydroepiandrosterone/toxicity , Disease Models, Animal , Disease Progression , Eflornithine/toxicity , Estradiol/blood , Female , Gene Expression/drug effects , Male , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Precancerous Conditions/drug therapy , Precancerous Conditions/prevention & control , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats , Transgenes/drug effects
16.
Cancer Res ; 60(7): 1864-70, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10766173

ABSTRACT

Genetic knockout or pharmacological inhibition of cyclooxygenase-2 decreases the number and size of adenomas in mouse models of familial adenomatous polyposis. Epidemiological and clinical studies in humans indicate that the entire class of nonsteroidal anti-inflammatory drugs (NSAIDs) that inhibit both COX-1 and COX-2 enzymes are promising colon cancer chemopreventive agents. We used the Apc mutant Min mouse model to test combinations of agents that might maximize preventive benefit with minimal toxicity because they act via different mechanisms. Min mice (n = 144) were exposed to low doses of the nonselective COX inhibitor piroxicam and the ornithine decarboxylase (ODC) inhibitor difluoromethylornithine (DFMO), beginning at the time they were weaned and continuing throughout the duration of the experiment. Piroxicam at 12, 25, and 50 ppm in the diet caused dose-dependent decreases in the number of tumors in the middle and distal portions of the small intestine. This decrease in tumor multiplicity was associated with a striking decrease in the size of those tumors that did grow out. In contrast, none of the doses of piroxicam alone decreased tumor multiplicity in the proximal portion of the intestine (duodenum). Exposure to DFMO (0.5 or 1.0% in water) caused a dose-dependent decrease in tumor multiplicity in the middle and distal portions of the small intestine. However, this decreased multiplicity was not associated with a striking decrease in the size of the tumors. Combined treatment of mice with piroxicam plus DFMO was much more effective than either agent alone and resulted in a significant number of mice totally free of any intestinal adenomas (P < 0.001), in contrast to the 100% incidence and high multiplicity in control Min mice. In addition to this profound effectiveness in reducing tumor number, the few residual tumors in mice treated with the combined drugs were markedly smaller in size than tumors that arose from control Min mice. These experiments suggest that selective COX-2 inhibition combined with ODC inhibition is a very promising approach for colon cancer prevention. These COX-2 and ODC inhibitor drugs were not overtly toxic at the doses used when administered to mice after weaning. However, when treatment was begun in utero, the Mendelian expected progeny ratio of 1:1 that we routinely obtained in untreated control litters was no longer observed. Apc(min)/+ progeny of pregnant dams treated with piroxicam and/or DFMO were reduced in number and their ratio to Apc+/+ progeny was decreased to approximately 0.28:1. Thus, these agents are effective against adenomas that have homozygous mutation of the APC gene and also select against fetuses bearing a heterozygous mutation in the APC gene.


Subject(s)
Adenoma/prevention & control , Anticarcinogenic Agents/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Eflornithine/toxicity , Eflornithine/therapeutic use , Genes, APC , Intestinal Neoplasms/prevention & control , Piroxicam/toxicity , Piroxicam/therapeutic use , Adenoma/pathology , Animals , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Drug Therapy, Combination , Embryo, Mammalian/drug effects , Female , Intestinal Neoplasms/pathology , Isoenzymes/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Ornithine Decarboxylase Inhibitors , Pregnancy , Prostaglandin-Endoperoxide Synthases/metabolism
17.
Cancer Res ; 59(15): 3610-20, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10446971

ABSTRACT

To be informative for chemoprevention, animal models must both closely emulate human disease and possess surrogate endpoint biomarkers that facilitate rapid drug screening. This study elucidated site-specific histopathological and biochemical surrogate endpoint biomarkers of spontaneous epidermal carcinogenesis in K14-HPV16 transgenic mice and demonstrated that the incidence and severity of these markers were decreased by the ornithine decarboxylase (ODC) inhibitor difluoromethylornithine (DFMO). The cumulative incidence of visible epidermal cancers in 127 untreated transgenic mice was 42% by 52 weeks of age, most frequently affecting the chest as flat lesions in association with chronic ulcers, or in the ear as protuberant masses. Microscopic malignancies were detected in 39% of 32-week-old transgenic mice and were found to emerge from precursor lesions that were of two distinct types: dysplastic sessile ear papillomas and hyperproliferative follicular/interfollicular chest dysplasias. ODC activity and tissue polyamine contents were differentially elevated in ear and chest skin during carcinogenesis, such that there was a marked elevation of both parameters of polyamine metabolism as early as 4 weeks of age in the ear, whereas in the chest, polyamine metabolism was increased significantly only in the late stages of neoplastic progression and in epidermal cancers. Administration of 1.0% DFMO in the drinking water from 4 to 32 weeks of age prevented both visible and microscopic malignancies and significantly decreased the incidence of chest and ear precursor lesions. ODC activity and tissue putrescine content were markedly diminished by DFMO chemoprevention in ear skin, whereas there was a more modest decline of these parameters in chest skin. DFMO treatment of transgenic mice from 28 to 32 weeks of age was associated with an absence of ear cancer and a marked regression of dysplastic papillomas. In contrast, the results in chest skin were complex in that the severity of chest precursors diminished, but their incidence was unchanged, and microscopic cancers were still detectable within these lesions. Collectively, this study highlights the utility of multistage epidermal carcinogenesis in K14-HPV16 transgenic mice both for the study of the biology of, and as a screening tool for, novel drugs and chemopreventive regimens.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Squamous Cell/prevention & control , Eflornithine/therapeutic use , Epidermis/drug effects , Genes, Viral , Keratins/genetics , Papilloma/prevention & control , Papillomaviridae/genetics , Skin Neoplasms/prevention & control , Transgenes , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DNA Replication/drug effects , Disease Progression , Ear , Eflornithine/administration & dosage , Eflornithine/pharmacology , Epidermis/metabolism , Gene Expression Regulation , Keratin-14 , Mice , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Organ Specificity , Ornithine Decarboxylase Inhibitors , Papilloma/genetics , Papilloma/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Precancerous Conditions/prevention & control , Putrescine/biosynthesis , Skin Diseases/genetics , Skin Diseases/pathology , Skin Diseases/prevention & control , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Thorax
18.
Cancer Res ; 59(17): 4285-90, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10485473

ABSTRACT

7,12-Dimethylbenz(a)anthracene (DMBA) is a prototype carcinogen that induces a high yield of mammary tumors in rats after a single feeding. We investigated the induction and chemoprevention of DNA adducts in female Sprague Dawley rats receiving DMBA by gavage according to a variety of treatment schedules. The patterns of 32P-postlabeled DNA adducts in liver and mammary epithelial cells were similar to those produced by the in vitro reaction of metabolically activated DMBA with calf thymus DNA. There was a high and statistically significant correlation between dose of DMBA administered to rats (0, 0.6, 2.4, and 12 mg/kg body weight) and levels of DNA adducts in both types of cells. The regression lines relating DMBA doses to total DNA adduct levels were significantly divergent and crossed at 1.5 mg/kg body weight, indicating that, at lower doses, the formation of DNA adducts is more intense in target mammary cells, whereas at higher doses, DNA adduct levels are more elevated in liver cells, presumably due to the greater metabolic capacity of this organ. When the rats were sacrificed 7 days rather than 2 days after DMBA administration, DNA adduct levels were approximately halved in both liver and mammary cells. The observed patterns can be interpreted based on toxicokinetic factors, local and distant metabolism, removal of DNA adducts by excision repair, and cell proliferation rate. Of three chemopreventive agents given with the diet to rats treated with 12 mg of DMBA, 5,6-benzoflavone (1650 ppm) was the most effective, inhibiting DNA adduct formation in liver and mammary cells by 96.5 and 83.5%, respectively. Feeding of 1,2-dithiole-3-thione (600 ppm) inhibited this biomarker by 68.5 and 50.2%, whereas butyl hydroxyanisole (BHA; 5000 ppm) showed a significant inhibition in the liver (46.5%) but was ineffective in mammary cells (29.0%, not significant). These data correlate nicely with the results of a parallel study in which 5,6-benzoflavone, 1,2-dithiole-3-thione, and BHA inhibited formation of hemoglobin adducts by 80.0, 44.0, and 0%, respectively; the incidence of mammary tumors by 82.4, 47.1, and 5.9%, respectively; and their multiplicity by 92.6, 80.0, and 7.4%, respectively. Therefore, biomarkers of biologically effective dose are highly predictive of the efficacy of chemopreventive agents in the DMBA rat mammary model. The selective inhibition by BHA of DNA adducts in the liver but not in mammary cells is consistent with the finding that this phenolic antioxidant stimulated phase II activities in the liver but not in the mammary gland (L. L. Song et al., manuscript in preparation). In any case, the broad-spectrum inducer 5,6-BF appears to be more effective than the two monofunctional phase II inducers, presumably because an enhanced activation of DMBA to reactive metabolites is coordinated with their blocking, detoxification, and excretion.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/metabolism , Anticarcinogenic Agents/pharmacology , Carcinogens/metabolism , DNA Adducts/metabolism , Liver/drug effects , Mammary Glands, Animal/drug effects , Animals , Butylated Hydroxyanisole/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Glutathione/metabolism , Liver/metabolism , Mammary Glands, Animal/metabolism , Rats , Rats, Sprague-Dawley , Thiones/pharmacology , Thiophenes/pharmacology
19.
Cancer Res ; 56(8): 1724-6, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620482

ABSTRACT

To determine if the chemopreventive activity of dehydroepiandrosterone (DHEA) in the rat mammary gland can be dissociated from its toxicity, two studies were conducted in which low doses of DHEA were administered alone and in combination with other agents to rats treated with N-methyl-N-nitrosourea. Beginning 1 week prior to administration of 35 mg N-methyl-N-nitrosourea per kg body weight, groups of 20 female Sprague-Dawley rates were fed AIN-76A diet supplemented with DHEA alone (800 or 400 mg/kg diet), DHEA + tamoxifen (80 or 40 microgram/kg diet), DHEA + carbenoxolone (3500 or 1750 mg/kg diet), or DHEA + tamoxifen + carbenoxolone. When administered alone at either 800 or 400 mg/kg diet, DHEA reduced mammary cancer incidence from >70% in dietary controls to 0%; mammary cancer incidence from >70% in dietary controls to 0%; mammary cancer incidence in all DHEA combination regimens was also < or = 5%. The dose levels of DHEA used induced no toxicity or alteration in body weight gain. These results indicate that dietary supplementation with low doses of DHEA has chemopreventive efficacy greater than or equal to that of endocrine ablation. This protection may be mediated by the induction of differentiation in the mammary parenchyma.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carbenoxolone/pharmacology , Dehydroepiandrosterone/therapeutic use , Mammary Neoplasms, Experimental/prevention & control , Tamoxifen/pharmacology , Animals , Anticarcinogenic Agents/administration & dosage , Carcinogens , Dehydroepiandrosterone/administration & dosage , Diet , Drug Administration Schedule , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Rats , Rats, Sprague-Dawley
20.
Cancer Res ; 57(3): 420-5, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9012468

ABSTRACT

Epidemiological studies suggest that consumption of diets containing fruits and vegetables, major sources of phytochemicals and micronutrients, may reduce the risk of developing cancer of the colon. Several phytochemicals and micronutrients present in fruits and vegetables are known to exert cancer-chemopreventive effects in several organs, including the colon. Monoterpenes such as d-limonene and perillyl alcohol derived from orange peels and lavender, respectively, have been shown to possess chemopreventive properties against mammary, liver, and/or lung carcinogenesis. The present study was designed to investigate the efficacy of dietary 40 and 80% maximum tolerated dose (MTD) levels of perillyl alcohol on azoxymethane (AOM)-induced colon carcinogenesis. The effect of this agent on the process of apoptosis in colon tumors was also investigated. Prior to the efficacy study, the MTD of perillyl alcohol was determined in male F344 rats in a 6-week subchronic toxicity study and found to be a 2.5-g/kg diet when added to the AIN-76A diet. At 5 weeks of age, groups of male F344 rats were fed control (AIN-76A) diet or diets containing 1 and 2 g perillyl alcohol/kg diet, representing 40 and 80% MTD levels, respectively. At 7 weeks of age, all animals except those in the vehicle-treated groups were given two weekly s.c. injections of AOM (15 mg/kg body weight/week). All animals were continued on their respective dietary regimen for 52 weeks after AOM treatment and then sacrificed. Colon tumors were evaluated histopathologically using routine procedures. Perillyl alcohol at the 1-g/kg level significantly inhibited the incidence (percentage of animals with tumors) and multiplicity (tumors/ animals) of invasive adenocarcinomas of the colon, whereas perillyl alcohol at 2 g/kg diet inhibited the incidence of total adenocarcinomas of the colon and small intestine as compared to the control diet. Our studies also indicate that the colon tumors of animals fed perillyl alcohol exhibited increased apoptosis as compared to those fed the control diet. These results demonstrate the potential chemopreventive activity of perillyl alcohol against colon carcinogenesis. The chemopreventive activity of perillyl alcohol is mediated through the tumor cell loss by apoptosis.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/administration & dosage , Colonic Neoplasms/prevention & control , Monoterpenes , Terpenes/administration & dosage , Animals , Apoptosis/drug effects , Azoxymethane/toxicity , Diet , Male , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL