Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Bioelectromagnetics ; 39(3): 190-199, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29537695

ABSTRACT

Radiofrequency radiation (RFR) causes heating, which can lead to detrimental biological effects. To characterize the effects of RFR exposure on body temperature in relation to animal size and pregnancy, a series of short-term toxicity studies was conducted in a unique RFR exposure system. Young and old B6C3F1 mice and young, old, and pregnant Harlan Sprague-Dawley rats were exposed to Global System for Mobile Communication (GSM) or Code Division Multiple Access (CDMA) RFR (rats = 900 MHz, mice = 1,900 MHz) at specific absorption rates (SARs) up to 12 W/kg for approximately 9 h a day for 5 days. In general, fewer and less severe increases in body temperature were observed in young than in older rats. SAR-dependent increases in subcutaneous body temperatures were observed at exposures ≥6 W/kg in both modulations. Exposures of ≥10 W/kg GSM or CDMA RFR induced excessive increases in body temperature, leading to mortality. There was also a significant increase in the number of resorptions in pregnant rats at 12 W/kg GSM RFR. In mice, only sporadic increases in body temperature were observed regardless of sex or age when exposed to GSM or CDMA RFR up to 12 W/kg. These results identified SARs at which measurable RFR-mediated thermal effects occur, and were used in the selection of exposures for subsequent toxicology and carcinogenicity studies. Bioelectromagnetics. 39:190-199, 2018. © 2018 The Authors. Bioelectromagnetics Published by Wiley Periodicals, Inc.


Subject(s)
Body Temperature/radiation effects , Cell Phone , Radiation Exposure/adverse effects , Radio Waves/adverse effects , Aging/physiology , Animals , Female , Mice , Pilot Projects , Pregnancy , Rats , Rats, Sprague-Dawley
2.
PLoS One ; 10(10): e0141849, 2015.
Article in English | MEDLINE | ID: mdl-26516762

ABSTRACT

Peroxisome-proliferator-activated receptor γ (PPARγ) is a ligand-activated transcription factor that regulates cell proliferation, differentiation, and apoptosis. In vivo studies were performed to evaluate the activities of two thiazolidinedione PPARγ agonists, rosiglitazone and pioglitazone, as inhibitors of oral carcinogenesis in rats. Oral squamous cell carcinomas (OSCC) were induced in male F344 rats by 4-nitroquinoline-1-oxide (NQO; 20 ppm in the drinking water for 10 weeks). In each study, groups of 30 NQO-treated rats were exposed to a PPARγ agonist beginning at week 10 (one day after completion of NQO administration) or at week 17 (7 weeks post-NQO); chemopreventive agent exposure was continued until study termination at week 22 (rosiglitazone study) or week 24 (pioglitazone study). Administration of rosiglitazone (800 mg/kg diet) beginning at week 10 increased survival, reduced oral cancer incidence, and reduced oral cancer invasion score in comparison to dietary controls; however, chemopreventive activity was largely lost when rosiglitazone administration was delayed until week 17. Administration of pioglitazone (500 mg/kg diet beginning at week 10 or 1000 mg/kg diet beginning at week 17) induced significant reductions in oral cancer incidence without significant effects on OSCC invasion scores. Transcript levels of PPARγ and its three transcriptional variants (PPARγv1, PPARγv2, and PPARγv3) were not significantly different in OSCC versus age- and site-matched phenotypically normal oral tissues from rats treated with NQO. These data suggest that PPARγ provides a useful molecular target for oral cancer chemoprevention, and that overexpression of PPARγ at the transcriptional level in neoplastic lesions is not essential for chemopreventive efficacy.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , PPAR gamma/agonists , Quinolones/therapeutic use , Thiazolidinediones/therapeutic use , 4-Nitroquinoline-1-oxide/administration & dosage , 4-Nitroquinoline-1-oxide/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Male , PPAR gamma/genetics , PPAR gamma/metabolism , Pioglitazone , Quinolones/administration & dosage , Rats , Rats, Inbred F344 , Rosiglitazone , Thiazolidinediones/administration & dosage
3.
Toxicol Appl Pharmacol ; 281(3): 303-9, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25448049

ABSTRACT

NSC-743380 (1-[(3-chlorophenyl)-methyl]-1H-indole-3-carbinol) is in early stages of development as an anticancer agent. Two metabolites reflect sequential conversion of the carbinol functionality to a carboxaldehyde and the major metabolite, 1-[(3-chlorophenyl)-methyl]-1H-indole-3-carboxylic acid. In an exploratory toxicity study in rats, NSC-743380 induced elevations in liver-associated serum enzymes and biliary hyperplasia. Biliary hyperplasia was observed 2 days after dosing orally for 2 consecutive days at 100mg/kg/day. Notably, hepatotoxicity and biliary hyperplasia were observed after oral administration of the parent compound, but not when major metabolites were administered. The toxicities of a structurally similar but pharmacologically inactive molecule and a structurally diverse molecule with a similar efficacy profile in killing cancer cells in vitro were compared to NSC-743380 to explore scaffold versus target-mediated toxicity. Following two oral doses of 100mg/kg/day given once daily on two consecutive days, the structurally unrelated active compound produced hepatic toxicity similar to NSC-743380. The structurally similar inactive compound did not, but, lower exposures were achieved. The weight of evidence implies that the hepatotoxicity associated with NSC-743380 is related to the anticancer activity of the parent molecule. Furthermore, because biliary hyperplasia represents an unmanageable and non-monitorable adverse effect in clinical settings, this model may provide an opportunity for investigators to use a short-duration study design to explore biomarkers of biliary hyperplasia.


Subject(s)
Acute Disease , Biliary Tract Diseases/chemically induced , Biliary Tract/drug effects , Indoles/adverse effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Biliary Tract/metabolism , Biliary Tract/pathology , Biliary Tract Diseases/blood , Biliary Tract Diseases/metabolism , Biliary Tract Diseases/pathology , Biomarkers/blood , Biotransformation , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacokinetics , Hyperplasia , Indoles/administration & dosage , Indoles/blood , Indoles/metabolism , Indoles/pharmacokinetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Random Allocation , Rats, Inbred F344 , Structure-Activity Relationship
4.
Cancer Chemother Pharmacol ; 69(5): 1301-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22302406

ABSTRACT

PURPOSE: CP-31398 (N0-[2-[(E)-2-(4-methoxyphenyl)ethenyl] quinazolin-4-yl]-N,N-dimethylpropane-1,3-diamine hydrochloride) is one of the new class of agents that can stabilize the DNA-binding domain of p53 and thereby maintain the activity of p53 as a tumor suppressor and transcription factor. Through its activity as a p53 stabilizer, CP-31398 demonstrates significant cancer preventive and therapeutic activity in several in vivo animal models. The objective of the current study was to describe the pharmacokinetic profile and tissue distribution of this novel agent following intravenous or oral (gavage and dietary) administration. METHODS: CP-31398 was administered to male CD and F344 rats as a single intravenous bolus dose or by daily oral gavage dosing. Male F344 rats also received drug as an ad libitum dietary supplement. Plasma, liver, skin, colon, and colon tumor samples were collected after oral dosing. Concentrations of CP-31398 in plasma and tissue samples were analyzed using LC­MS/MS, and the resultant data were subjected to a non-compartmental pharmacokinetic analysis. RESULTS: Bioavailability (12­32%), elimination half-life (14­20 h), clearance (4.2­4.8 l/h/kg), and volume of distribution (70­82 l/kg) were determined. Tissue levels of CP-31398 after oral (gavage or diet) administration were several orders of magnitude higher than were corresponding plasma concentrations; CP-31398 levels were especially high in colon and liver. Levels of CP-31398 in tissues were higher after gavage dosing than after dietary administration. CONCLUSIONS: CP-31398 is bioavailable and has a relatively long elimination half-life, which supports the achievement of plasma steady-state levels with a once daily dosing regimen. CP-31398 exhibits a dramatically high volume of distribution, which is consistent with its tissue concentrations being much higher than corresponding plasma levels. It is accumulated in colon tumor tissues, albeit at lower concentrations than found in liver, skin, and colon.


Subject(s)
Colon/metabolism , Liver/metabolism , Pyrimidines/pharmacokinetics , Tumor Suppressor Protein p53/drug effects , Administration, Oral , Animals , Biological Availability , Chromatography, Liquid , Half-Life , Injections, Intravenous , Male , Pyrimidines/administration & dosage , Rats , Rats, Inbred F344 , Tandem Mass Spectrometry , Tissue Distribution , Tumor Suppressor Protein p53/metabolism
5.
Int J Toxicol ; 30(2): 143-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21378370

ABSTRACT

A recombinant ricin toxin A-chain 1-33/44-198 vaccine (RVEc) was administered to male and female New Zealand white (NZW) rabbits (10/sex/group) in a repeat-dose toxicity study. The RVEc vaccine was administered on study days 1, 29, 57, and 85 via intramuscular (IM) injection (0, 100, or 200 µg/dose). All study animals were observed throughout treatment until euthanized and submitted for necropsy on study day 88 or 99 (recovery period). There were no treatment-related or toxicologically significant effects observed. There were no statistically significant differences noted in the antibody titers and/or concentrations in 100 µg RVEc-treated animals when compared to 200 µg RVEc-treated animals, suggesting that both doses produced comparable antibody titers/concentrations during the study. The highest immune response was observed on study day 99 (ie, 2 weeks after the last dose). The immune response observed demonstrated that RVEc is biologically active in the rabbit model, with no apparent marked sex differences.


Subject(s)
Dose-Response Relationship, Immunologic , Ricin/administration & dosage , Ricin/toxicity , Vaccines, Synthetic/administration & dosage , Animals , Antibodies, Neutralizing/blood , Biological Products , Biological Warfare Agents , Drug Evaluation, Preclinical , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Immunoglobulin G/blood , Male , Models, Animal , No-Observed-Adverse-Effect Level , Rabbits , Ricin/immunology , Vaccines, Synthetic/immunology
6.
Cancer Chemother Pharmacol ; 68(2): 513-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21085965

ABSTRACT

PURPOSE: Characterize the preclinical pharmacokinetics, metabolic profile, multi-species toxicology, and antitumor efficacy of azurin-p28 (NSC 745104), an amphipathic, 28 amino acid fragment (aa 50-77) of the copper containing redox protein azurin that preferentially enters cancer cells and is currently under development for treatment of p53-positive solid tumors. METHODS: An LC/MS/MS assay was developed, validated, and applied to liver microsomes, serum, and tumor cells to assess cellular uptake and metabolic stability. Pharmacokinetics was established after administration of a single intravenous dose of p28 in preclinical species undergoing chronic toxicity testing. Antitumor efficacy was assessed on human tumor xenografts. A human therapeutic dose was predicted based on efficacy and pharmacokinetic parameters. RESULTS: p28 is stable, showed tumor penetration consistent with selective entry into tumor cells and significantly inhibited p53-positive tumor growth. Renal clearance, volume of distribution, and metabolic profile of p28 was relatively similar among species. p28 was non-immunogenic and non-toxic in mice and non-human primates (NHP). The no observed adverse effect level (NOAEL) was 120 mg/kg iv in female mice. A NOAEL was not established for male mice due to decreased heart and thymus weights that was reversible and did not result in limiting toxicity. In contrast, the NOAEL for p28 in NHP was defined as the highest dose (120 mg/kg/dose; 1,440 mg/m(2)/dose) studied. The maximum-tolerated dose (MTD) for subchronic administration of p28 to mice is >240 mg/kg/dose (720 mg/m(2)/dose), while the MTD for subchronic administration of p28 to Cynomolgous sp. is >120 mg/kg (1,440 mg/m(2)/dose). The efficacious (murine) dose of p28 was 10 mg/kg ip per day. CONCLUSIONS: p28 does not exhibit preclinical immunogenicity or toxicity, has a similar metabolic profile among species, and is therapeutic in xenograft models.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Azurin/adverse effects , Azurin/pharmacokinetics , Neoplasms/drug therapy , Peptide Fragments/pharmacokinetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Azurin/metabolism , Azurin/therapeutic use , Biotransformation , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Nude , No-Observed-Adverse-Effect Level , Peptide Fragments/adverse effects , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Specific Pathogen-Free Organisms , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
7.
Int J Toxicol ; 29(2): 157-64, 2010.
Article in English | MEDLINE | ID: mdl-20335511

ABSTRACT

The synthetic retinoic acid analog, 9-cis-UAB30 [(2E,4E,6Z,8E)-8-(3',4'-dihydro-1'(2'H)-naphthalen-1'-ylidene)-3,7-dimethyl-2,4,6-octatrienoic acid], is a specific ligand for the retinoid X receptor. Murine oncogenicity and pharmacokinetics studies were performed as part of the preclinical development of 9-cis-UAB30 for breast cancer chemoprevention. In the oncogenicity study, TSG-p53((+/-)) (p53 knockout) mice (25 per sex per group) received daily gavage exposure to 9-cis-UAB30 doses of 0 (control), 30, 100, or 300 mg/kg/d for 6 months. Positive controls received p-cresidine (400 mg/kg/d) for 6 months. 9-cis-UAB30 had no biologically significant effects on survival, body weight, body weight gain, clinical signs, hematology, or clinical chemistry but induced dose-related hepatomegaly in both sexes and decreased thymus weights in high-dose females. Gross and microscopic pathology provided no evidence of 9-cis-UAB30 toxicity or oncogenicity; by contrast, p-cresidine induced urinary bladder neoplasms in more than 60% of male and female mice. It was concluded that 9-cis-UAB30 is not oncogenic in p53((+/-)) mice. In the pharmacokinetics study, C57BL/6 mice received daily gavage exposure to 9-cis-UAB30 (100 or 300 mg/kg/d) for 1 or 7 days. Pharmacokinetic parameters were similar after 1 and 7 days of dosing. Dose-related peak plasma levels of 9-cis-UAB30 were seen between 0.25 and 3 hours; volume of distribution was comparable at both dose levels. Increases in area under the curve were less than proportional to dose and were associated with an increased rate of apparent clearance and decreased elimination half-life. These results suggest decreased absorption and/or possible induction of clearance mechanisms. Enzyme induction may underlie the hepatomegaly seen in mice treated with 9-cis-UAB30 for 6 months in the oncogenicity study.


Subject(s)
Breast Neoplasms/prevention & control , Fatty Acids, Unsaturated/toxicity , Naphthalenes/toxicity , Animals , Area Under Curve , Carcinogenicity Tests , Dose-Response Relationship, Drug , Fatty Acids, Unsaturated/blood , Fatty Acids, Unsaturated/pharmacokinetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthalenes/blood , Naphthalenes/pharmacokinetics , Retinoid X Receptors/agonists , Tumor Suppressor Protein p53/genetics
8.
Cancer Prev Res (Phila) ; 3(1): 73-81, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20051374

ABSTRACT

Oral squamous cell carcinomas induced in rats by 4-nitroquinoline-1-oxide (NQO) show substantial overexpression of cyclooxygenase-2 (COX-2) when compared with adjacent phenotypically normal oral tissues. By contrast, neither 5-lipoxygenase (LOX) nor 12-LOX is overexpressed in rat oral cancers. Two chemoprevention studies were done to test the resulting hypothesis that COX-2 is a useful target for oral cancer chemoprevention in the rat. In both studies, male F344 rats received drinking water exposure to NQO (20 ppm) for 10 weeks, followed by administration of chemopreventive agents from week 10 until study termination at week 26. In the first study, groups of rats were fed basal diet (control), or basal diet supplemented with the selective COX-2 inhibitor celecoxib (500 or 1,500 mg/kg diet), the nonselective COX inhibitor piroxicam (50 or 150 mg/kg diet), or the 5-LOX inhibitor zileuton (2,000 mg/kg diet). In the second study, rats were fed basal diet (control) or basal diet supplemented with nitric oxide-naproxen (180 or 90 mg/kg diet), a nonselective COX inhibitor that shows reduced gastrointestinal toxicity. When compared with dietary controls, celecoxib decreased oral cancer incidence, cancer invasion score, and cancer-related mortality. Piroxicam decreased cancer-related mortality and cancer invasion score, whereas nitric oxide-naproxen decreased oral cancer incidence and cancer invasion score. By contrast, zileuton showed no chemopreventive activity by any parameter assessed. These data show that both selective and nonselective inhibitors of COX-2 can prevent NQO-induced oral carcinogenesis in rats. The chemopreventive activity of COX inhibitors may be linked to overexpression of their enzymatic target in incipient oral neoplasms.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/prevention & control , Cyclooxygenase 2/biosynthesis , Cyclooxygenase Inhibitors/pharmacology , Mouth Neoplasms/enzymology , Mouth Neoplasms/prevention & control , Animals , Celecoxib , Gene Expression/drug effects , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Lipoxygenase Inhibitors/pharmacology , Male , Naproxen/pharmacology , Piroxicam/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Up-Regulation
9.
Toxicology ; 248(1): 8-17, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18423834

ABSTRACT

Non-clinical studies were conducted to evaluate the toxicity of Antalarmin, a corticotropin-releasing hormone type 1 receptor antagonist being developed for therapy of stress-related pathologies. Antalarmin was not genotoxic in bacterial mutagenesis assays, mammalian cell mutagenesis assays, or in vivo DNA damage assays. In a 14-day range-finding study in rats, Antalarmin doses >or=500 mg/kg/day (3,000 mg/m(2)/day) induced mortality. In a 90-day toxicity study in rats, no gross toxicity was seen at doses of 30, 100, or 300 mg/kg/day (180, 600, or 1,800 mg/m(2)/day, respectively). Antalarmin (300 mg/kg/day) induced mild anemia, increases in serum gamma-glutamyl transferase activity, and microscopic hepatic pathology (bile duct hyperplasia and epithelial necrosis, periportal inflammation). Microscopic renal changes (cortical necrosis, inflammation, hypertrophy, nephropathy) were observed in rats at all Antalarmin doses. In a 14-day range-finding study in dogs, Antalarmin doses >or=50mg/kg/day (1,000 mg/m(2)/day) induced repeated emesis and bone marrow suppression. In a 90-day toxicity study in dogs, Antalarmin (4, 8, or 16 mg/kg/day (80, 160, or 320 mg/m(2)/day, respectively)) induced bone marrow and lymphoid depletion, but no gross toxicity. Comparative in vitro studies using rat, dog, and human neutrophil progenitors demonstrated that canine bone marrow cells are highly sensitive to Antalarmin cytotoxicity, while rat and human bone marrow cells are relatively insensitive. As such, the bone marrow toxicity observed in dogs is considered likely to over-predict Antalarmin toxicity in humans. The hepatic and renal toxicities seen in rats exposed to Antalarmin identify those tissues as the most likely targets for Antalarmin toxicity in humans.


Subject(s)
Pyrimidines/toxicity , Pyrroles/toxicity , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Line, Tumor , Colony-Forming Units Assay , Dogs , Female , Humans , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Mice , Mutagenicity Tests , No-Observed-Adverse-Effect Level , Rats , Rats, Inbred F344 , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Vomiting/chemically induced
10.
Vaccine ; 26(14): 1773-85, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18308432

ABSTRACT

Tularemia is a severe disease for which there is no licensed vaccine. An attenuated F. tularensis live vaccine strain (LVS) was protective when administered to humans but safety concerns precluded its licensure and use in large-scale immunization. An improved F. tularensis LVS preparation was produced under current good manufacturing practice (cGMP) guidelines for evaluation in clinical trials. Preclinical safety, tolerability and immunogenicity were investigated in rabbits that received LVS in escalating doses (1 x 10(5)-1 x 10(9)CFU) by the intradermal, subcutaneous or percutaneous (scarification) route. This improved LVS formulation was well tolerated at all doses; no death or adverse clinical signs were observed and necropsies showed no signs of pathology. No live organisms were detected in liver or spleen. Transient local reactogenicity was observed after scarification injection. Erythema and edema developed after intradermal injection in the highest dose cohorts. High levels of F. tularensis-specific IgM, IgG and IgA developed early after immunization, in a dose-dependent fashion. Scarification elicited higher levels of IgA. Antibodies elicited by LVS also recognized F. tularensis Schu-S4 antigens and there was a significant correlation between antibody titers measured against both LVS and Schu-S4. The ELISA titers also correlated closely with those measured by microagglutination. This is the first report describing comprehensive toxicological and immunological studies of F. tularensis LVS in rabbits. This animal model, which closely resembles human disease, proved adequate to assess safety and immunogenicity of F. tularensis vaccine candidates. This new LVS vaccine preparation is being evaluated in human clinical studies.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Agglutination Tests , Animals , Antibody Formation/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/adverse effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Injections, Intradermal , Injections, Subcutaneous , Kinetics , Male , Rabbits , United States , United States Food and Drug Administration , Vaccines, Attenuated/immunology
11.
Toxicol Sci ; 99(2): 612-27, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17630414

ABSTRACT

Tamoxifen (TAM) is a nonsteroidal antiestrogen that prevents estrogen receptor-positive breast cancer in rodents and humans. Bexarotene (BEX), a selective agonist for retinoid X receptors, inhibits mammary carcinogenesis in rodents. The present study was conducted to support the preclinical development of TAM (tamoxifen citrate) + BEX for use in breast cancer chemoprevention, and to investigate the influence of these agents on hepatic gene expression. Female CD rats (20 per group) received daily oral (gavage) exposure to TAM (0 or 60 microg/kg/day) and/or BEX (0, 5, 15, or 45 mg/kg/day) for a minimum of 90 days. BEX induced mild, dose-related anemia and dose-related increases in serum alkaline phosphatase, cholesterol, triglycerides, and calcium levels, and increased platelet counts. TAM had no biologically significant effect on any clinical pathology parameter and did not alter the effects of BEX on these endpoints. Microscopic alterations induced by BEX included epidermal hyperplasia, hyperkeratosis (stomach), and cytoplasmic clearing (liver). Microscopic changes in TAM-treated rats were limited to mucous cell hypertrophy in the cervix and vagina. The toxicity of administration of the combination of TAM + BEX can generally be predicted on the basis of the toxicity of each drug as a single agent. BEX induced dose-related alterations in the expression of several genes involved in steroid, drug, and/or fatty acid metabolism; TAM did not alter these effects of BEX. Differential expression of genes involved in drug and lipid metabolism may underlie the observed effects of BEX on cholesterol and triglyceride levels and its effects on liver histology.


Subject(s)
Tamoxifen/toxicity , Tetrahydronaphthalenes/toxicity , Animals , Bexarotene , Dimerization , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , Liver/drug effects , Liver/metabolism , Oligonucleotide Array Sequence Analysis , PPAR alpha/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/agonists , Retinoid X Receptors/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/blood , Tetrahydronaphthalenes/blood , Toxicogenetics
12.
Chem Biol Interact ; 152(2-3): 79-99, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15840382

ABSTRACT

Farnesol demonstrates antitumor activity in several animal models for human cancer and was being considered for development as a cancer chemopreventive agent. This study was performed to characterize the effects of minimally toxic doses of farnesol on the activity of phase I and II drug metabolizing enzymes. CD((R)) rats (20/sex/group) received daily gavage exposure to farnesol doses of 0, 500, or 1000 mg/kg/day for 28 days; 10 rats/sex/group were necropsied at the termination of farnesol exposure; remaining animals were necropsied after a 28-day recovery period. No deaths occurred during the study, and farnesol had no significant effects on body weight, food consumption, clinical signs, or hematology/coagulation parameters. Modest but statistically significant alterations in several clinical chemistry parameters were observed at the termination of farnesol exposure; all clinical pathology effects were reversed during the recovery period. At the termination of dosing, the activities of CYP1A, CYP2A1-3, CYP2B1/2, CYP2C11/12, CYP2E1, CYP3A1/2, CYP4A1-3, CYP19, glutathione reductase, NADPH/quinone oxidoreductase and UDP-glucuronosyltransferase were significantly increased in the livers of farnesol-treated rats; farnesol also increased the activity of glutathione S-transferase in the kidney. The effects of farnesol on hepatic and renal enzymes were reversed during the recovery period. At the end of the dosing period, increases in absolute and relative liver and kidney weights were seen in farnesol-treated rats. These increases may be secondary to induction of drug metabolizing enzymes, since organ weight increases were not associated with histopathologic alterations and were reversed upon discontinuation of farnesol exposure. Administration of farnesol at doses of up to 1000 mg/kg/day induced reversible increases in the activities of several hepatic and renal drug metabolizing enzymes in rats, while inducing only minimal toxicity. It is concluded that non-toxic or minimally toxic doses of farnesol could alter the metabolism, efficacy, and/or toxicity of drugs with which it is co-administered.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Farnesol/pharmacology , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Animals , Farnesol/blood , Farnesol/toxicity , Female , Glucuronosyltransferase/metabolism , Glutathione Reductase/metabolism , Glutathione Transferase/metabolism , Male , NAD(P)H Dehydrogenase (Quinone)/metabolism , Organ Size/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley
13.
Biochem Pharmacol ; 64(3): 393-404, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12147290

ABSTRACT

Indole-3-carbinol (I3C), found in cruciferous vegetables, has been shown to suppress tumorigenesis at estrogen-responsive sites. This effect may be mediated through modification by I3C of the cytochrome P450 (CYP) complement and activities leading to estrogen detoxication. In this study, we examined the effects of 4- and 10-day treatments of female Sprague-Dawley rats with I3C at 5, 25, and 250 mg/kg body weight, administered by oral gavage, on CYP mRNA expression in the liver and mammary gland, CYP-dependent activities, and the metabolism of 17beta-estradiol (E2) and estrone (E1) by liver microsomes. The mRNA transcripts for hepatic CYP1A1, 1B1, and 2B1/2 and mammary CYP1A1 were up-regulated after treatment with I3C at 250 mg/kg. However, the level of expression of CYP1B1 in the liver was lower than that of other CYPs. In the mammary gland, CYP1B1 mRNA levels were unaltered by treatment and similar to those of I3C-induced CYP1A1. Hepatic P450 probe activities indicative of induction of CYP1A1, 1A2, and 2B1/2 were increased by I3C in a dose-dependent manner. Treatment with I3C at 250 mg/kg increased the capacity of liver microsomes to metabolize E2 to 2-OH-E2, 2-OH-E1, 6alpha-OH-E2, 6beta-OH-E2, estriol, and 15alpha-OH-E2, and E1 to 2-OH-E1, 2-OH-E2, 6(alpha+beta)-OH-E1, and 6alpha-OH-E2. The magnitudes of increases of CYP-dependent activities and rates of estrogen metabolite formation achieved with I3C at 250 mg/kg were smaller after ten than four treatments. The increased rates of formation of 6alpha-OH-E2, 6beta-OH-E2, and 15alpha-OH-E2 from E2 were also detected after treatment with I3C at 25mg/kg, and, except for increased 6beta-OH-E2 from E2, no other changes in E2 or E1 metabolism occurred after treatment with I3C at 5mg/kg. The data indicate that alterations in the CYP complement and, thus, metabolite composition from E2 and E1 are I3C dose- and treatment duration-dependent, and suggest that potential biological activity of I3C administered at low doses to rats may not involve changes in estrogen metabolism.


Subject(s)
Breast/drug effects , Cytochrome P-450 Enzyme System/metabolism , Estrogen Antagonists/pharmacology , Estrogens/metabolism , Indoles/pharmacology , Liver/drug effects , Animals , Breast/enzymology , Cytochrome P-450 Enzyme System/genetics , Estradiol/metabolism , Estrone/metabolism , Female , Gene Expression/drug effects , Liver/enzymology , Models, Animal , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...