Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.230
Filter
1.
Nature ; 632(8027): 1137-1144, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085612

ABSTRACT

Exposure to environmental pollutants and human microbiome composition are important predisposition factors for tumour development1,2. Similar to drug molecules, pollutants are typically metabolized in the body, which can change their carcinogenic potential and affect tissue distribution through altered toxicokinetics3. Although recent studies demonstrated that human-associated microorganisms can chemically convert a wide range of xenobiotics and influence the profile and tissue exposure of resulting metabolites4,5, the effect of microbial biotransformation on chemical-induced tumour development remains unclear. Here we show that the depletion of the gut microbiota affects the toxicokinetics of nitrosamines, which markedly reduces the development and severity of nitrosamine-induced urinary bladder cancer in mice6,7. We causally linked this carcinogen biotransformation to specific gut bacterial isolates in vitro and in vivo using individualized bacterial culture collections and gnotobiotic mouse models, respectively. We tested gut communities from different human donors to demonstrate that microbial carcinogen metabolism varies between individuals and we showed that this metabolic activity applies to structurally related nitrosamine carcinogens. Altogether, these results indicate that gut microbiota carcinogen metabolism may be a contributing factor for chemical-induced carcinogenesis, which could open avenues to target the microbiome for improved predisposition risk assessment and prevention of cancer.


Subject(s)
Carcinogenesis , Carcinogens , Gastrointestinal Microbiome , Nitrosamines , Urinary Bladder Neoplasms , Animals , Female , Humans , Male , Mice , Biotransformation , Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogens/chemistry , Carcinogens/metabolism , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Gastrointestinal Microbiome/physiology , Germ-Free Life , Mice, Inbred C57BL , Nitrosamines/chemistry , Nitrosamines/metabolism , Nitrosamines/pharmacokinetics , Nitrosamines/toxicity , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/etiology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/prevention & control , Disease Susceptibility
2.
Toxicol Appl Pharmacol ; 438: 115830, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34933053

ABSTRACT

Dibenzo[def,p]chrysene (DBC) is an environmental polycyclic aromatic hydrocarbon (PAH) that causes tumors in mice and has been classified as a probable human carcinogen by the International Agency for Research on Cancer. Animal toxicity studies often utilize higher doses than are found in relevant human exposures. Additionally, like many PAHs, DBC requires metabolic bioactivation to form the ultimate toxicant, and species differences in DBC and DBC metabolite metabolism have been observed. To understand the implications of dose and species differences, a physiologically based pharmacokinetic model (PBPK) for DBC and major metabolites was developed in mice and humans. Metabolism parameters used in the model were obtained from experimental in vitro metabolism assays using mice and human hepatic microsomes. PBPK model simulations were evaluated against mice dosed with 15 mg/kg DBC by oral gavage and human volunteers orally microdosed with 29 ng of DBC. DBC and its primary metabolite DBC-11,12-diol were measured in blood of mice and humans, while in urine, the majority of DBC metabolites were obeserved as conjugated DBC-11,12-diol, conjugated DBC tetrols, and unconjugated DBC tetrols. The PBPK model was able to predict the time course concentrations of DBC, DBC-11,12-diol, and other DBC metabolites in blood and urine of human volunteers and mice with reasonable accuracy. Agreement between model simulations and measured pharmacokinetic data in mice and human studies demonstrate the success and versatility of our model for interspecies extrapolation and applicability for different doses. Furthermore, our simulations show that internal dose metrics used for risk assessment do not necessarily scale allometrically, and that PBPK modeling provides a reliable approach to appropriately account for interspecies differences in metabolism and physiology.


Subject(s)
Chrysenes/administration & dosage , Chrysenes/pharmacokinetics , Cystine/analogs & derivatives , Animals , Carcinogens/administration & dosage , Carcinogens/pharmacokinetics , Cystine/administration & dosage , Cystine/pharmacokinetics , Female , Humans , Male , Mice , Models, Biological , Neoplasms/chemically induced
3.
Regul Toxicol Pharmacol ; 119: 104819, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33189748

ABSTRACT

Studies demonstrate that with sufficient dose and duration, 1,4-dioxane (1,4-DX) induces liver tumors in laboratory rodent models. The available evidence aligns with a threshold-dependent, tumor promotion mode of action (MOA). The MOA and key events (KE) in rats are well developed but less so in the mouse. Therefore, we conducted a 90-day drinking water study in female mice to evaluate early KE at 7, 28, and 90 days. Female B6D2F1/Crl mice consumed drinking water containing 0, 40, 200, 600, 2000 or 6000 ppm 1,4-DX. 1,4-DX was detected in blood at 90-days of exposure to 6000 ppm, but not in the other exposure groups, indicating a metabolic clearance threshold between 2000 and 6000. Early events identified in this study include glycogen-like vacuolization, centrilobular hypertrophy, centrilobular GST-P staining, apoptosis, and pan-lobular increase in cell proliferation observed after 90-days of exposure to 6000 ppm 1,4-DX. There was minimal evidence of hepatotoxicity over the duration of this study. These findings demonstrate a previously unreported direct mitogenic response following exposures exceeding the metabolic clearance threshold of 1,4-DX. Collectively, the information generated in this study supports a threshold MOA for the development of liver tumors in mice after exposure to 1,4-DX.


Subject(s)
Carcinogens/toxicity , Dioxanes/toxicity , Liver Neoplasms/chemically induced , Animals , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Carcinogens/pharmacokinetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Dioxanes/blood , Dioxanes/pharmacokinetics , Dose-Response Relationship, Drug , Drinking Water , Female , Liver/drug effects , Liver/pathology , Liver Neoplasms/pathology , Mice , Toxicity Tests, Subchronic
4.
Regul Toxicol Pharmacol ; 110: 104539, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31765675

ABSTRACT

Arsenic, a metalloid and naturally occurring element, is one of the most abundant elements in the earth's crust. Water is contaminated by arsenic through natural sources (underground water, minerals and geothermal processes) and anthropogenic sources such as mining, industrial processes, and the production and use of pesticides. Humans are exposed to arsenic mainly by drinking contaminated water, and secondarily through inhalation and skin contact. Arsenic exposure is associated with the development of vascular disease, including stroke, ischemic heart disease and peripheral vascular disease. Also, arsenic increases the risk of tumors of bladder, lungs, kidneys and liver, according to the International Agency for Research on Cancer and the Food and Drug Administration. Once ingested, an estimated 70-90% of inorganic arsenic is absorbed by the gastrointestinal tract and widely distributed through the blood to different organs, primarily to the liver, kidneys, lungs and bladder and secondarily to muscle and nerve tissue. Arsenic accumulates in the organs, especially in the liver. Its excretion mostly takes place through urination. The toxicokinetics of arsenic depends on the duration of exposure, pathway of ingestion, physicochemical characteristics of the compound, and affected biological species. The present review outlines of arsenic toxic effects focusing on different cancer types whit highest prevalence's by exposure to this metalloid and signaling pathways of carcinogenesis.


Subject(s)
Arsenic/toxicity , Carcinogens/toxicity , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Neoplasms/chemically induced , Animals , Arsenic/pharmacokinetics , Carcinogens/pharmacokinetics , Environmental Pollutants/pharmacokinetics , Environmental Pollution , Humans , Neoplasms/genetics , Toxicokinetics
5.
Regul Toxicol Pharmacol ; 110: 104548, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31805361

ABSTRACT

Areca Nut (AN), the seed of tropical palm tree Areca catechu, is a widely chewed natural product with estimated 600 million users across the world. Various AN products, thriving in the market, portray 'Areca nut' or 'Supari' as mouth freshener and safe alternative to smokeless tobacco. Unfortunately, AN is identified as a Group 1 human carcinogen by International Agency for Research on Cancer (IARC). Wide variation in the level of alkaloids, broadly ranging from 2 to 10 mg/gm dry weight, is observed in diverse variety of AN sold worldwide. For the first time, various factors influencing the formation of carcinogenic alkaloids in AN at various stages, including during the growth, processing, and storage of the nut, are discussed. Current review illustrates the mechanism of cancer induction by areca alkaloids in humans and also compiles dose-dependent pharmacology and toxicology data of arecoline, the most potent carcinogenic alkaloid in AN. Careful monitoring of the arecoline content in AN can potentially be used as a tool in product surveillance studies to identify the variations in characteristics of various AN sample sold worldwide. The article will help to generate public awareness and sensitize the government bodies to initiate campaigns against AN use and addiction.


Subject(s)
Alkaloids , Areca , Carcinogens , Neoplasms/chemically induced , Nuts , Alkaloids/pharmacokinetics , Alkaloids/pharmacology , Alkaloids/toxicity , Animals , Areca/chemistry , Carcinogens/pharmacokinetics , Carcinogens/pharmacology , Carcinogens/toxicity , Dose-Response Relationship, Drug , Humans , Neoplasms/metabolism , Nuts/chemistry
6.
Environ Geochem Health ; 42(5): 1291-1303, 2020 May.
Article in English | MEDLINE | ID: mdl-31515640

ABSTRACT

This study focused on the oral bioaccessibility and children health risks of metal(loid)s (As, Cd, Cr, Cu, Ni, Pb and Zn) in soil/indoor dust of school and households from Lanzhou, China. The simple bioaccessibility extraction test method was applied to assess bioaccessibility, and children's health risk was assessed via statistical modeling (hazard quotients, hazard index and incremental lifetime carcinogenic risk). Metal(loid) content and bioaccessibility in indoor dust samples were significantly higher than those in corresponding soil samples (p < 0.05). The order for mean values of bioaccessibility of the elements in soil was as follows: Cd (57.1%) > Zn (44.6%) > Pb (39.9%) > Cu (33.2%) > Ni (12.4%) > Cr (5.3%) > As (4.4%), while for indoor dust, the order was: As (73.0%) > Cd (68.4%) > Pb (63.3%) > Zn (60.4%) > Cu (36.5%) > Ni (25.2%) > Cr (13.6%). The Pearson correlation coefficient showed that metal(loid) bioaccessibility was in general significantly negatively correlated to the Al, Fe and Mn contents. Neither noncarcinogenic nor carcinogenic risks exceeded the tolerance interval for 3-5- and 6-9-year-old children for all elements. They both were mostly attributed to As considering metal(loid)s types and to school indoor dust considering sources. Therefore, maintaining interior sanitation would be an effective measure to reduce the potential health effects of indoor dust on children.


Subject(s)
Metalloids/pharmacokinetics , Metalloids/toxicity , Metals/pharmacokinetics , Metals/toxicity , Risk Assessment/methods , Air Pollution, Indoor/adverse effects , Air Pollution, Indoor/analysis , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Child , Child, Preschool , China , Dust/analysis , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Environmental Monitoring/methods , Humans , Metalloids/analysis , Metals/analysis , Metals, Heavy/analysis , Rural Population , Schools , Soil Pollutants/analysis , Soil Pollutants/pharmacokinetics , Soil Pollutants/toxicity , Urban Population
7.
Toxicol Appl Pharmacol ; 364: 97-105, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30582946

ABSTRACT

Benzo[a]pyrene (BaP), is a known human carcinogen (International Agency for Research on Cancer (IARC) class 1). The remarkable sensitivity (zepto-attomole 14C in biological samples) of accelerator mass spectrometry (AMS) makes possible, with de minimus risk, pharmacokinetic (PK) analysis following [14C]-BaP micro-dosing of humans. A 46 ng (5 nCi) dose was given thrice to 5 volunteers with minimum 2 weeks between dosing and plasma collected over 72 h. [14C]-BaPeq PK analysis gave plasma Tmax and Cmax values of 1.25 h and 29-82 fg/mL, respectively. PK parameters were assessed by non- compartment and compartment models. Intervals between dosing ranged from 20 to 420 days and had little impact on intra-individual variation. DNA, extracted from peripheral blood mononuclear cells (PBMCs) of 4 volunteers, showed measurable levels (LOD ~ 0.5 adducts/1011 nucleotides) in two individuals 2-3 h post-dose, approximately three orders of magnitude lower than smokers or occupationally-exposed individuals. Little or no DNA binding was detectable at 48-72 h. In volunteers the allelic variants CYP1B1*1/*⁎1, *1/*3 or *3/*3 and GSTM1*0/0 or *1 had no impact on [14C]-BaPeq PK or DNA adduction with this very limited sample. Plasma metabolites over 72 h from two individuals (one CYP1B1*1/*1 and one CYP1B1*3/*3) were analyzed by UPLC-AMS. In both individuals, parent [14C]-BaP was a minor constituent even at the earliest time points and metabolite profiles markedly distinct. AMS, coupled with UPLC, could be used in humans to enhance the accuracy of pharmacokinetics, toxicokinetics and risk assessment of environmental carcinogens.


Subject(s)
Benzo(a)pyrene/pharmacokinetics , Carcinogens/pharmacokinetics , Chromatography, Liquid/methods , Mass Spectrometry , Administration, Oral , Adult , Aged , Benzo(a)pyrene/administration & dosage , Benzo(a)pyrene/adverse effects , Carcinogens/administration & dosage , Carcinogens/toxicity , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , DNA Adducts/metabolism , Female , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Male , Middle Aged , Models, Biological , Pharmacogenomic Variants , Risk Assessment , Young Adult
8.
Toxicol Pathol ; 47(1): 82-92, 2019 01.
Article in English | MEDLINE | ID: mdl-30585133

ABSTRACT

The 6-month Tg.rasH2 mouse carcinogenicity model provides an acceptable alternative to the 2-year carcinogenicity study in CD-1 mice. However, key questions related to the use of this model for testing antisense oligonucleotides (ASOs) include the similarity in the biologic response between mouse strains and the feasibility of using data from the CD-1 mouse to set doses and dose schedules for a Tg.rasH2 carcinogenicity study. To evaluate the potential strain differences, four distinct 2'- O-(2-methoxyethyl) ASOs were administered to CByB6F1 (wild type), Tg.rasH2 (hemizygous), and CD-1 mice. There were no meaningful differences in clinical signs, body weight, food consumption, or serum chemistry and hematology parameters. Histopathology evaluation indicated little to no difference in the spectrum or magnitude of changes present. The cytokine/chemokine response was also not appreciably different between the strains. This was consistent with the similarity in ASO concentration in the liver between the mouse strains tested. As the class effects of the ASOs were not meaningfully different between CD-1, CByB6F1, or Tg.rasH2 mice, data from nonclinical studies in CD-1 mice can be used for dose selection and expectation of effect in the Tg.rasH2 mouse.


Subject(s)
Carcinogens/toxicity , Genes, ras , Oligonucleotides, Antisense/toxicity , Oligoribonucleotides/toxicity , Toxicity Tests , Animals , Base Sequence , Carcinogens/classification , Carcinogens/pharmacokinetics , Cytokines/blood , Female , Hemizygote , Male , Mice, Inbred ICR , Mice, Transgenic , Oligonucleotides, Antisense/classification , Oligonucleotides, Antisense/pharmacokinetics , Oligoribonucleotides/classification , Oligoribonucleotides/pharmacokinetics , Organ Size/drug effects , Organ Specificity , Species Specificity , Time Factors , Tissue Distribution , Toxicity Tests/methods , Toxicity Tests/standards
9.
Part Fibre Toxicol ; 16(1): 34, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477126

ABSTRACT

BACKGROUND: Potassium octatitanate fibers (K2O•8TiO2, POT fibers) are used as an asbestos substitute. Their physical characteristics suggest that respirable POT fibers are likely to be carcinogenic in the lung and pleura. However, previous 2-year inhalation studies reported that respired POT fibers had little or no carcinogenic potential. In the present study ten-week old male F344 rats were left untreated or were administered vehicle, 0.25 or 0.5 mg rutile-type nano TiO2 (r-nTiO2), 0.25 or 0.5 mg POT fibers, or 0.5 mg MWCNT-7 by intra-tracheal intra-pulmonary spraying (TIPS), and then observed for 2 years. RESULTS: There were no differences between the r-nTiO2 and control groups. The incidence of bronchiolo-alveolar cell hyperplasia was significantly increased in the groups treated with 0.50 mg POT and 0.50 mg MWCNT-7. The overall incidence of lung tumors, however, was not increased in either the POT or MWCNT-7 treated groups. Notably, the carcinomas that developed in the POT and MWCNT-7 treated rats were accompanied by proliferative fibrous connective tissue while the carcinomas that developed in the untreated rats and the r-nTiO2 treated rats were not (carcinomas did not develop in the vehicle control rats). In addition, the carcinoma that developed in the rat treated with 0.25 mg POT was a squamous cell carcinoma, a tumor that develops spontaneously in about 1 per 1700 rats. The incidence of mesothelial cell hyperplasia was 4/17, 7/16, and 10/14 and the incidence of malignant mesothelioma was 3/17, 1/16, and 2/14 in the 0.25 mg POT, 0.5 mg POT, and MWCNT-7 treated groups, respectively. Neither mesothelial cell hyperplasia nor mesothelioma developed in control rats or the rats treated with r-nTiO2. Since the incidence of spontaneously occurring malignant mesothelioma in rats is extremely low, approximately 1 per 1000 animals (Japan Bioassay Research Center [JBRC] historical control data), the development of multiple malignant mesotheliomas in the POT and MWCNT-7 treated groups was biologically significant. CONCLUSION: The incidence of pleural mesotheliomas in male F344 rats administered POT fibers and MWCNT-7 was significantly higher than the JBRC historical control data, indicating that the incidence of pleural mesothelioma in the groups administered POT fibers and MWCNT-7 fibers via the airway using TIPS was biologically significant. The incidence of type II epithelial cell hyperplasia and the histology of the carcinomas that developed in the POT treated rats also indicates that respirable POT fibers are highly likely to be carcinogenic in the lungs of male F344 rats.


Subject(s)
Carcinogens/toxicity , Lung Neoplasms/chemically induced , Lung/drug effects , Mesothelioma/chemically induced , Pleura/drug effects , Titanium/toxicity , Animals , Carcinogens/chemistry , Carcinogens/pharmacokinetics , Inhalation Exposure , Lung/pathology , Lung Neoplasms/pathology , Male , Mesothelioma/pathology , Mesothelioma, Malignant , Mineral Fibers , Pleura/pathology , Rats, Inbred F344 , Surface Properties , Tissue Distribution , Titanium/chemistry , Titanium/pharmacokinetics
10.
Regul Toxicol Pharmacol ; 95: 29-51, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29510166

ABSTRACT

Afidopyropen is a novel insecticide that acts as a TRPV channel modulator in chordotonal organs of target insects. In two carcinogenicity studies with Fischer rats, an increased incidence of uterine adenocarcinomas was observed at 1000 and 3000 ppm. This finding prompted an investigation of the mechanism of the tumor formation as well as the relevance of this mechanism to humans. The mechanistic work took parallel paths: one path investigated the pharmacokinetic properties of the test substance at the doses where the tumors were found; while the second path examined the key mechanistic events that culminated in uterine adenocarcinomas. The results of the investigation indicated that the tumors only occurred at doses where excretion of test substance was saturated - indicating that homeostatic biological and/or physiological processes were overwhelmed. At the doses where these processes were overwhelmed, the test substance acted through a mechanism of dopamine agonism, triggering a cascade key events that resulted in uterine adenocarcinomas. An analysis of these mechanisms observed in rat showed that they are both quantitatively (pharmacokinetic mechanism) and qualitatively (dopamine agonism mechanism) not relevant to humans. Therefore the uterine adenocarcinomas observed in the rat associated with high doses of Afidopyropen are not expected to pose a carcinogenic risk to humans.


Subject(s)
Adenocarcinoma/chemically induced , Carcinogens/toxicity , Dopamine Agonists/toxicity , Heterocyclic Compounds, 4 or More Rings/toxicity , Insecticides/toxicity , Lactones/toxicity , Uterine Neoplasms/chemically induced , Animals , Carcinogens/pharmacokinetics , Disease Progression , Dopamine Agonists/pharmacokinetics , Female , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Humans , Insecticides/pharmacokinetics , Lactones/pharmacokinetics , Male , Rats, Inbred F344 , Risk Assessment , Toxicity Tests
11.
Regul Toxicol Pharmacol ; 95: 17-28, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29496518

ABSTRACT

Based on 13 chronic studies, styrene exposure causes lung tumors in mice, but no tumor increases in other organs in mice or rats. Extensive research into the mode of action demonstrates the key events and human relevance. Key events are: metabolism of styrene by CYP2F2 in mouse lung club cells to ring-oxidized metabolites; changes in gene expression for metabolism of lipids and lipoproteins, cell cycle and mitotic M-M/G1 phases; cytotoxicity and mitogenesis in club cells; and progression to preneoplastic/neoplastic lesions in lung. Although styrene-7,8-oxide (SO) is a common genotoxic styrene metabolite in in vitro studies, the data clearly demonstrate that SO is not the proximate toxicant and that styrene does not induce a genotoxic mode of action. Based on complete attenuation of styrene short-term and chronic toxicity in CYP2F2 knockout mice and similar attenuation in CYP2F1 (humanized) transgenic mice, limited metabolism of styrene in human lung by CYP2F1, 2 + orders of magnitude lower SO levels in human lung compared to mouse lung, and lack of styrene-related increase in lung cancer in humans, styrene does not present a risk of cancer to humans.


Subject(s)
Carcinogens/toxicity , Lung Neoplasms/chemically induced , Styrene/toxicity , Animals , Carcinogens/pharmacokinetics , Cell Survival/drug effects , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , Humans , Lipid Metabolism/genetics , Lung/drug effects , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice, Knockout , Rats , Risk Assessment , Species Specificity , Styrene/pharmacokinetics
12.
Chem Res Toxicol ; 30(1): 81-93, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-28092947

ABSTRACT

Pyrrolizidine alkaloids (PAs) and PA N-oxides are a class of phytochemical carcinogens contained in over 6000 plant species spread around the world. It has been estimated that approximately half of the 660 PAs and PA N-oxides that have been characterized are cytotoxic, genotoxic, and tumorigenic. It was recently determined that a genotoxic mechanism of liver tumor initiation mediated by PA-derived DNA adducts is a common metabolic activation pathway of a number of PAs. We proposed this set of PA-derived DNA adducts could be a common biological biomarker of PA exposure and a potential biomarker of PA-induced liver tumor formation. We have also found that several reactive secondary pyrrolic metabolites can dissociate and interconvert to other secondary pyrrolic metabolites, resulting in the formation of the same exogenous DNA adducts. This present perspective reports the current progress on these new findings and proposes future research needed for obtaining a greater understanding of the role of this activation pathway and validating the use of this set of PA-derived DNA adducts as a biological biomarker of PA-induced liver tumor initiation.


Subject(s)
Carcinogens/pharmacokinetics , Carcinogens/toxicity , Liver Neoplasms/chemically induced , Pyrrolizidine Alkaloids/pharmacokinetics , Pyrrolizidine Alkaloids/toxicity , Activation, Metabolic , Animals , Humans , Liver Neoplasms/metabolism
13.
Chem Res Toxicol ; 30(1): 126-144, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28092946

ABSTRACT

Worldwide, cancers of the oral cavity and pharynx comprise the sixth most common malignancies. Histologically, more than 90% of oral cancers are squamous cell carcinoma (SCC). Epidemiologic data strongly support the role of exogenous factors such as tobacco, alcohol, and human papilloma virus infection as major causative agents. Avoidance of risk factors has only been partially successful, and survival rates have not improved despite advances in therapeutic approaches. Therefore, new or improved approaches to prevention and/or early detection are critical. Better understanding of the mechanisms of oral carcinogenesis can assist in the development of novel biomarkers for early detection and strategies for disease prevention. Toward this goal, several animal models for carcinogenesis in the oral cavity have been developed. Among these are xenograft, and transgenic animal models, and others employing the synthetic carcinogens such as 7,12-dimethylbenz[a]anthracene in hamster cheek pouch and 4-nitroquinoline-N-oxide in rats and mice. Additional animal models employing environmental carcinogens such as benzo[a]pyrene and N'-nitrosonornicotine have been reported. Each model has certain advantages and disadvantages. Models that (1) utilize environmental carcinogens, (2) reflect tumor heterogeneity, and (3) accurately represent the cellular and molecular changes involved in the initiation and progression of oral cancer in humans could provide a realistic platform. To achieve this goal, we introduced a novel nonsurgical mouse model to study oral carcinogenesis induced by dibenzo[a,l]pyrene (DB[a,l]P), an environmental pollutant and tobacco smoke constituent, and its diol epoxide metabolite (±)-anti-11,12-dihydroxy-13,14-epoxy-11,12,13,14-tetrahydrodibenzo[a,l]pyrene [(±)-anti-DB[a,l]PDE]. On the basis of a detailed comparison of oral cancer induced by DB[a,l]P with that induced by the other above-mentioned oral carcinogens with respect to dose, duration, species and strain, cellular and molecular targets, and relative carcinogenic potency, our animal model may offer a more realistic platform to study oral carcinogenesis. In this perspective, we also discuss our preclinical studies to demonstrate the potential of black raspberry extracts on the prevention of OSCC. Specifically, we were the first to demonstrate that black raspberry inhibited DB[a,l]P-DNA binding and of particular importance its capacity to enhance the repair of DB[a,l]P-induced bulky lesions in DNA. We believe that the information presented in this perspective will stimulate further research on the impact of environmental carcinogens in the development of oral cancer and may lead to novel strategies toward the control and prevention of this disease.


Subject(s)
Carcinogens/toxicity , Mouth Neoplasms/prevention & control , Plant Extracts/pharmacology , Rubus , Activation, Metabolic , Animals , Carcinogenesis , Carcinogens/pharmacokinetics , DNA Adducts , DNA Repair , Disease Models, Animal , Humans , Mouth Neoplasms/etiology , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mutation , Tumor Suppressor Protein p53/genetics
14.
Arch Toxicol ; 91(9): 3185-3188, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28536864

ABSTRACT

The rabbit was the initial animal model to investigate the acetylation polymorphism expressed in humans. Use of the rabbit model is compromised by lack of a rapid non-invasive method for determining acetylator phenotype. Slow acetylator phenotype in the rabbit results from deletion of the N-acetyltransferase 2 (NAT2) gene. A relatively quick and non-invasive method for identifying the gene deletion was developed and acetylator phenotypes confirmed by measurement of N- and O-acetyltransferase activities in hepatic cytosols. Rabbit liver cytosols catalyzed the N-acetylation of sulfamethazine (p = 0.0014), benzidine (p = 0.0257), 4-aminobiphenyl (p = 0.0012), and the O-acetylation of N-hydroxy-2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (N-OH-PhIP; p = 0.002) at rates significantly higher in rabbits possessing NAT2 gene than rabbits with NAT2 gene deleted. In contrast, hepatic cytosols catalyzed the N-acetylation of p-aminobenzoic acid (an N-acetyltransferase 1 selective substrate) at rates that did not differ significantly (p > 0.05) between rabbits positive and negative for NAT2. The new NAT2 genotyping method facilitates use of the rabbit model to investigate the role of acetylator polymorphism in the metabolism of aromatic and heterocyclic amine drugs and carcinogens.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Carcinogens/pharmacokinetics , Genotyping Techniques/methods , Polymorphism, Genetic , Acetylation , Aminobiphenyl Compounds/pharmacokinetics , Animals , Arylamine N-Acetyltransferase/metabolism , Benzidines/pharmacokinetics , Cytosol/enzymology , Genotype , Rabbits , Sulfamethazine/pharmacokinetics
15.
Arch Toxicol ; 91(10): 3317-3325, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28283705

ABSTRACT

3-Nitrobenzanthrone (3-NBA), a potent environmental mutagen and carcinogen, is known to be activated in vivo to 3-benzanthronylnitrenium ion which forms both NH and C2-bound adducts with DNA and also reacts with glutathione giving rise to urinary 3-aminobenzanthron-2-ylmercapturic acid. In this study, acid hydrolysate of globin from rats dosed intraperitoneally with 3-NBA was analysed by HPLC/MS to identify a novel type of cysteine adduct, 3-aminobenzanthron-2-ylcysteine (3-ABA-Cys), confirmed using a synthesised standard. The 3-ABA-Cys levels in globin peaked after single 3-NBA doses of 1 and 2 mg/kg on day 2 to attain 0.25 and 0.49 nmol/g globin, respectively, thereafter declining slowly to 70-80% of their maximum values during 15 days. After dosing rats for three consecutive days with 1 mg 3-NBA/kg a significant cumulation of 3-ABA-Cys in globin was observed. 3-ABA-Cys was also found in the plasma hydrolysate. Herein, after dosing with 1 and 2 mg 3-NBA/kg the adduct levels peaked on day 1 at 0.15 and 0.51 nmol/ml plasma, respectively, thereafter declining rapidly to undetectable levels on day 15. In addition, sulphinamide adducts were also found in the exposed rats, measured indirectly as 3-aminobenzanthrone (3-ABA) split off from globin by mild acid hydrolysis. Levels of both types of adducts in the globin samples parallelled very well with 3-ABA/3-ABA-Cys ratio being around 1:8. In conclusion, 3-ABA-Cys is the first example of arylnitrenium-cysteine adduct in globin representing a new promising class of biomarkers to assess cumulative exposures to aromatic amines, nitroaromatics and heteroaromatic amines.


Subject(s)
Benz(a)Anthracenes/pharmacokinetics , Carcinogens/pharmacokinetics , Globins/chemistry , Animals , Benz(a)Anthracenes/metabolism , Carcinogens/metabolism , Chromatography, High Pressure Liquid , Cysteine/chemistry , Cysteine/metabolism , Environmental Biomarkers , Hydrolysis , Magnetic Resonance Spectroscopy , Male , Plasma/metabolism , Rats, Wistar
16.
Arch Toxicol ; 91(9): 3093-3108, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28357488

ABSTRACT

Considering the rapid developments in food safety in the past decade in China, it is of importance to obtain insight into what extent safety and risk assessments of chemicals performed for the Caucasian population apply to the Chinese population. The aim of the present study was to determine physiologically based kinetic (PBK) modeling-based predictions for differences between Chinese and Caucasians in terms of metabolic bioactivation and detoxification of the food-borne genotoxic carcinogen estragole. The PBK models were defined based on kinetic constants for hepatic metabolism derived from in vitro incubations using liver fractions of the two ethnic groups, and used to evaluate the inter-ethnic differences in metabolic activation and detoxification of estragole. The models predicted that at realistic dietary intake levels, only 0.02% of the dose was converted to the ultimate carcinogenic metabolite 1'-sulfooxyestragole in Chinese subjects, whereas this amounted to 0.09% of the dose in Caucasian subjects. Detoxification of 1'-hydroxyestragole, mainly via conversion to 1'-oxoestragole, was similar within the two ethnic groups. The 4.5-fold variation in formation of the ultimate carcinogenic metabolite of estragole accompanied by similar rates of detoxification may indicate a lower risk of estragole for the Chinese population at similar levels of exposure. The study provides a proof of principle for how PBK modeling can identify differences in ethnic sensitivity and provide a more refined risk assessment for a specific ethnic group for a compound of concern.


Subject(s)
Anisoles/pharmacokinetics , Models, Biological , Administration, Oral , Allylbenzene Derivatives , Anisoles/administration & dosage , Arylsulfotransferase/metabolism , Asian People , Carcinogens/administration & dosage , Carcinogens/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Humans , Inactivation, Metabolic , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , White People
17.
Arch Toxicol ; 91(7): 2515-2538, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28175954

ABSTRACT

The detoxification of toxic substances is of general relevance in all biological systems. The plethora of exogenous xenobiotic compounds and endogenous toxic metabolic products explains the evolutionary pressure of all organisms to develop molecular mechanisms to detoxify and excrete harmful substances from the body. P-glycoprotein and other members of the ATP-binding cassette (ABC) transporter family extrude innumerous chemical compounds out of cells. Their specific expression in diverse biological contexts cause different phenotypes: (1) multidrug resistance (MDR) and thus failure of cancer chemotherapy, (2) avoidance of accumulation of carcinogens and prevention of carcinogenesis in healthy tissues, (3) absorption, distribution, metabolization and excretion (ADME) of pharmacological drugs in human patients, (4) protection from environmental toxins in aquatic organisms (multi-xenobiotic resistance, MXR). Hence ABC-transporters may have opposing effects for organismic health reaching from harmful in MDR of tumors to beneficial for maintenance of health in MXR. While their inhibition by specific inhibitors may improve treatment success in oncology and avoid carcinogenesis, blocking of ABC-transporter-driven efflux by environmental pollutants leads to ecotoxicological consequences in marine biotopes. Poisoned seafood may enter the food-chain and cause intoxications in human beings. As exemplified with ABC-transporters, joining forces in interdisciplinary research may, therefore, be a wise strategy to fight problems in human medicine and environmental sciences.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Carcinogens/toxicity , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Xenobiotics/toxicity , ATP-Binding Cassette Transporters/metabolism , Carcinogens/pharmacokinetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Ecotoxicology/methods , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Humans , Inactivation, Metabolic , Xenobiotics/pharmacokinetics
18.
Arch Toxicol ; 91(2): 713-734, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27334372

ABSTRACT

The present study describes physiologically based kinetic (PBK) models for the alkenylbenzene myristicin that were developed by extension of the PBK models for the structurally related alkenylbenzene safrole in rat and human. The newly developed myristicin models revealed that the formation of the proximate carcinogenic metabolite 1'-hydroxymyristicin in liver is at most 1.8 fold higher in rat than in human and limited for the ultimate carcinogenic metabolite 1'-sulfoxymyristicin to (2.8-4.0)-fold higher in human. In addition, a comparison was made between the relative importance of bioactivation for myristicin and safrole. Model predictions indicate that for these related compounds, the formation of the 1'-sulfoxy metabolites in rat and human liver is comparable with a difference of <2.2-fold over a wide dose range. The results from this PBK analysis support that risk assessment of myristicin may be based on the BMDL10 derived for safrole of 1.9-5.1 mg/kg bw per day. Using an estimated daily intake of myristicin of 0.0019 mg/kg bw per day resulting from the use of herbs and spices, this results in MOE values for myristicin that amount to 1000-2700, indicating a priority for risk management. The results obtained illustrate that PBK modeling provides insight into possible species differences in the metabolic activation of myristicin. Moreover, they provide an example of how PBK modeling can facilitate a read-across in risk assessment from a compound for which in vivo toxicity studies are available to a related compound for which tumor data are not reported, thus contributing to alternatives in animal testing.


Subject(s)
Benzyl Compounds/pharmacokinetics , Dioxolanes/pharmacokinetics , Models, Theoretical , Pyrogallol/analogs & derivatives , Activation, Metabolic , Allylbenzene Derivatives , Animals , Carcinogens/pharmacokinetics , Humans , Inactivation, Metabolic , Kinetics , Liver/drug effects , Liver/metabolism , Male , Microsomes/drug effects , Microsomes/metabolism , Oxidation-Reduction , Pyrogallol/pharmacokinetics , Rats, Sprague-Dawley , Risk Assessment/methods , Safrole/pharmacokinetics
19.
J Appl Toxicol ; 37(5): 621-640, 2017 05.
Article in English | MEDLINE | ID: mdl-27885692

ABSTRACT

In cancer bioassays, inhalation, but not drinking water exposure to ethyl tertiary-butyl ether (ETBE), caused liver tumors in male rats, while tertiary-butyl alcohol (TBA), an ETBE metabolite, caused kidney tumors in male rats following exposure via drinking water. To understand the contribution of ETBE and TBA kinetics under varying exposure scenarios to these tumor responses, a physiologically based pharmacokinetic model was developed based on a previously published model for methyl tertiary-butyl ether, a structurally similar chemical, and verified against the literature and study report data. The model included ETBE and TBA binding to the male rat-specific protein α2u-globulin, which plays a role in the ETBE and TBA kidney response observed in male rats. Metabolism of ETBE and TBA was described as a single, saturable pathway in the liver. The model predicted similar kidney AUC0-∞ for TBA for various exposure scenarios from ETBE and TBA cancer bioassays, supporting a male-rat-specific mode of action for TBA-induced kidney tumors. The model also predicted nonlinear kinetics at ETBE inhalation exposure concentrations above ~2000 ppm, based on blood AUC0-∞ for ETBE and TBA. The shift from linear to nonlinear kinetics at exposure concentrations below the concentration associated with liver tumors in rats (5000 ppm) suggests the mode of action for liver tumors operates under nonlinear kinetics following chronic exposure and is not relevant for assessing human risk. Copyright © 2016 The Authors Journal of Applied Toxicology Published by John Wiley & Sons Ltd.


Subject(s)
Alpha-Globulins/metabolism , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Ethyl Ethers/pharmacokinetics , Ethyl Ethers/toxicity , tert-Butyl Alcohol/pharmacokinetics , tert-Butyl Alcohol/toxicity , Administration, Inhalation , Administration, Oral , Animals , Area Under Curve , Computer Simulation , Female , Inhalation Exposure , Kidney/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Metabolic Networks and Pathways , Nonlinear Dynamics , Protein Binding , Rats
20.
Int J Toxicol ; 36(6): 427-439, 2017.
Article in English | MEDLINE | ID: mdl-29153032

ABSTRACT

The carcinogenic potential of roxadustat (FG-4592), a novel orally active, heterocyclic small molecule inhibitor of hypoxia-inducible factor prolyl hydroxylase (HIF-PH) enzymes in clinical development for treatment of anemia, was evaluated in CD-1 mice and Sprague Dawley rats. Inhibition of HIF-PH by roxadustat leads to a rapid increase in cytoplasmic HIF-α concentrations, followed by translocation of HIF-α to the nucleus and upregulation of HIF-responsive genes, including erythropoietin. Roxadustat was dosed by oral gavage 3 times weekly (TIW) for up to 104 weeks in mice at 0, 15, 30, and 60 mg/kg and in rats at 0, 2.5, 5, and 10 mg/kg. Treatment-associated changes in hematology parameters were consistent with the pharmacologic activity of roxadustat and included elevations in hematocrit in mice at 30 and 60 mg/kg TIW and elevations in erythrocyte count, hemoglobin, hematocrit, and red cell distribution width in rats at 10 mg/kg TIW. No increase in mortality or neoplastic effects compared with vehicle controls was observed after roxadustat treatment in either species. No treatment-related nonneoplastic findings were observed in mice, whereas nonneoplastic microscopic findings in rats were limited to atrial/aortic thromboses at 10 mg/kg TIW males and bone marrow hypercellularity in all treated male and female groups, consistent with the pharmacology of roxadustat. In conclusion, roxadustat administered by oral gavage to mice and rats TIW for up to 104 weeks resulted in dose-dependent exposure and hematologic effects with no effect on survival or development of neoplastic lesions at up to 60 mg/kg in mice and up to 10 mg/kg in rats.


Subject(s)
Carcinogens/toxicity , Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Isoquinolines/toxicity , Administration, Oral , Animals , Carcinogenicity Tests , Carcinogens/chemistry , Carcinogens/pharmacokinetics , Dose-Response Relationship, Drug , Female , Glycine/chemistry , Glycine/pharmacokinetics , Glycine/toxicity , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Male , Mice, Inbred Strains , Rats, Sprague-Dawley , Survival Analysis , Toxicokinetics
SELECTION OF CITATIONS
SEARCH DETAIL