27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

916 RESEARCH PLAN TO FILL DATA GAPS IN THE MODE<br />

OF ACTION FOR CANCER RISK ASSESSMENT OF<br />

HEXAVALENT CHROMIUM IN DRINKING WATER.<br />

L. Haws 3 , D. Proctor 1 , C. Thompson 2 and M. Harris 2 . 1 ToxStrategies, Inc.,<br />

Rancho Santa Margarita, CA, 2 ToxStrategies, Inc., Houston, TX and 3 ToxStrategies,<br />

Inc., Austin, TX.<br />

Using the mode-<strong>of</strong>-action (MOA) frameworks proposed by the U.S. EPA and<br />

ILSI/IPCS, we outlined key events and their temporal sequencing in a proposed<br />

MOA for intestinal tumors in mice following ingestion <strong>of</strong> Cr(VI). Data gaps identified<br />

during this MOA analysis were used to inform the design <strong>of</strong> a 90-day drinking<br />

water study to better inform the MOA for the observed responses. Sequentially,<br />

the proposed key events are saturation <strong>of</strong> the reductive capacity <strong>of</strong> the upper gastrointestinal<br />

tract, absorption <strong>of</strong> Cr(VI) into the intestinal epithelium, oxidative<br />

stress and inflammation, cell proliferation, DNA modifications (direct and/or epigenetic),<br />

and mutagenesis. Given clear species differences in carcinogenic response,<br />

key data gaps include direct measures <strong>of</strong> species-specific pharmacokinetic determinants<br />

(e.g. reduction and uptake), direct evidence for oxidative stress and inflammation<br />

as contributors to cell proliferation, and the nature <strong>of</strong> DNA modifications.<br />

Additionally, it is not known whether DNA modification and mutation occur at<br />

lower doses and early time points during exposure to Cr(VI) in drinking water. In<br />

order to address these data gaps, rats and mice were exposed to Cr(VI) for 7 or 90<br />

days, followed by evaluation <strong>of</strong> histopathology, toxicogenomics, oxidative and inflammatory<br />

responses, DNA damage, in vivo mutations, target tissue chromium<br />

concentrations, and rates and capacities <strong>of</strong> Cr(VI) reduction in gastric fluid. <strong>The</strong>se<br />

in vivo measures were collected in target tissues <strong>of</strong> both species at doses carcinogenic<br />

to rodents, as well as at concentrations more environmentally relevant to humans.<br />

Comparisons between responsive and unresponsive species and tissues will<br />

be used to further inform the carcinogenic MOA and provide mechanistic and<br />

dosimetric information necessary for human health risk assessment.<br />

917 TOXICOGENOMIC ANALYSIS OF CR(VI) EFFECTS ON<br />

INTESTINAL AND ORAL EPITHELIUM.<br />

T. R. Zacharewski 1 , A. K. Kopec 1 , A. L. Forgacs 1 , S. Kim 1 , S. D. Grimes 2 and<br />

C. D. Hébert 2 . 1 Biochemistry & Molecular Biology and Center for Integrative<br />

<strong>Toxicology</strong>, Michigan State University, East Lansing, MI and 2 Southern Research<br />

Institute, Birmingham, AL.<br />

Chronic administration <strong>of</strong> hexavalent chromium (CrVI)-containing sodium<br />

dichromate dihydrate (SDD) is linked to intestinal and oral cancer in rodents. In<br />

this study, the dose-dependent gene expression effects <strong>of</strong> SDD (0, 0.3, 4, 14, 60,<br />

170 or 520 mg/L) in female B6C3F1 duodenal, jejunal and oral epithelial tissues<br />

were examined after 7 days <strong>of</strong> exposure in drinking water. 4x44 K Agilent two-color<br />

oligonucleotide arrays identified 6453, 4360 and 698 unique gene expression<br />

changes in duodenum, jejunum and oral mucosa, respectively (|fold change|>1.5,<br />

P1(t)>0.999). Robust dose-dependent differential gene expression was identified in<br />

duodenal and jejunal epithelium. Automated dose-response modeling <strong>of</strong> the microarray<br />

data identified 2491 duodenal, 965 jejunal but only 141 oral mucosa<br />

genes displaying a sigmoidal dose-response pr<strong>of</strong>ile with ED50 values within the experimental<br />

dose range. Comparisons between duodenal and jejunal expression pr<strong>of</strong>iles<br />

showed significant overlap with duodenal differential gene expression responses<br />

exhibiting greater efficacy compared to jejunum. <strong>The</strong> median and<br />

interquartile ED50 range overlapped between duodenum and jejunum with greater<br />

than 70% <strong>of</strong> all sigmoidal ED50 values between 10 and 100 mg/L SDD.<br />

Complementary histopathology identified crypt hyperplasia, cytoplasmic vacuolization<br />

in the intestinal epithelium and histiocytic infiltration in the villous lamina<br />

propria, consistent with the differential expression <strong>of</strong> genes associated with cell<br />

cycle, lipid metabolism and immune response in the duodenum and jejunum.<br />

Cancer associated genes were also over-represented within intestinal differential<br />

gene expression, and to a lesser extent in the oral mucosa. Collectively, these results<br />

indicate that intestinal epithelium is highly responsive to CrVI and early gene expression<br />

signatures may mediate neoplastic changes observed with prolonged SDD<br />

exposure.<br />

918 GENETIC TOXICITY OF HEXAVALENT CHROMIUM IN<br />

THE GASTROINTESTINAL TRACT FOLLOWING ORAL<br />

EXPOSURE.<br />

T. J. O’Brien 1 , H. Ding 1 , C. D. Hébert 2 , R. D. May 2 and S. R. Patierno 1 .<br />

1 Department <strong>of</strong> Pharmacology and Physiology, <strong>The</strong> George Washington University<br />

Medical Center, Washington, DC and 2 Southern Research Institute, Birmingham, AL.<br />

Occupational exposure to hexavalent chromium [Cr(VI))]-containing compounds<br />

has been shown to cause lung cancer. Cr(VI)-induced carcinogenesis has been proposed<br />

to involve direct modification <strong>of</strong> DNA by Cr, inflammation and alteration <strong>of</strong><br />

196 SOT 2011 ANNUAL MEETING<br />

cell signaling pathways. Recently Cr(VI) has been shown to induce tumors <strong>of</strong> the<br />

duodenum <strong>of</strong> rodents following drinking water exposure at concentrations exceeding<br />

60 mg/L sodium dichromate dihydrate (SDD). Because some chromium<br />

species interact with DNA, it is thought that DNA damage and mutagenecity play<br />

a role in the mode <strong>of</strong> action (MOA). To investigate the genotoxicity <strong>of</strong> Cr(VI) in<br />

the GI tract <strong>of</strong> rodents, SDD at 0, 0.3, 4, 14, 60, 170, 520 mg/L was administered<br />

ad libitum for 7 or 90 days. Samples were analyzed for 8-Oxo-2’-deoxyguanosine<br />

(8-OHdG) at 90 days (10 animals/group), and Cr-DNA binding at 7 and 90 days<br />

(5 animals/group). For Cr-DNA binding, DNA was isolated from scrapings <strong>of</strong> the<br />

oral, duodenal and jejunal epithelium and analyzed by ICP/MS. 8-OHdG analysis<br />

was performed on whole tissue from the duodenum and oral cavity using an<br />

ELISA. 8-OHdG levels were 2-3 times higher in the duodenal epithelium versus<br />

oral mucosa; however, a concentration-dependent increase in oxidized DNA bases<br />

was not observed. Concentration-dependent increases in Cr-DNA binding were<br />

observed in oral, jejunal and duodenal epithelium at 7 and 90 days, with evidence<br />

<strong>of</strong> the accumulation <strong>of</strong> Cr-binding observed overtime in all tissues. Slightly higher,<br />

but not statistically significant, levels <strong>of</strong> Cr-DNA binding were observed in the jejunum,<br />

as compared to the duodenum. In addition, an apparent threshold seems to<br />

exist for Cr-DNA binding in all tissues examined. Because we also observed villous<br />

cytotoxicity histopathologically, these results suggest that cytotoxicity in the duodenum<br />

villi may contribute to Cr(VI) carcinogenicity through a wound-rehealing<br />

mechanism involving the crypt cells.<br />

919 A FRAMEWORK FOR EVALUATING DATA IN SUPPORT<br />

OF A MUTAGENIC MOA.<br />

R. Schoeny. Office <strong>of</strong> Water, U.S. EPA, Washington, DC. Sponsor: D. Proctor.<br />

To support its 2005 Guidelines for Cancer Risk Assessment the U.S. EPA Risk<br />

Assessment Forum convened a technical panel to develop guidance in evaluating<br />

data for determination that a chemical carcinogen acts through a mutagenic mode<br />

<strong>of</strong> action (MOA). <strong>The</strong> panel proposed definitions for mutagenicity and genotoxicity.<br />

Mutagenicity is the induction <strong>of</strong> permanent, transmissible changes in the<br />

amount, chemical properties, or structure <strong>of</strong> the genetic material. Genotoxicity is a<br />

broader term encompassing any alterations to genetic material including those that<br />

are transient. <strong>The</strong> technical panel noted that there is no “default MOA” for carcinogenicity<br />

but that all such judgements are data intensive exercises. A framework<br />

for data evaluation was proposed. While acknowledging the iterative nature <strong>of</strong> risk<br />

assessment, the framework considers 4 steps: 1. assemble all relevant data; 2. evaluate<br />

the data against current acceptance and quality criteria; 3. judge weight <strong>of</strong> evidence<br />

that the chemical has mutagenic activity, and if so, what type(s) <strong>of</strong> mutational<br />

activity; 4. by applying the Cancer Guidelines MOA framework, assess the<br />

evidence as to whether mutation is an early key event in the induction <strong>of</strong> tumors.<br />

<strong>The</strong> technical panel further proposed a hierarchy <strong>of</strong> data preference in making the<br />

weight <strong>of</strong> evidence decisions. <strong>The</strong> ideal data are cancer-relevant oncogene or tumor<br />

suppressor gene mutations detected in the tumor target tissue following chemical<br />

exposure. However, these results are rarely available, and the risk assessor must rely<br />

on more standard test batteries such as those from OCSPP guideline studies (i.e.,<br />

gene mutations, cytogenetic damage, DNA adducts and/or primary DNA damage).<br />

Several case studies have been published that use principles <strong>of</strong> the proposed<br />

framework. Chlor<strong>of</strong>orm has sufficient evidence that the MOA does not include<br />

mutagenicity; whereas cyclophosphamide has sufficient evidence <strong>of</strong> a mutagenic<br />

MOA (McCarroll et al. 2008); and CrVI has adequate, but not overwhelming evidence<br />

<strong>of</strong> a mutagenic MOA (McCarrroll et al. 2010). [<strong>The</strong> opinions in this abstract<br />

are those <strong>of</strong> the author and do not necessarily reflect policies <strong>of</strong> the U.S. EPA.]<br />

920 PHARMACOKINETICS OF INGESTED CR(VI) IN<br />

RODENTS: EXTRAPOLATIONS TO TARGET TISSUE<br />

DOSE IN HUMANS.<br />

S. Hays 1 , L. Aylward 2 and C. Kirman 3 . 1 Summit <strong>Toxicology</strong>, Inc., Lyons, Co.,<br />

2 Summit <strong>Toxicology</strong>, Inc., Falls Church, VA and 3 Summit <strong>Toxicology</strong>, Inc., Orange<br />

Village, OH.<br />

<strong>The</strong> target tissues for carcinogenic effects following chronic oral exposure to Cr(VI)<br />

in drinking water are the oral cavity in rats, and the small intestines (duodenum<br />

and jejunum) in mice, for which a response gradient is readily apparent (duodenum>jejunum>ileum)<br />

suggesting that neoplastic and non-neoplastic effects are due<br />

to direct contact <strong>of</strong> Cr(VI) in the lumen <strong>of</strong> the small intestine rather than systemic<br />

absorption. <strong>The</strong> ability <strong>of</strong> gastric and other body fluids to reduce Cr(VI) to Cr(III)<br />

before absorption occurs is recognized as a detoxification process providing protection<br />

against Cr(VI) toxicity following ingestion. However, saturation <strong>of</strong> reductive<br />

capacity occurs at some level <strong>of</strong> exposure, and serves as a potential source <strong>of</strong> nonlinear<br />

toxicokinetics for Cr(VI) . A series <strong>of</strong> studies were conducted to develop<br />

PBPK models to estimate species-specific target tissue dose following Cr(VI) ingestion.<br />

<strong>The</strong>se included an ex vivo study <strong>of</strong> Cr(VI) reduction kinetics (rate and capacity)<br />

using real rat, mouse and human gastric fluid, and measures <strong>of</strong> Cr and Fe tissue

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!