26.11.2012 Views

Contemporary ConCepts in toxiCology (CCt) Workshop

Contemporary ConCepts in toxiCology (CCt) Workshop

Contemporary ConCepts in toxiCology (CCt) Workshop

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.

<strong>Contemporary</strong> <strong>ConCepts</strong> <strong>in</strong><br />

<strong>toxiCology</strong> (<strong>CCt</strong>) <strong>Workshop</strong><br />

Hemangiosarcoma <strong>in</strong> Rodents:<br />

Mode-of-Action Evaluation<br />

and Human Relevance<br />

<strong>Workshop</strong><br />

DeCember 4–5, 2008<br />

West<strong>in</strong> arl<strong>in</strong>gton gateWay hotel<br />

arl<strong>in</strong>gton, Virg<strong>in</strong>ia<br />

<strong>Workshop</strong> Co-Chairs:<br />

Samuel M. Cohen<br />

University of Nebraska Medical Center, Omaha, NE,<br />

United States<br />

Jon C. Cook<br />

Pfizer Inc., Groton, CT, United States


notes<br />

2


2008–2009 COUNCIL<br />

PresIdeNt<br />

Kenneth S. Ramos,<br />

B.s.Ph., Ph.d., Ats<br />

University of Louisville<br />

VICe PresIdeNt<br />

Cheryl Lyn Walker,<br />

Ph.d.<br />

University of texas<br />

Md Anderson Cancer Center<br />

VICe PresIdeNt-eLeCt<br />

Michael P. Holsapple,<br />

Ph.d., Ats<br />

International Life sciences<br />

Institute (ILsI)<br />

Health and environmental<br />

sciences Institute (HesI)<br />

treAsUrer<br />

William Slikker, Jr.,<br />

Ph.d., Ats<br />

National Center for<br />

toxicological research<br />

treAsUrer-eLeCt<br />

Lawrence R. Curtis,<br />

Ph.d.<br />

Oregon state University<br />

seCretAry<br />

Mart<strong>in</strong> A. Philbert,<br />

Ph.d., Ats<br />

University of Michigan<br />

PAst PresIdeNt<br />

George B. Corcoran,<br />

Ph.d., Ats<br />

Wayne state University<br />

COUNCILOrs<br />

Kim Boekelheide,<br />

M.d., Ph.d.<br />

Brown University<br />

Patricia E. Ganey,<br />

Ph.d.<br />

Michigan state University<br />

Denise Rob<strong>in</strong>son Gravatt,<br />

Ph.d.<br />

Pfizer Global research<br />

and development<br />

Ronald N. H<strong>in</strong>es,<br />

Msc, Ph.d., Ats<br />

Medical College of Wiscons<strong>in</strong><br />

exeCUtIVe dIreCtOr<br />

Shawn Douglas Lamb<br />

Thursday, December 4, 2008<br />

Dear Participants:<br />

I am writ<strong>in</strong>g to thank you for your participation <strong>in</strong> the <strong>Contemporary</strong><br />

Concepts <strong>in</strong> Toxicology <strong>Workshop</strong> entitled, “Hemangiosarcoma <strong>in</strong> Rodents:<br />

Mode-of-Action Evaluation and Human Relevance.” The major driver for<br />

these workshops is the desire to offer participants an opportunity to<br />

exam<strong>in</strong>e timely scientific topics of <strong>in</strong>terest over the course of two days.<br />

This workshop br<strong>in</strong>gs together government officials and scientists from<br />

around the world to discuss key issues related to mode of action <strong>in</strong> the<br />

<strong>in</strong>duction of hemangiosarcomas and the assessment of human risk associated<br />

with this disease.<br />

Session One of the workshop opens with an overview and discussion of<br />

workshop objectives. Session Two <strong>in</strong>cludes a discussion titled, “Pathology and<br />

Biology,” which is chaired by Drs. Richard D. Storer and David E. Malarkey.<br />

Session Three opens with a discussion focused around, “Hemangiosarcoma<br />

Induced by Non-Pharmaceuticals.” This session is co-chaired by<br />

Drs. James E. Klaunig and James A. Swenberg. Sessions Four and Five focus<br />

on “Hemangiosarcoma Induced by PPAR Agonists—HESI Initiative” and<br />

“Hemangiosarcoma Induced by Other Pharmaceuticals” and are co-chaired<br />

by Drs. Samuel M. Cohen and Jon C. Cook. Session Six, “Regulatory<br />

Perspectives on Data Gaps,” is co-chaired by Drs. Vicki L. Dellarco and Abigail<br />

C. Jacobs. The f<strong>in</strong>al session, titled “Mode(s) of Action for Hemgangiosarcoma<br />

Induction” is co-chaired by Drs. Samuel M. Cohen and Jon C. Cook.<br />

I would like to thank the follow<strong>in</strong>g members of the Organiz<strong>in</strong>g<br />

Committee for their efforts <strong>in</strong> mak<strong>in</strong>g this CCT workshop a reality:<br />

Neil G. Carmichael, Ph.D.; Samuel M. Cohen, M.D., Ph.D., ATS; Jon C. Cook,<br />

Ph.D., DABT; Vicki L. Dellarco, Ph.D.; Nancy G. Doerrer, M.S.;<br />

Tim G. Hammond; Jerry F. Hardisty, D.V.M., DACVP; Heike Hellmold, Ph.D.;<br />

Abigail C. Jacobs, Ph.D.; David Jacobson-Kram, Ph.D., DABT; James E. Klaunig,<br />

Ph.D., ATS; Mart<strong>in</strong> A. Philbert, Ph.D.; Christopher J. Powell, D.Sc., FRCPath;<br />

Richard D. Storer, Ph.D.; and James A. Swenberg, D.V.M., Ph.D., DACVP.<br />

In clos<strong>in</strong>g, I would also like to thank the follow<strong>in</strong>g sponsors of this workshop<br />

for their generous support: ILSI Health and Environmental Sciences<br />

Institute, Aclairo Pharmaceutical Development Group, Inc., AstraZeneca,<br />

Daiichi-Sankyo, GlaxoSmithKl<strong>in</strong>e, Merck Research Laboratories, Pfizer Inc.,<br />

sanofi aventis, the Society of Toxicologic Pathology, SOT Regulatory and<br />

Safety Evaluation Specialty Section, and Takeda Pharmaceutical Company.<br />

I wish everyone a most successful two days.<br />

S<strong>in</strong>cerely,<br />

Kenneth S. Ramos, B.S.Ph., Ph.D., ATS<br />

SOT President<br />

3<br />

letter from the presiDent


notes<br />

4


sponsors:<br />

SOT Regulatory and Safety Evaluation Specialty Section<br />

5<br />

sponsors


Why Attend the SOT Annual Meet<strong>in</strong>g?<br />

48 th 48 th<br />

Baltimore, Maryland<br />

MARCH 15–19, 2009<br />

www.toxicology.org/2009am • www.toxexpo.com<br />

6<br />

Annual Meet<strong>in</strong>g<br />

& ToxExpo Annual Meet<strong>in</strong>g<br />

& ToxExpo Science—the catch<br />

of the day<br />

Photo courtesy of Baltimore Area Convention and Visitors Association<br />

Cutt<strong>in</strong>g-Edge Science and New Perspectives<br />

The SOT Annual Meet<strong>in</strong>g provides the most comprehensive coverage of toxicology. The scheduled<br />

scientific sessions and poster and platform presentations will present the latest “cutt<strong>in</strong>g-edge”<br />

research.<br />

Depth of Analysis<br />

Five scientific themes will allow attendees to ga<strong>in</strong> depth of analysis on Biomarkers, Epigenetics,<br />

Inflammation and Disease, Nanotechnology, and Neurodegenerative Disease.<br />

Network<strong>in</strong>g<br />

The SOT Annual Meet<strong>in</strong>g, toxicology’s largest meet<strong>in</strong>g, allows you to network with colleagues and<br />

other lead<strong>in</strong>g scientists from around the world.<br />

Value<br />

The SOT Annual Meet<strong>in</strong>g is cost-effective, with low registration fees, <strong>in</strong>expensive high-quality<br />

cont<strong>in</strong>u<strong>in</strong>g education courses, and exposure to the very latest advances <strong>in</strong> science. International<br />

attendees benefit from the good exchange rate.<br />

ToxExpo<br />

The SOT Exhibition, ToxExpo, is the profession’s largest tradeshow and a one-of-a k<strong>in</strong>d event<br />

where attendees from around the globe gather to exchange ideas and debut cutt<strong>in</strong>g-edge<br />

research and technology <strong>in</strong> the field of toxicology. Attendees have the opportunity to ga<strong>in</strong> firsthand<br />

knowledge about the latest products and services offered by more than 350 exhibitors.<br />

Visit www.toxexpo.com for toxicology-related products and services.<br />

D eaD l<strong>in</strong>es<br />

Photos courtesy of Baltimore Area Convention and Visitors Association. All text and graphics © 2008 by the Society of Toxicology unless noted.<br />

January 30, 2009<br />

Early Bird Registration<br />

February 6, 2009<br />

Hous<strong>in</strong>g Reservation<br />

February 20, 2009<br />

Standard Registration<br />

February 20, 2009<br />

Cancellations


organiz<strong>in</strong>g Committee:<br />

Co-Chairpersons<br />

Samuel M. Cohen, M.D., Ph.D., ATS—Professor, Department of Pathology and<br />

Microbiology, Havlik-Wall Professor of Oncology, University of Nebraska Medical Center,<br />

Omaha, NE, United States<br />

Jon C. Cook, Ph.D., DABT—Senior Director, Investigative Toxicology, Pfizer Inc.,<br />

Groton, CT, United States<br />

members<br />

Neil G. Carmichael, Ph.D.—Secretary General, ECETOC AISBL, Brussels, Belgium<br />

Vicki L. Dellarco, Ph.D.—Senior Science Advisor, Office of Pesticide Programs,<br />

U.S. Environmental Protection Agency, Health Effects Division, Wash<strong>in</strong>gton, DC,<br />

United States<br />

Nancy G. Doerrer, M.S.—Associate M.S.— Director, Scientific Program Stewardship,<br />

ILSI Health and Environmental Sciences Institute, Wash<strong>in</strong>gton, DC, United States<br />

Tim G. Hammond Hammond—Vice —Vice President, Safety Assessment UK, AstraZeneca R&D,<br />

United K<strong>in</strong>gdom<br />

Jerry F. Hardisty, D.V.M., DACVP—Experimental Pathology Laboratories, Inc.,<br />

Research Triangle Park, NC, United States<br />

Heike Hellmold, Ph.D.—Pr<strong>in</strong>cipal Scientist and Associate Director Molecular<br />

Toxicology, Safety Assessment, AstraZeneca R&D Sweden, Sweden<br />

Abigail C. Jacobs, Ph.D.—Associate Ph.D.— Director, Pharmacology/Toxicology, Center for<br />

Drug Evaluation and Research, ONDIO, U.S. Food and Drug Adm<strong>in</strong>istration,<br />

Silver Spr<strong>in</strong>g, MD, United States<br />

David Jacobson-Kram, Ph.D., DABT—Office of New Drugs, Center for Drug<br />

Evaluation and Research, U.S. Food and Drug Adm<strong>in</strong>istration, Silver Spr<strong>in</strong>g, MD,<br />

United States<br />

James E. Klaunig, Ph.D., ATS—Robert B. Forney Professor of Toxicology, Director,<br />

Center for Environmental Health, Indiana University School of Medic<strong>in</strong>e, Indianapolis,<br />

IN, United States<br />

Mart<strong>in</strong> A. Philbert, Ph.D.—Professor of Toxicology, University of Michigan, Toxicology<br />

Program, School of Public Health, Ann Arbor, MI, United States<br />

Christopher J. Powell, D.Sc., FRCPath—Director, Safety Assessment Europe,<br />

GlaxoSmithKl<strong>in</strong>e R&D, United K<strong>in</strong>gdom<br />

Richard D. Storer, Ph.D.—Senior Scientific Director, Department of Laboratory<br />

Sciences and Investigative Toxicology, Merck Research Laboratories, West Po<strong>in</strong>t, PA,<br />

United States<br />

James A. Swenberg, D.V.M., Ph.D., DACVP—Director, Curriculum <strong>in</strong> Toxicology,<br />

University of North Carol<strong>in</strong>a at Chapel Hill, School of Health and Medic<strong>in</strong>e, Chapel Hill,<br />

NC, United States<br />

7<br />

organiz<strong>in</strong>g Committee


Be a member of the premier group that is creat<strong>in</strong>g<br />

a safer and healthier world by advanc<strong>in</strong>g the<br />

science of toxicology<br />

As an SOT member you can . . .<br />

Stay connected at www.toxicology.org<br />

Access member-restricted <strong>in</strong>formation<br />

Use the on-l<strong>in</strong>e Member Directory<br />

Receive Reduced Registration Costs<br />

for SOT meet<strong>in</strong>gs<br />

Receive SOT Publications<br />

The Toxicologist (CD-Rom)<br />

Toxicological Sciences<br />

Communiqué<br />

Others<br />

Jo<strong>in</strong> one of 22 Specialty Sections<br />

Choose a Special Interest Group<br />

Participate <strong>in</strong> Your Regional SOT Chapter<br />

Communicate the Importance of your Discipl<strong>in</strong>e<br />

Utilize Career Resources<br />

Register for Mentor Match<br />

Nom<strong>in</strong>ate for Awards<br />

Volunteer and Demonstrate Your Leadership Skills<br />

F<strong>in</strong>d Products and Services Easily at Tox Expo<br />

www.toxicology.org<br />

For complete <strong>in</strong>formation about membership<br />

<strong>in</strong> the Society of Toxicology, visit the<br />

SOT Web site at www.toxicology.org<br />

and select Member Information.<br />

8<br />

Membership Fees:<br />

Full Membership __________________ $135<br />

Associate Membership______________$135<br />

Postdoctoral Membership ____________$35<br />

Student Membership ________________$20<br />

Retired Membership ________________ $0<br />

Easy on-l<strong>in</strong>e membership application takes<br />

approximately 15 m<strong>in</strong>utes to complete.<br />

Special Offer to Non-Member SOT<br />

2009 Annual Meet<strong>in</strong>g Attendees:<br />

• Apply for SOT membership between<br />

January 15, 2009, and the May 1, 2009,<br />

deadl<strong>in</strong>e, and if accepted, SOT will waive<br />

your 2009 dues.<br />

• Applications must be complete and sponsor<br />

letters received by the May 1 deadl<strong>in</strong>e.<br />

March 15–19, 2009


purpose and objectives of the<br />

sot-<strong>CCt</strong> <strong>Workshop</strong><br />

The purpose of the <strong>Workshop</strong> is to explore the modes of action (MOAs)<br />

and human relevance of hemangiosarcoma <strong>in</strong>duced <strong>in</strong> rodents by various<br />

classes of compounds.<br />

background:<br />

Hemangiosarcoma is a malignant tumor of endothelial cells that is rare <strong>in</strong><br />

humans, but common <strong>in</strong> certa<strong>in</strong> breeds of dogs. In carc<strong>in</strong>ogenicity studies<br />

conducted for safety assessment, nongenotoxic agents typically <strong>in</strong>duce<br />

hemangiosarcoma <strong>in</strong> mice but not rats, and the most common target<br />

organs are the liver, spleen, and bone marrow. To date, the MOA for<br />

hemangiosarcoma <strong>in</strong>duction by nongenotoxic compounds is not known,<br />

nor is the human relevance of these tumors understood.<br />

objectives:<br />

1. Summarize current understand<strong>in</strong>g of MOAs for various<br />

compound classes.<br />

2.<br />

3.<br />

Share data and <strong>in</strong>formation with the scientific and regulatory<br />

communities to promote and guide future research on<br />

nongenotoxic MOAs for hemangiosarcoma <strong>in</strong> rodents.<br />

Identify research tools and approaches to study<strong>in</strong>g<br />

hemangiosarcoma and related vascular lesions.<br />

9<br />

meet<strong>in</strong>g objeCtiVes


notes<br />

10


Day D 1<br />

thursday, December 4, 2008 8:00 am–5:00 pm<br />

Session One: Welcome<br />

Session Two: Pathology and Biology<br />

Session Three: Hemangiosarcoma Induced by Non-Pharmaceuticals<br />

Day D 2<br />

friday, December 5, 2008 8:00 am–3:15 pm<br />

Session Four: Hemangiosarcoma Induced by PPAR Agonists—<br />

HESI Initiative<br />

Session Five: Hemangiosarcoma Induced by Other Pharmaceuticals<br />

Session Six: Regulatory Perspectives on Data Gaps<br />

Session Seven: Mode(s) of Action for Hemangiosarcoma Induction<br />

11<br />

agenDa


Day 1<br />

thursday, December 4, 2008<br />

session one, 8:00 am–8:15 am: Welcome<br />

Session Co-Chairs: Samuel M. Cohen, M.D., Ph.D., ATS, University of Nebraska Medical Center,<br />

Omaha, NE, United States and Jon C. Cook, Ph.D., DABT, Pfizer Inc., Groton, CT, United States<br />

time: topic: speaker:<br />

7:00 AM–8:00 AM Open Registration<br />

8:00 AM–8:05 AM Welcome George B. Corcoran, Wayne State<br />

University, Detroit, MI<br />

8:05 AM–8:15 AM Welcom<strong>in</strong>g Remarks and<br />

<strong>Workshop</strong> Objectives<br />

12<br />

Jon C. Cook, Pfizer Inc,<br />

Groton, CT<br />

Samuel M. Cohen, University<br />

of Nebraska Medical Center,<br />

Omaha, NE<br />

session tWo, 8:15 am–11:45 am: pathology and biology<br />

Session Co-Chairs: Richard D. Storer, Ph.D., Merck Research Laboratories, West Po<strong>in</strong>t, PA,<br />

United States and David E. Malarkey, D.V.M., Ph.D., DACVP, NIEHS National Toxicology<br />

Program, Research Triangle Park, NC, United States<br />

time: topic: speaker:<br />

8:15 AM–8:30 AM Speaker Introductions Richard D. Storer, Merck<br />

Research Laboratories,<br />

West Po<strong>in</strong>t, PA<br />

8:30 AM–9:15 AM Trivial to Catastrophic,<br />

Necessary to Deadly: Vascular<br />

Proliferations and Malignancies<br />

<strong>in</strong> Humans and Animals<br />

9:15 AM–10:00 AM Pathology of Treatment-Induced<br />

Hemangiosarcoma <strong>in</strong> NTP<br />

Studies<br />

10:00 AM–10:15 AM BREAK<br />

10:15 AM–11:00 AM The Effect of Genetic Diversity<br />

on Angiogenesis<br />

11:00 AM–11:45 AM Bark<strong>in</strong>g Up the Right Tree:<br />

Uncover<strong>in</strong>g the Influence of<br />

Heritable Factors on Can<strong>in</strong>e<br />

Hemangiosarcoma<br />

lunCh anD poster VieW<strong>in</strong>g<br />

11:45 am–2:00 pm<br />

Sir Col<strong>in</strong> Berry, Queen Mary,<br />

University of London, London,<br />

United K<strong>in</strong>gdom<br />

David E. Malarkey, NIEHS<br />

National Toxicology Program,<br />

Research Triangle Park, NC<br />

Michael S. Rogers, Harvard<br />

Medical School, Boston, MA<br />

Jaime F. Modiano, University of<br />

M<strong>in</strong>nesota, M<strong>in</strong>neapolis, MN


session three, 2:00 pm–4:30 pm: hemangiosarcoma <strong>in</strong>duced by<br />

non-pharmaceuticals<br />

Session Co-Chairs: James E. Klaunig, Ph.D., ATS, Indiana University School of Medic<strong>in</strong>e,<br />

Indianapolis, IN, United States and James A. Swenberg, D.V.M., Ph.D., DACVP, University of<br />

North Carol<strong>in</strong>a at Chapel Hill, Chapel Hill, NC, United States<br />

time: topic: speaker:<br />

2:00 PM–2:15 PM Speaker Introductions James E. Klaunig, Indiana<br />

University School of Medic<strong>in</strong>e,<br />

Indianapolis, IN<br />

2:15 PM–2:45 PM Us<strong>in</strong>g Mode-of-Action Data to<br />

Assess the Human Relevance of<br />

Animal Tumors<br />

2:45 PM–3:15 PM Mode of Action of V<strong>in</strong>yl<br />

Chloride-Induced Hepatic<br />

Hemangiosarcoma<br />

3:15 PM–3:30 PM BREAK<br />

3:30 PM–4:00 PM Vascular Tumor Potential of<br />

Carbaryl <strong>in</strong> the Heterozygous p53<br />

Knockout Mouse Model<br />

4:00 PM–4:30 PM Nongenotoxic Agents: Studies<br />

with 2-Butoxyethanol<br />

Day 1 Clos<strong>in</strong>g<br />

time: topic: speaker:<br />

13<br />

Vicki L. Dellarco, Office of<br />

Pesticide Programs, U.S.<br />

Environmental Protection<br />

Agency, Wash<strong>in</strong>gton, DC<br />

James A. Swenberg, University<br />

of North Carol<strong>in</strong>a at Chapel Hill,<br />

Chapel Hill, NC<br />

Dom<strong>in</strong>ique Lasserre-Bigot, Bayer<br />

CropScience, Sophia Antipolis,<br />

France<br />

James E. Klaunig, Indiana<br />

University School of Medic<strong>in</strong>e,<br />

Indianapolis, IN<br />

4:30 PM–5:00 PM Comments and Questions Jon C. Cook, Pfizer Inc.,<br />

Groton, CT<br />

5:00 PM ADJOURN DAY ONE (d<strong>in</strong>ner on your own)


Day 2<br />

friday, December 5, 2008<br />

session four, 8:00 am–10:15 am: hemangiosarcoma <strong>in</strong>duced by<br />

ppar agonists—hesi <strong>in</strong>itiative<br />

Session Co-Chairs: Samuel M. Cohen, M.D., Ph.D., ATS, University of Nebraska Medical Center,<br />

Omaha, NE, United States and Jon C. Cook, Ph.D., DABT, Pfizer Inc., Groton, CT, United States<br />

time: topic: speaker:<br />

7:00 AM–8:00 AM Open Registration<br />

8:00 AM–8:10 AM Speaker Introductions Samuel M. Cohen, University<br />

of Nebraska Medical Center,<br />

Omaha, NE<br />

8:10 AM–8:45 AM Angiogenesis and Adipogenesis Keith L. March, Indiana<br />

University School of Medic<strong>in</strong>e,<br />

Indianapolis, IN<br />

8:45 AM–9:20 AM Troglitazone: An Illustration<br />

of the HESI Mode-of-Action<br />

Framework for PPAR Gamma-<br />

Induced Hemangiosarcoma<br />

9:20 AM–9:55 AM Troglitazone Effects on<br />

Endothelial Cells In Vivo and<br />

In Vitro: Differences between<br />

Mice and Humans<br />

9:55 AM–10:15 AM BREAK<br />

14<br />

Jon C. Cook, Pfizer Inc.,<br />

Groton, CT<br />

Samuel M. Cohen, University<br />

of Nebraska Medical Center,<br />

Omaha, NE<br />

session fiVe, 10:15 am–11:35 am: hemangiosarcoma <strong>in</strong>duced by<br />

other pharmaceuticals<br />

Session Co-Chairs: Samuel M. Cohen, M.D., Ph.D., ATS, University of Nebraska Medical Center,<br />

Omaha, NE, United States and Jon C. Cook, Ph.D., DABT, Pfizer Inc., Groton, CT, United States<br />

time: topic: speaker:<br />

10:15 AM–10:25 AM Speaker Introductions Jon C. Cook, Pfizer Inc.,<br />

Groton, CT<br />

10:25 AM–11:00 AM Ret<strong>in</strong>oid-Induced<br />

Hemangiosarcoma <strong>in</strong> Mice:<br />

Potential Insight from In Vivo and<br />

In Vitro Studies<br />

11:00 AM–11:35 AM Epigenetic Mode of Action<br />

Associated with Induction of<br />

Hemangiosarcoma <strong>in</strong> Mice<br />

Treated with Pregabal<strong>in</strong><br />

lunCh (on your oWn)<br />

11:35 am–12:45 pm<br />

Timothy E. Johnson, Merck<br />

Research Laboratories,<br />

West Po<strong>in</strong>t, PA<br />

Kay A. Criswell, Pfizer Global<br />

Research and Development,<br />

Groton, CT


session six, 12:45 pm–2:00 pm: regulatory perspectives on Data gaps<br />

Session Co-Chairs: Vicki L. Dellarco, Ph.D., Office of Pesticide Programs, U.S. Environmental<br />

Protection Agency, Wash<strong>in</strong>gton, DC, United States and Abigail C. Jacobs, Ph.D., Center for Drug<br />

Evaluation and Research, U.S. Food and Drug Adm<strong>in</strong>istration, Silver Spr<strong>in</strong>g, MD, United States<br />

time: topic: speaker:<br />

12:45 PM–1:00 PM Speaker Introductions Vicki L. Dellarco, Office of<br />

Pesticide Programs,<br />

U.S. Environmental Protection<br />

Agency, Wash<strong>in</strong>gton, DC<br />

1:00 PM–1:10 PM EPA Perspectives Vicki L. Dellarco, Office of<br />

Pesticide Programs,<br />

U.S. Environmental Protection<br />

Agency, Wash<strong>in</strong>gton, DC<br />

1:10 PM–1:35 PM Hemangiosarcomas and<br />

Pharmaceuticals: An FDA<br />

Perspective<br />

1:35 PM–2:00 PM European Perspectives on<br />

Carc<strong>in</strong>ogenicity Test<strong>in</strong>g<br />

15<br />

Abigail C. Jacobs, Center<br />

for Drug Evaluation and<br />

Research, U.S. Food and Drug<br />

Adm<strong>in</strong>istration, Silver Spr<strong>in</strong>g,<br />

MD<br />

Jan Willem van der Laan,<br />

National Institute of Public<br />

Health and the Environment<br />

(RIVM), Bilthoven,<br />

The Netherlands<br />

session seVen, 2:00 pm–3:00 pm: mode(s) of action for<br />

hemangiosarcoma <strong>in</strong>duction<br />

Session Co-Chairs: Samuel M. Cohen, M.D., Ph.D., ATS, University of Nebraska Medical Center,<br />

Omaha, NE, United States and Jon C. Cook, Ph.D., DABT, Pfizer Inc., Groton, CT, United States<br />

time: topic: speaker:<br />

2:00 PM–3:00 PM Panel-Led Forum:<br />

Open Discussion<br />

Day 2 Clos<strong>in</strong>g<br />

time: topic: speaker:<br />

Moderators:<br />

Samuel M. Cohen, University<br />

of Nebraska Medical Center,<br />

Omaha, NE<br />

Jon C. Cook, Pfizer Inc.,<br />

Groton, CT<br />

3:00 PM–3:15 PM Clos<strong>in</strong>g Comments Jon C. Cook, Pfizer Inc.,<br />

Groton, CT<br />

3:15 PM ADJOURN


notes<br />

16


iographies<br />

Sir Col<strong>in</strong> Berry<br />

Samuel M. Cohen<br />

Jon C. Cook<br />

George B. Corcoran<br />

Kay A. Criswell<br />

Vicki L. Dellarco<br />

Abigail C. Jacobs<br />

Timothy E. Johnson<br />

James E. Klaunig<br />

Dom<strong>in</strong>ique R. Lasserre-Bigot<br />

David E. Malarkey<br />

Keith L. March<br />

Jaime F. Modiano<br />

Michael S. Rogers<br />

Richard D. Storer<br />

James A. Swenberg<br />

Jan Willem van der Laan<br />

17<br />

biographies


speaker biographies<br />

Sir Col<strong>in</strong> Berry, D.Sc., M.D., Ph.D., FRCPath, FRCP, Queen Mary, University of London,<br />

London, United K<strong>in</strong>gdom<br />

After <strong>in</strong>itial tra<strong>in</strong><strong>in</strong>g <strong>in</strong> Medic<strong>in</strong>e and Pathology at Char<strong>in</strong>g Cross Hospital, where he won the Governors<br />

Cl<strong>in</strong>ical Gold Medal, Sir Col<strong>in</strong> became Lecturer at the Institute of Child Health, Great Ormond Street, and<br />

was there for six years, becom<strong>in</strong>g a British Heart Foundation Research Fellow and Senior Lecturer three<br />

years later. He was appo<strong>in</strong>ted Reader <strong>in</strong> Pathology at Guy’s Hospital <strong>in</strong> 1970 and Professor and Head of the<br />

Department of Pathology at the Royal London Hospital <strong>in</strong> 1976.<br />

He has worked <strong>in</strong> Vascular Pathology and Paediatric Pathology, has written standard texts on both subjects,<br />

and has a body of experimental work <strong>in</strong> vascular pathology, ma<strong>in</strong>ly concerned with biomechanics.<br />

His <strong>in</strong>terest <strong>in</strong> toxicology—developed as a result of work <strong>in</strong> the early days of the culture of vertebrate<br />

embryos—has resulted <strong>in</strong> his serv<strong>in</strong>g as a member of the Committee on Safety of Medic<strong>in</strong>es and as a<br />

member of a number of other related committees of the European Commission. He was Chairman of the<br />

Advisory Committee on Pesticides for 10 years; this aspect of his work means that he has advised the<br />

British Potato Council and addressed the World Banana Congress.<br />

Sir Col<strong>in</strong> is a former member of the Medical Research Council of Great Brita<strong>in</strong> and has rema<strong>in</strong>ed an<br />

advisor to that body. He is a former President of the European Society of Pathology and of the British<br />

Academy of Forensic Sciences. He was elected to the German Academy of Sciences (Deutsche Akademie<br />

der Natureforscher Leopold<strong>in</strong>a) <strong>in</strong> 1993 and made Knight Bachelor <strong>in</strong> the Queen’s Birthday Honours List<br />

<strong>in</strong> the same year, for services to Medic<strong>in</strong>e and Science. He is an Honorary Fellow of the Royal College of<br />

Physicians, of the Faculty of Occupational Medic<strong>in</strong>e, the Faculty of Pharmaceutical Medic<strong>in</strong>e and of the<br />

Royal College of Physicians of Ed<strong>in</strong>burgh, and has a number of honorary degrees from overseas<br />

universities.<br />

S<strong>in</strong>ce retirement from his University Chair, he has been <strong>in</strong>creas<strong>in</strong>gly <strong>in</strong>volved <strong>in</strong> work on the public<br />

understand<strong>in</strong>g of science, and serves on the Steer<strong>in</strong>g Committee of the European Science Open Forum and<br />

is Chairman of its Programme Committee. He is on the Management Board of Sense about Science, a body<br />

that advises Parliamentarians about the consequences of scientific legislation and regularly presents topics<br />

to the Parliamentary and Scientific Committee. He is an Honorary Curator of the Deutches Museum <strong>in</strong><br />

Munich.<br />

In 2005, he was made an honorary fellow of the German Pathological Society and of the British Society of<br />

Toxicology.<br />

Samuel M. Cohen, M.D., Ph.D., ATS, University of Nebraska Medical Center, Omaha, NE, United States<br />

Dr. Samuel M. Cohen (M.D., Ph.D., University of Wiscons<strong>in</strong>, 1972) tra<strong>in</strong>ed <strong>in</strong> anatomic and cl<strong>in</strong>ical<br />

pathology and is board certified (1976). He has been professor at the University of Nebraska Medical<br />

Center s<strong>in</strong>ce 1981, serv<strong>in</strong>g as Vice Chair for eleven years and Chair for fifteen years. He has been actively<br />

<strong>in</strong>volved <strong>in</strong> chemical carc<strong>in</strong>ogenesis research and risk assessment, has greater than 300 publications, has<br />

served on numerous national and <strong>in</strong>ternational agency committees and panels, has received several awards<br />

<strong>in</strong>clud<strong>in</strong>g the SOT Lehman Award, and is a member of the ILSI Health and Environmental Sciences<br />

Institute PPAR Agonist Project Committee.<br />

18


Jon C. Cook, Ph.D., DABT, Pfizer Inc., Groton, CT, United States<br />

Dr. Jon C. Cook received a BS <strong>in</strong> Physiology from the University of California at Davis (1979) and<br />

Ph.D. <strong>in</strong> Toxicology from North Carol<strong>in</strong>a State University (1985). He was a postdoctoral fellow at the<br />

Chemical Industry Institute of Toxicology before jo<strong>in</strong><strong>in</strong>g DuPont <strong>in</strong> 1987. At DuPont, Jon developed an<br />

<strong>in</strong>terest <strong>in</strong> carc<strong>in</strong>ogenesis focus<strong>in</strong>g on endocr<strong>in</strong>e-mediated mechanisms. In 1998, Jon jo<strong>in</strong>ed Pfizer where<br />

he contributed to the registration of a COX-2 <strong>in</strong>hibitor (Celebrex) and a SERM (Fablyn). He currently<br />

leads an <strong>in</strong>vestigative toxicology group that focuses on discovery toxicology and late-stage portfolio<br />

support. In 1998, he received the Robert A. Scala Award <strong>in</strong> Toxicology. Jon has been President of the SOT<br />

Carc<strong>in</strong>ogenesis Specialty Section and is co-chair of the ILSI Health and Environmental Sciences Institute<br />

PPAR Agonist Project Committee.<br />

George B. Corcoran, Ph.D., ATS, Wayne State University, Detroit, MI, United States<br />

Dr. George B. Corcoran is Professor and the Chairman of the Department of Pharmaceutical Sciences,<br />

College of Pharmacy and Health Sciences, Wayne State University, and Adjunct Professor of Pediatrics,<br />

Wayne State University School of Medic<strong>in</strong>e. He received his bachelor’s degree <strong>in</strong> Chemistry from Ithaca<br />

College and his master’s degree <strong>in</strong> Organic Chemistry from Bucknell University. He received his doctoral<br />

degree <strong>in</strong> Pharmacology/Toxicology from George Wash<strong>in</strong>gton University and completed postdoctoral<br />

tra<strong>in</strong><strong>in</strong>g <strong>in</strong> toxicology at the Baylor College of Medic<strong>in</strong>e.<br />

Prior to com<strong>in</strong>g to Wayne State University, Dr. Corcoran served as Assistant Professor Pharmaceutics at the<br />

State University of New York at Buffalo. He also served for n<strong>in</strong>e years at the University of New Mexico<br />

as Associate Professor, Professor, and Director of the Toxicology Graduate Program. He has authored<br />

more than 170 orig<strong>in</strong>al research papers, abstracts and other reports. His research <strong>in</strong>terests focus on factors<br />

govern<strong>in</strong>g drug and chemical-<strong>in</strong>duced <strong>in</strong>juries and cellular <strong>in</strong>jury and cell death. He served as President of<br />

the Society of Toxicology (SOT) <strong>in</strong> 2007 and is currently serv<strong>in</strong>g as Past President of SOT. He is a Fellow<br />

of the Academy of Toxicological Sciences, a Delegate to the International Congress of Toxicology and is a<br />

member of the International Union of Toxicology Develop<strong>in</strong>g Countries Committee. Dr. Corcoran is also<br />

a member of the Scientific Advisory Board of the U.S. Environmental Protection Agency and a member of<br />

The Executive Board of the Council of Scientific Society Presidents.<br />

Kay A. Criswell, Ph.D., ASCP, Pfizer Global Research and Development, Groton, CT, United States<br />

Dr. Kay A. Criswell received her bachelor’s degree <strong>in</strong> Medical Technology from Northern Michigan<br />

University and her master’s and Ph.D. <strong>in</strong> Toxicology from the University of Michigan. She served as the<br />

Director of Laboratory Services at Providence Hospital <strong>in</strong> Southfield, Michigan for 13 years cover<strong>in</strong>g<br />

hematology, coagulation, immunology and flow cytometric services. She jo<strong>in</strong>ed Warner-Lambert Parke-<br />

Davis <strong>in</strong> 1994 as a Scientist <strong>in</strong> Toxicology. She has assumed positions of <strong>in</strong>creas<strong>in</strong>g responsibility <strong>in</strong><br />

Pfizer Global Research & Development, advanc<strong>in</strong>g to Senior Director of Investigative Toxicology at the<br />

Ann Arbor, Michigan site. In June 2008, she assumed a role as Senior Director <strong>in</strong> charge of Drug Safety<br />

Biomarker Laboratories <strong>in</strong> Groton, CT, and lead for the global biomarker leadership team with<strong>in</strong> Pfizer.<br />

Her current areas of leadership <strong>in</strong>clude cl<strong>in</strong>ical pathology, flow cytometry, bioanalytical development,<br />

and biomarker development and translation. She is the global lead for several issues management teams<br />

regard<strong>in</strong>g hematologic toxicities. She is also the current drug safety team lead for Lyrica, <strong>in</strong>clud<strong>in</strong>g<br />

management of regulatory issues, and lead for the mouse-specific hemangiosarcoma mechanism of action<br />

team. She serves as a liaison between Drug Safety and Cl<strong>in</strong>ical and Discovery teams provid<strong>in</strong>g guidance<br />

for development of safety biomarkers for early research programs and translation of biomarkers from<br />

precl<strong>in</strong>ical safety test<strong>in</strong>g to first <strong>in</strong> human trials. She has served as a former Secretary of the Society of<br />

Toxicology and is currently the chair-elect of the Division of Animal Cl<strong>in</strong>ical Chemistry. She has held<br />

adjunct faculty positions at the University of Michigan School of Public Health and Eastern Michigan<br />

University Program of Toxicology.<br />

19<br />

(Cont<strong>in</strong>ued on next page)


Vicki L. Dellarco, Ph.D., Office of Pesticide Programs, U.S. Environmental Protection Agency,<br />

Wash<strong>in</strong>gton, DC, United States<br />

Dr. Vicki L. Dellarco is a Senior Science Advisor <strong>in</strong> the Office of Pesticide Programs (OPP) at the U.S.<br />

Environmental Protection Agency. She has led the development of several major science policies <strong>in</strong>volv<strong>in</strong>g<br />

the implementation of the 1996 Food Quality Protection Act, <strong>in</strong>clud<strong>in</strong>g the development of mode-of-action<br />

decisions, cumulative risk assessment methods and guidance, as well as the development of toxicity test<strong>in</strong>g<br />

strategies for improv<strong>in</strong>g and ref<strong>in</strong><strong>in</strong>g approaches to health risk assessment. She has also served as a Senior<br />

Science Advisor <strong>in</strong> other Agency programs <strong>in</strong>clud<strong>in</strong>g the National Center for Environmental Assessment<br />

and the Office of Water. She is a member of U.S. EPA’s Risk Assessment Forum, Human Health Oversight<br />

Committee, and also serves on several <strong>in</strong>ternational committees. Dr. Dellarco is the 2008 recipient of the<br />

Arnold J. Lehman Award from the Society of Toxicology.<br />

Abigail C. Jacobs, Ph.D., Center for Drug Evaluation and Research, U.S. Food and Drug Adm<strong>in</strong>istration,<br />

Silver Spr<strong>in</strong>g, MD, United States<br />

Dr. Abigail (Abby) C. Jacobs is currently an Associate Director for Pharmacology/Toxicology for<br />

several U.S. FDA/CDER Offices of Drug Evaluation. She received a BS <strong>in</strong> Chemistry at the University<br />

of Michigan, Ann Arbor, and a Ph.D. <strong>in</strong> Biochemistry at the University of California, Berkeley. After<br />

postdoctoral work <strong>in</strong> immunochemistry and mast cell biochemistry, she became a toxicologist. She spent<br />

numerous years work<strong>in</strong>g for the NCI/NTP and the Division of AIDS, NIAID, NIH, as a contractor for<br />

toxicologic and carc<strong>in</strong>ogenicity evaluation before jo<strong>in</strong><strong>in</strong>g the Division of Antiviral Drug Products,<br />

U.S. FDA/CDER, as a Toxicology Reviewer <strong>in</strong> 1991. For eight years, Abby was Pharmacology/Toxicology<br />

Supervisor of the Division of Dermatologic and Dental Products, U.S. FDA/CDER. She is a stand<strong>in</strong>g<br />

member of the CDER, U.S. FDA, Executive Carc<strong>in</strong>ogenicity Assessment Committee. She represents<br />

U.S. FDA on ICCVAM and on OECD pharm/tox issues, and is on the OECD expert work<strong>in</strong>g group on<br />

carc<strong>in</strong>ogenicity evaluation. She is the rapporteur and U.S. FDA topic leader for revisions to the ICHM3<br />

guidel<strong>in</strong>e.<br />

Timothy E. Johnson, Ph.D., Merck Research Laboratories, West Po<strong>in</strong>t, PA, United States<br />

Dr. Timothy E. Johnson, Research Fellow <strong>in</strong> Laboratory Science and Investigative Toxicology at Merck<br />

Research Laboratories, holds a B.A. <strong>in</strong> Biology from Glassboro State College and an M.S. and a Ph.D. <strong>in</strong><br />

Cell and Developmental Biology from Rutgers University. Before jo<strong>in</strong><strong>in</strong>g Merck, he worked at the Coriell<br />

Institute for Medical Research <strong>in</strong> Camden, NJ. He has held various positions over his 20 years at Merck.<br />

Dr. Johnson currently leads a group of scientists <strong>in</strong>volved <strong>in</strong> <strong>in</strong>vestigat<strong>in</strong>g drug-<strong>in</strong>duced toxicities that occur<br />

<strong>in</strong> precl<strong>in</strong>ical species.<br />

James E. Klaunig, Ph.D., ATS, Indiana University School of Medic<strong>in</strong>e, Indianapolis, IN, United States<br />

Dr. James E. Klaunig is the Robert B. Forney Professor and Director of Toxicology <strong>in</strong> the Department<br />

of Pharmacology and Toxicology, the found<strong>in</strong>g Director of the Center for Environmental Health, and,<br />

the Associate Director of the Cancer Center at Indiana University. He received his B.S. <strong>in</strong> Biology from<br />

Urs<strong>in</strong>us College, Collegeville, PA, and a Ph.D. <strong>in</strong> Experimental Pathology (B. Trump, Mentor) from the<br />

University of Maryland, Baltimore, MD. After postdoctoral studies, he became Assistant Professor and<br />

then Associate Professor <strong>in</strong> the Departments of Pathology and Pharmacology at the Medical University of<br />

Ohio, Toledo, OH. Follow<strong>in</strong>g a sabbatical year at the Chemical Industry Institute of Toxicology, he took<br />

the professorship at Indiana University <strong>in</strong> 1991. His research has focused on the mechanisms of chemically<br />

<strong>in</strong>duced carc<strong>in</strong>ogenesis with emphasis on the epigenetic modes of action. This has <strong>in</strong>volved studies <strong>in</strong>to<br />

the role of oxidative stress/oxidative damage, Kupffer cell activation, modulation of gap junctions, and<br />

20


cell growth/apoptosis <strong>in</strong> this process. His research is supported by the National Institutes of Health (NIH),<br />

U.S. EPA, and non federal sources of support. He is active <strong>in</strong> the Society of Toxicology hav<strong>in</strong>g served on<br />

elected and appo<strong>in</strong>ted committees over the past 16 years. He serves as a member of the U.S. EPA Science<br />

Advisory Board, a Trustee of the Board and Executive Committee of the ILSI Health and Environmental<br />

Sciences Institute, and a Member of the Board of Directors of the Toxicology Forum. In 2002, he received<br />

the Kenneth P. DuBois Award from the Midwest Society of Toxicology Chapter. From Indiana University,<br />

he has also received the Otis R. Bowen, M.D. Dist<strong>in</strong>guished Leadership Award and the Indiana University<br />

Board of Trustees’ Teach<strong>in</strong>g Award. He received the Sagamore of the Wabash, the highest award given<br />

for extraord<strong>in</strong>ary service to the State of Indiana by the Governor. He is a Fellow <strong>in</strong> the Academy of<br />

Toxicological Sciences. He has published over 195 peer reviewed manuscripts and book chapters, and has<br />

mentored over 50 M.S., Ph.D., and postdoctoral fellows <strong>in</strong> toxicology and chemical carc<strong>in</strong>ogenesis. He was<br />

Editor of Toxicologic Pathology from 2004–2007.<br />

Dom<strong>in</strong>ique R. Lasserre-Bigot, Ph.D., Bayer CropScience, Sophia Antipolis, France<br />

Dr. Dom<strong>in</strong>ique Lasserre-Bigot received her bachelor’s degree <strong>in</strong> Pharmaceutical Studies at the University<br />

of Paris V <strong>in</strong> 1986, a masters degree <strong>in</strong> Experimental Pharmacology, Molecular Pharmacochemistry and<br />

Metabolism the follow<strong>in</strong>g year, and her doctoral degree <strong>in</strong> 1991 after hav<strong>in</strong>g spent three years at the<br />

MRC <strong>in</strong> Cambridge, UK work<strong>in</strong>g on the reorganization of the neuronal skeleton after excitatory am<strong>in</strong>o<br />

acid stimulation. S<strong>in</strong>ce then, she has been work<strong>in</strong>g <strong>in</strong> toxicology for agrochemical companies (Rhône-<br />

Poulenc, Aventis, and now Bayer CropScience), start<strong>in</strong>g as Study Director <strong>in</strong> Neurotoxicology and then <strong>in</strong><br />

Hepatotoxicology. After 13 years <strong>in</strong> research experimental toxicology she moved to regulatory toxicology<br />

for the development of new active <strong>in</strong>gredients and the support of marketed agrochemicals.<br />

David E. Malarkey, D.V.M., Ph.D., DACVP, National Institute of Environmental Health Sciences (NIEHS)<br />

National Toxicology Program, Research Triangle Park, NC, United States<br />

Dr. David E. Malarkey is currently the Head of the National Toxicology Program’s Pathology Group at the<br />

National Institute of Environmental Health Sciences. He received his D.V.M. from Tufts University School<br />

of Veter<strong>in</strong>ary Medic<strong>in</strong>e; Pathology Residency tra<strong>in</strong><strong>in</strong>g at Angell Memorial Animal Hospital <strong>in</strong> Boston;<br />

and Ph.D. from North Carol<strong>in</strong>a State University (NCSU). He has been a Diplomate of the American<br />

College of Veter<strong>in</strong>ary Pathologists s<strong>in</strong>ce 1993, and has over 50 publications <strong>in</strong> the areas of toxicological<br />

pathology, carc<strong>in</strong>ogenesis, and molecular pathology. Dr. Malarkey was previously a Research Fellow under<br />

the direction of Dr. Robert Maronpot at NIEHS and Assistant Professor at NCSU College of Veter<strong>in</strong>ary<br />

Medic<strong>in</strong>e.<br />

Keith L. March, M.D., Ph.D., Indiana University School of Medic<strong>in</strong>e, Indianapolis, IN, United States<br />

Dr. Keith L. March has dedicated his career to br<strong>in</strong>g<strong>in</strong>g new medical approaches to patients. His<br />

publications <strong>in</strong>clude more than 75 manuscripts. He was the editor of the first book dedicated to<br />

cardiovascular gene transfer. Dr. March’s research has resulted <strong>in</strong> more than 40 worldwide (19 U.S.)<br />

patents, with others pend<strong>in</strong>g. He is well-known for <strong>in</strong>vent<strong>in</strong>g the Closer, a patented suture-mediated closure<br />

device that is now used annually worldwide <strong>in</strong> 500,000 patients, which closes the puncture wound <strong>in</strong> an<br />

artery follow<strong>in</strong>g heart catheterization. He has served as a scientific advisor to numerous pharmaceutical,<br />

biotechnology and medical device companies.<br />

His laboratory focuses on vascular biology, with a particular emphasis on the function and translational<br />

study of stem cells found <strong>in</strong> the adipose tissue, which his laboratory identified as peri-vascular cells with<br />

critical roles <strong>in</strong> vasculogenesis, angiogenesis, and adipose tissue regulation.<br />

21<br />

(Cont<strong>in</strong>ued on next page)


As Director of the Indiana Center for Vascular Biology & Medic<strong>in</strong>e, Dr. March leads a research team of<br />

more than 30 <strong>in</strong>vestigators dedicated to the development of revolutionary medical therapies, devices, drugs<br />

and genetic <strong>in</strong>terventions for patients of all vascular diseases. Dr. March is widely recognized as a lead<strong>in</strong>g<br />

expert <strong>in</strong> the field of adult stem cell research, particularly that <strong>in</strong>volv<strong>in</strong>g adipose-derived stromal stem cells.<br />

His accomplishments with basic studies of adult stem cells and his <strong>in</strong>volve ment <strong>in</strong> pioneer<strong>in</strong>g cl<strong>in</strong>ical trials<br />

ongo<strong>in</strong>g at the ICVBM have led to his appo<strong>in</strong>tment as a National Institutes of Health (NIH) Chairperson to<br />

oversee all U.S. cell therapy trials <strong>in</strong> the areas of heart lung and blood diseases.<br />

The advancements made by Dr March’s ICVBM research team have stimulated a commitment from<br />

Indiana University-Purdue University <strong>in</strong> Indianapolis (IUPUI) to form, under his direction the “Signature<br />

Center” IUPUI Vascular and Cardiac Center of Adult Stem Cell Cast (VC-CAST) to foster adult stem cell<br />

research, particularly as related to vascular and cardiac disease.<br />

In addition to his research roles, Dr. March has served as the President (2007) of the International<br />

Federation of Adipose Therapeutics and Science Association (IFATS), and serves as the Chief Medical<br />

Advisor for the Cell Therapy Foundation. In both affiliations, he reaches out to advance general awareness<br />

of adult stem cell research by contribut<strong>in</strong>g to educat<strong>in</strong>g laypersons about the significance of adult stem<br />

cells.<br />

Dr. March is Professor, Departments of Medic<strong>in</strong>e, Cellular and Integrative Physiology, and Biomedical<br />

Eng<strong>in</strong>eer<strong>in</strong>g, Indiana University School of Medic<strong>in</strong>e and the Krannert Institute of Cardiology; Director,<br />

Indiana Center for Vascular Biology & Medic<strong>in</strong>e (ICVBM); and Director, IUPUI Vascular and Cardiac<br />

Center of Adult Stem Cell Cast (VC-CAST)<br />

Jaime F. Modiano, V.M.D., Ph.D., University of M<strong>in</strong>nesota, M<strong>in</strong>neapolis, MN, United States<br />

Dr. Jaime F. Modiano did undergraduate work at Texas A&M, earned his V.M.D. and Ph.D. <strong>in</strong> Immunology<br />

at the University of Pennsylvania, tra<strong>in</strong>ed as a resident <strong>in</strong> Cl<strong>in</strong>ical Pathology at Colorado State, and<br />

completed his post-doc at National Jewish. He has led an <strong>in</strong>dependent research program focused on cancer<br />

genetics and therapeutic development with an emphasis on immunotherapy for 13 years. Currently, he<br />

cont<strong>in</strong>ues his research program as Professor and Program Leader of Comparative Oncology, hold<strong>in</strong>g the<br />

Al and June Perlman Endowed Chair, jo<strong>in</strong>tly appo<strong>in</strong>ted at the College of Veter<strong>in</strong>ary Medic<strong>in</strong>e, the School<br />

of Medic<strong>in</strong>e, and the Masonic Cancer Center of the University of M<strong>in</strong>nesota.<br />

Michael S. Rogers, Ph.D., Harvard University, Boston, MA, United States<br />

Dr. Michael S. Rogers graduated with a BS <strong>in</strong> Molecular Biology and Microbiology from Brigham Young<br />

University <strong>in</strong> 1993. He received a Ph.D. <strong>in</strong> Tumor Biology from the Mayo Graduate School <strong>in</strong> 2000. S<strong>in</strong>ce<br />

that time, he has worked <strong>in</strong> the Vascular Biology Program at Children’s Hospital Boston, where he is<br />

currently an Instructor at Harvard Medical School. His major area of research has been elucidation of the<br />

genetic regulation of angiogenesis. He has identified several quantitative trait loci regulat<strong>in</strong>g differential<br />

response to VEGF and bFGF <strong>in</strong> <strong>in</strong>bred stra<strong>in</strong> crosses, as well as loci regulat<strong>in</strong>g choroidal angiogenesis.<br />

22


Richard D. Storer, Ph.D., Merck Research Laboratories, West Po<strong>in</strong>t, PA, United States<br />

Dr. Richard D. Storer received a Bachelor of Arts <strong>in</strong> English from Harvard College <strong>in</strong> 1973, a Master of<br />

Public Health Degree <strong>in</strong> Environmental Health from the University of Michigan School of Public Health<br />

<strong>in</strong> 1979, and a Ph.D. degree <strong>in</strong> Toxicology from the University of Michigan <strong>in</strong> 1983. His thesis work with<br />

Dr. Rory Conolly <strong>in</strong>volved <strong>in</strong> vivo genotoxicity assessment of 1,2-dihaloethanes. He did his postdoctoral<br />

work with Dr. Matthews Bradley at Merck Research Laboratories (MRL) on oncogene cooperativity <strong>in</strong><br />

the malignant transformation of rodent cells. In 1985, he jo<strong>in</strong>ed Merck as a Genetic Toxicologist <strong>in</strong> the<br />

Department of Safety Assessment. He is currently a Senior Scientific Director <strong>in</strong> Genetic and Cellular<br />

Toxicology <strong>in</strong> the Department of Laboratory Sciences and Investigative Toxicology, MRL Safety<br />

Assessment. He is the Scientific Advisor to the Molecular Carc<strong>in</strong>ogenesis and DNA Damage lab group,<br />

with responsibility for <strong>in</strong> vitro and vivo genotoxicity assays for DNA damage and <strong>in</strong>vestigative approaches<br />

to understand<strong>in</strong>g mechanisms of chemical carc<strong>in</strong>ogenesis. His research <strong>in</strong>terests <strong>in</strong>clude the development<br />

of primary DNA damage assays (Comet and Alkal<strong>in</strong>e Elution) for genotoxicity assessment, understand<strong>in</strong>g<br />

the role of oncogene activation and tumor suppressor gene <strong>in</strong>activation <strong>in</strong> chemical carc<strong>in</strong>ogenesis,<br />

development of short-term carc<strong>in</strong>ogenicity bioassays <strong>in</strong> transgenic mice, epigenetic mechanisms <strong>in</strong><br />

tumorigenesis, and genetic susceptibility factors for development of hemangiosarcomas <strong>in</strong> mice. He was a<br />

member of the Steer<strong>in</strong>g Committee for the ILSI Health and Environmental Sciences Institute’s Alternatives<br />

to Carc<strong>in</strong>ogenicity Test<strong>in</strong>g Technical Committee from 1996 to 2003. From 2000 to 2004, he served as a<br />

member of the National Toxicology Program Board of Scientific Counselors, Technical Reports Review<br />

Subcommittee. His current responsibilities also <strong>in</strong>clude precl<strong>in</strong>ical compound management for compounds<br />

<strong>in</strong> development for Type II Diabetes.<br />

James A. Swenberg, D.V.M., Ph.D., DACVP, University of North Carol<strong>in</strong>a at Chapel Hill, Chapel Hill,<br />

NC, United States<br />

James Swenberg is a Kenan Dist<strong>in</strong>guished Professor of Environmental Sciences and Eng<strong>in</strong>eer<strong>in</strong>g and<br />

Professor of Nutrition, Pathology and Laboratory Medic<strong>in</strong>e at the University of North Carol<strong>in</strong>a at Chapel<br />

Hill. He also serves as the Director of the Center for Environmental Health and Susceptibility, the UNC<br />

Superfund Basic Research Program, and the Curriculum <strong>in</strong> Toxicology at the University of North Carol<strong>in</strong>a<br />

at Chapel Hill. His research focuses on mechanisms of carc<strong>in</strong>ogenesis and toxicology, with emphasis on<br />

the roles of DNA damage and repair and cell proliferation. He has published extensively on the use of<br />

mass spectrometry for DNA and prote<strong>in</strong> adducts aris<strong>in</strong>g from environmental and endogenous chemicals,<br />

<strong>in</strong>clud<strong>in</strong>g direct and <strong>in</strong>direct DNA damage aris<strong>in</strong>g from oxidative stress. Dr. Swenberg has published<br />

over 320 scientific papers. He was awarded the George Scott Award from the Toxicology Forum, the John<br />

Barnes Prize Lectureship from the British Toxicology Society, the Dist<strong>in</strong>guished Alumnus Award from<br />

The Ohio State University College of Veter<strong>in</strong>ary Medic<strong>in</strong>e, and the Dist<strong>in</strong>guished Research Alumnus Award<br />

from the University of M<strong>in</strong>nesota, College of Veter<strong>in</strong>ary Medic<strong>in</strong>e, and the Society of Toxicology Merit<br />

Award for 2007.<br />

Jan Willem van der Laan, Ph.D., National Institute of Public Health and the Environment (RIVM),<br />

Bilthoven, The Netherlands<br />

S<strong>in</strong>ce 1990, Dr. Jan Willem van der Laan is Senior Assessor of the Section of Pharmacology and<br />

Toxicology Assessment at the National Institute for Public Health and the Environment (RIVM) <strong>in</strong> The<br />

Netherlands. In this role, he is a member and Vice-Chairperson of the Safety Work<strong>in</strong>g Party of the CHMP.<br />

He has been (and still is) rapporteur for several topics <strong>in</strong> the toxicology area, such as Carc<strong>in</strong>ogenicity and<br />

Immunotoxicology. He was <strong>in</strong>volved <strong>in</strong> the ILSI Health and Environmental Sciences Institute <strong>in</strong>itiative<br />

on Alternatives to Carc<strong>in</strong>ogenicity Test<strong>in</strong>g, the project on transgenic mice stra<strong>in</strong>s <strong>in</strong> test<strong>in</strong>g. As a delegate<br />

from the CHMP-SWP, Dr. van der Laan was a participant <strong>in</strong> the ICH discussions on Carc<strong>in</strong>ogenicity, and<br />

Rapporteur of the ICH Expert Work<strong>in</strong>g Group on Immunotoxicity.<br />

23


notes<br />

24


speaker abstracts<br />

Sir Col<strong>in</strong> Berry<br />

Samuel M. Cohen<br />

Jon C. Cook<br />

Kay A. Criswell<br />

Vicki L. Dellarco<br />

Abigail C. Jacobs<br />

Timothy E. Johnson<br />

James E. Klaunig<br />

Dom<strong>in</strong>ique R. Lasserre-Bigot<br />

David E. Malarkey<br />

Keith L. March<br />

Jamie F. Modiano<br />

Michael S. Rogers<br />

James A. Swenberg<br />

Jan Willem van der Laan<br />

25<br />

session anD speaker presentations


speaker abstracts list<strong>in</strong>g <strong>in</strong> presentation order<br />

abstract name speaker<br />

Trivial to Catastrophic, Necessary to Deadly: Vascular Proliferations and<br />

Malignancies <strong>in</strong> Humans and Animals<br />

Sir Col<strong>in</strong> Berry<br />

Pathology of Treatment-Induced Hemangiosarcoma <strong>in</strong> NTP Studies David E. Malarkey<br />

The Effect of Genetic Diversity on Angiogenesis Michael S. Rogers<br />

Bark<strong>in</strong>g Up the Right Tree: Uncover<strong>in</strong>g the Influence of Heritable<br />

Factors on Can<strong>in</strong>e Hemangiosarcoma<br />

Jamie F. Modiano<br />

Us<strong>in</strong>g Mode-of-Action Data to Assess the Human Relevance of Animal<br />

Tumors<br />

Vicki L. Dellarco<br />

Mode of Action of V<strong>in</strong>yl Chloride-Induced Hepatic Hemangiosarcoma James A. Swenberg<br />

Vascular Tumor Potential of Carbaryl <strong>in</strong> the Heterozygous p53<br />

Knockout Mouse Model<br />

Dom<strong>in</strong>ique R. Lasserre-Bigot<br />

Nongenotoxic Agents: Studies with 2-Butoxyethanol James E. Klaunig<br />

Angiogenesis and Adipogenesis Keith L. March<br />

Troglitazone: An Illustration of the HESI Mode-of-Action Framework<br />

for PPAR Gamma-Induced Hemangiosarcoma<br />

Jon C. Cook<br />

Troglitazone Effects on Endothelial Cells In Vivo and In Vitro:<br />

Differences between Mice and Humans<br />

Samuel M. Cohen<br />

Ret<strong>in</strong>oid-Induced Hemangiosarcoma <strong>in</strong> Mice: Potential Insight from<br />

In Vivo and In Vitro Studies<br />

Timothy E. Johnson<br />

Epigenetic Mode of Action Associated with Induction of<br />

Hemangiosarcoma <strong>in</strong> Mice Treated with Pregabal<strong>in</strong><br />

Kay A. Criswell<br />

EPA Perspectives Vicki L. Dellarco<br />

Hemangiosarcomas and Pharmaceuticals: An FDA Perspective Abigail C. Jacobs<br />

European Perspectives on Carc<strong>in</strong>ogenicity Test<strong>in</strong>g Jan Willem van der Laan<br />

26


speaker abstracts list<strong>in</strong>g <strong>in</strong> alpha order by speaker<br />

speaker abstract name<br />

Sir Col<strong>in</strong> Berry Trivial to Catastrophic, Necessary to Deadly: Vascular Proliferations<br />

and Malignancies <strong>in</strong> Humans and Animals<br />

Samuel M. Cohen Troglitazone Effects on Endothelial Cells In Vivo and In Vitro:<br />

Differences between Mice and Humans<br />

Jon C. Cook Troglitazone: An Illustration of the HESI Mode-of-Action Framework<br />

for PPAR Gamma-Induced Hemangiosarcoma<br />

Kay A. Criswell Epigenetic Mode of Action Associated with Induction of<br />

Hemangiosarcoma <strong>in</strong> Mice Treated with Pregabal<strong>in</strong><br />

Vicki L. Dellarco Us<strong>in</strong>g Mode-of-Action Data to Assess the Human Relevance of Animal<br />

Tumors<br />

Vicki L. Dellarco EPA Perspectives<br />

Abigail C. Jacobs Hemangiosarcomas and Pharmaceuticals: An FDA Perspective<br />

Timothy E. Johnson Ret<strong>in</strong>oid-Induced Hemangiosarcoma <strong>in</strong> Mice: Potential Insight from<br />

In Vivo and In Vitro Studies<br />

James E. Klaunig Nongenotoxic Agents: Studies with 2-Butoxyethanol<br />

Dom<strong>in</strong>ique R. Lasserre-Bigot Vascular Tumor Potential of Carbaryl <strong>in</strong> the Heterozygous p53<br />

Knockout Mouse Model<br />

David E. Malarkey Pathology of Treatment-Induced Hemangiosarcoma <strong>in</strong> NTP Studies<br />

Keith L. March Angiogenesis and Adipogenesis<br />

Jamie F. Modiano Bark<strong>in</strong>g Up the Right Tree: Uncover<strong>in</strong>g the Influence of Heritable<br />

Factors on Can<strong>in</strong>e Hemangiosarcoma<br />

Michael S. Rogers The Effect of Genetic Diversity on Angiogenesis<br />

James A. Swenberg Mode of Action of V<strong>in</strong>yl Chloride-Induced Hepatic Hemangiosarcoma<br />

Jan Willem van der Laan European Perspectives on Carc<strong>in</strong>ogenicity Test<strong>in</strong>g<br />

27


Day 1<br />

thursDay, DeCember 4, 2008<br />

session tWo: pathology anD biology<br />

In the first session on Pathology and Biology, the spectrum of spontaneous and chemically-<strong>in</strong>duced<br />

benign and malignant vascular neoplasms <strong>in</strong> rodents and humans will be reviewed <strong>in</strong> order to highlight<br />

the important species differences <strong>in</strong> <strong>in</strong>cidence, tissue distribution, tumor pathology and susceptibility to<br />

tumorigenesis. Hemangiosarcoma is rare <strong>in</strong> humans and relatively common <strong>in</strong> domestic and experimental<br />

animals. Established causes <strong>in</strong> humans (Thorotrast, v<strong>in</strong>yl chloride, HHV-8 <strong>in</strong>fection and hereditary<br />

syndromes) differ widely <strong>in</strong> pathogenetic mechanisms. In addition, there are a number of syndromes <strong>in</strong><br />

man <strong>in</strong> which abnormalities of the vasculature and neoplasia are both significant features and where an<br />

<strong>in</strong>terrelationship between the changes lead<strong>in</strong>g to tumour formation <strong>in</strong> both epithelia and the vasculature<br />

are common and apparently genetically determ<strong>in</strong>ed. The relationship of sarcomas to other proliferative<br />

vascular changes may depend on a limited number of disturbances <strong>in</strong> the genetic control of physiologically<br />

essential angiogenic stimuli. These changes will be discussed and their relationships with proliferative and<br />

cytok<strong>in</strong>e related stimuli considered. It is clear that a number of different pathways to angiosarcoma exist<br />

and that some assumptions that have been made about pathogenesis are unfounded. Which are the relevant<br />

animal models and do they <strong>in</strong>form us about human risk assessments? Hemangiosarcoma spontaneously<br />

occurs <strong>in</strong> about 0.1–2% of rats and 2–5% of mice. More than twenty agents studied by the National<br />

Toxicology Program (NTP) have been associated with <strong>in</strong>duction of vascular neoplasms, a few caus<strong>in</strong>g<br />

hemangiosarcoma <strong>in</strong>cidences of > 80%. The comparative <strong>in</strong>cidences and pathology of agent-<strong>in</strong>duced<br />

hemangiosarcomas <strong>in</strong> rodents from NTP and other studies will be reviewed.<br />

S<strong>in</strong>ce vascular neoplasms are assumed to reflect aberrations <strong>in</strong> growth control of cells that are essential<br />

participants <strong>in</strong> normal vasculogenesis and angiogenesis, an overview of our current understand<strong>in</strong>g of the<br />

cell types and processes <strong>in</strong>volved <strong>in</strong> normal vessel development will be presented. Angiogenesis is an<br />

essential part of many cont<strong>in</strong>uously runn<strong>in</strong>g processes, <strong>in</strong>clud<strong>in</strong>g wound heal<strong>in</strong>g and repair and endometrial<br />

cycl<strong>in</strong>g. It is ma<strong>in</strong>ly an adaptive response to local hypoxia and is dependent on the local accumulation<br />

of hypoxia <strong>in</strong>ducible factors (HIF’s), transcription factors that can activate a number of angiogenic<br />

genes. Recent advances <strong>in</strong> our understand<strong>in</strong>g of tumor angiogenesis have highlight the important roles of<br />

circulat<strong>in</strong>g, bone marrow-derived progenitor cells <strong>in</strong>clud<strong>in</strong>g both endothelial and monocyte/macrophage<br />

l<strong>in</strong>eage cells. These f<strong>in</strong>d<strong>in</strong>gs suggest that hemangiosarcomas may arise not only from transformation<br />

of tissue-resident endothelial cell populations but also from circulat<strong>in</strong>g progenitors or adult stem cells<br />

recruited from bone marrow or possibly also from extramedullary sites of hematopoiesis such as the<br />

liver and spleen. The coord<strong>in</strong>ation of the recruitment, proliferation, localization, and differentiation of<br />

the different cell types that regulate angiogenic processes is exquisitely complex <strong>in</strong>volv<strong>in</strong>g hundreds of<br />

genes and offers a multitude of opportunities for genetic diversity (polymorphisms) to <strong>in</strong>troduce species,<br />

stra<strong>in</strong> and <strong>in</strong>dividual animal variation <strong>in</strong> angiogenic responses. The genetic diversity <strong>in</strong> angiogenesisregulat<strong>in</strong>g<br />

genes has been l<strong>in</strong>ked to <strong>in</strong>creased susceptibility to multiple angiogenesis-dependent<br />

diseases <strong>in</strong> humans <strong>in</strong>clud<strong>in</strong>g cancer. Ongo<strong>in</strong>g mapp<strong>in</strong>g studies have identified multiple loci that control<br />

angiogenic responsiveness <strong>in</strong> several mouse models rais<strong>in</strong>g the possibility that this approach may<br />

ultimately reveal genetic loci <strong>in</strong> mice that modulate susceptibility not only to angiogenic responsiveness<br />

but also to spontaneous and/or chemically-<strong>in</strong>duced hemangiosarcomas. Similarly, research on can<strong>in</strong>e<br />

hemangiosarcoma has tested the hypothesis that genetic background (def<strong>in</strong>ed as “breed”), <strong>in</strong>fluences<br />

phenotypes and behavior of sporadic tumors. Results of this work have shown that tumor-restricted<br />

expression of pro-<strong>in</strong>flammatory and angiogenic genes clusters hemangiosarcoma of Golden Retrievers<br />

separately from hemangiosarcoma of non-Golden Retrievers, suggest<strong>in</strong>g heritable factors mold phenotypes<br />

<strong>in</strong> sporadic, naturally occurr<strong>in</strong>g tumors. Together these data suggest that understand<strong>in</strong>g the mode-of-action<br />

and human relevance of chemically-<strong>in</strong>duced hemangiosarcomas <strong>in</strong> rodents will need to consider not<br />

only the dose response relationships and chemical mode-of-action but also the <strong>in</strong>tr<strong>in</strong>sic sensitivity and<br />

susceptibility of different rodent test species and stra<strong>in</strong>s and the genetic basis for these species differences.<br />

28


session two speaker abstracts:<br />

trivial to Catastrophic, necessary to Deadly: Vascular proliferations<br />

and malignancies <strong>in</strong> humans and animals<br />

Sir Col<strong>in</strong> Berry 1<br />

1 Queen Mary, University of London, London, United K<strong>in</strong>gdom<br />

Angiogenesis is an essential part of many cont<strong>in</strong>uously runn<strong>in</strong>g processes, <strong>in</strong>clud<strong>in</strong>g wound heal<strong>in</strong>g<br />

and repair and endometrial cycl<strong>in</strong>g. It is ma<strong>in</strong>ly an adaptive response to local hypoxia and is dependent<br />

on the local accumulation of hypoxia <strong>in</strong>ducible factors (HIF’s) which are composed of alpha and<br />

beta sub-units of a heterodimeric transcription factor. These are normally cont<strong>in</strong>uously degraded by<br />

oxygen dependent processes but <strong>in</strong> the absence of adequate oxygen levels the alpha sub-units are<br />

stabilized and form heterodimers with HIF beta which is not oxygen sensitive. These transcription<br />

factors then activate a number of angiogenic genes. There are a number of syndromes <strong>in</strong> Man <strong>in</strong><br />

which abnormalities of the vasculature and neoplasia are both significant features and where an<br />

<strong>in</strong>terrelationship between the changes lead<strong>in</strong>g to tumor formation <strong>in</strong> both epithelia and the vasculature<br />

are common and apparently genetically determ<strong>in</strong>ed. However, it is clear that a number of different<br />

pathways to angiosarcoma exist and that some assumptions that have been made about pathogenesis<br />

are unfounded. Which are the relevant models and do they <strong>in</strong>form us about human risk assessments?<br />

pathology of treatment-<strong>in</strong>duced hemangiosarcoma <strong>in</strong> ntp studies<br />

David E. Malarkey 1<br />

1 NIEHS National Toxicology Program, Research Triangle Park, NC, United States<br />

Hemangiosarcoma (also known as angiosarcoma or malignant hemangioendothelioma) is a<br />

highly aggressive and often lethal malignant neoplasm that arises from vascular endothelium.<br />

Hemangiosarcomas (HSA) are generally uncommon and have been reported <strong>in</strong> most species, <strong>in</strong>clud<strong>in</strong>g<br />

man, non-human primates, dogs, cats, mice, and rats. The neoplasm can arise <strong>in</strong> any organ but is more<br />

commonly found <strong>in</strong> heart (right atrium), liver, spleen, lung, sk<strong>in</strong>, soft tissues, mammary gland, and/<br />

or bone. Local <strong>in</strong>vasion and metastasis are common with metastasis frequently <strong>in</strong>volv<strong>in</strong>g the lung and<br />

liver. Metastatic lesions can be difficult to dist<strong>in</strong>guish from multicentrically aris<strong>in</strong>g HSA. There is<br />

some evidence that benign lesions, such as hemangioma, may rarely progress to malignancy. HSA has<br />

been reported spontaneously <strong>in</strong> CBA/J, CD1, BalbC, and B6C3F1 mice and average <strong>in</strong>cidences range<br />

from < 1% up to about 6 %, respectively, with slightly lower <strong>in</strong>cidences <strong>in</strong> female mice. F344 and<br />

Sprague Dawley rats have spontaneous <strong>in</strong>cidences of 0.4% and 1.6%, respectively. Risk factors <strong>in</strong> man<br />

and/or animals <strong>in</strong>clude exposure to chemicals such as v<strong>in</strong>yl chloride, arsenic, peroxisome proliferatoractivated<br />

receptor (PPAR) agonists, and thorium dioxide; hemochromatosis; radiation therapy;<br />

solar irradiation; and heritable defects. Twenty six agents studied by the U.S. National Toxicology<br />

Program (NTP) between 1978–2008 have been associated with the <strong>in</strong>duction of hemangioma and/or<br />

hemangiosarcoma, represent<strong>in</strong>g 9% (26/292) of rodent carc<strong>in</strong>ogens identified by the NTP <strong>in</strong> 2 year<br />

studies. p53 and K-ras alterations have been demonstrated <strong>in</strong> rodent hemangiosarcomas with chemicalspecific<br />

mutations identified <strong>in</strong> neoplasms <strong>in</strong>duced by o-nitrotoluene, riddelli<strong>in</strong>e, butadiene, and v<strong>in</strong>yl<br />

chloride. Studies focus<strong>in</strong>g on markers of <strong>in</strong>volution of vascular anomalies and benign neoplasms are<br />

beg<strong>in</strong>n<strong>in</strong>g to reveal angiogenic and angio<strong>in</strong>hibitory pathways for HSA growth driven by cytok<strong>in</strong>es such<br />

as bFGF, PIGF, VEGF, and angiopoiet<strong>in</strong>s. Understand<strong>in</strong>g the comparative histological, biological, and<br />

molecular aspects of spontaneous and chemically <strong>in</strong>duced endothelial tumors offers new <strong>in</strong>sights <strong>in</strong>to<br />

the pathogenesis and relevance of animal models <strong>in</strong> risk assessment.<br />

29<br />

(Cont<strong>in</strong>ued on next page)


the effect of genetic Diversity on angiogenesis<br />

Michael S. Rogers 1<br />

1 Harvard University, Boston, MA, United States<br />

Angiogenesis is the process by which new blood vessels are formed from exist<strong>in</strong>g vessels. The process<br />

is normally tightly regulated, however dysregulation of angiogenesis is observed <strong>in</strong> a number of<br />

pathologies, <strong>in</strong>clud<strong>in</strong>g cancer. Mammalian populations, <strong>in</strong>clud<strong>in</strong>g humans and mice, harbor genetic<br />

variations that alter angiogenesis. In humans, these changes can result <strong>in</strong> Mendelian traits of variable<br />

penetrance, with telangiectasia be<strong>in</strong>g a common symptom. Other, more common, variations exist, with<br />

promoter variations <strong>in</strong> the VEGF gene be<strong>in</strong>g particularly commonly studied. Angiogenesis-regulat<strong>in</strong>g<br />

gene variants can result <strong>in</strong> <strong>in</strong>creased susceptibility to multiple angiogenesis-dependant diseases <strong>in</strong><br />

humans. Our efforts to dissect the complexity of the genetic diversity that regulates angiogenesis have<br />

used laboratory animals due to the availability of genome sequence for many species and the ability to<br />

perform high volume controlled breed<strong>in</strong>g. Ongo<strong>in</strong>g mapp<strong>in</strong>g studies have identified multiple loci that<br />

control angiogenic responsiveness <strong>in</strong> several mouse models.<br />

Bark<strong>in</strong>g Up the Right Tree: Uncover<strong>in</strong>g the Influence of Heritable<br />

factors on Can<strong>in</strong>e hemangiosarcoma<br />

Jaime F. Modiano 1<br />

1 University of M<strong>in</strong>nesota, M<strong>in</strong>neapolis, MN, United States<br />

The role an <strong>in</strong>dividual’s genetic background plays on phenotype and biological behavior of sporadic<br />

tumors rema<strong>in</strong>s <strong>in</strong>completely understood. We showed previously that lymphomas from Golden<br />

Retrievers harbor def<strong>in</strong>ed, recurrent chromosomal aberrations that occur less frequently <strong>in</strong> lymphomas<br />

from other dog breeds suggest<strong>in</strong>g spontaneous can<strong>in</strong>e tumors provide suitable models to def<strong>in</strong>e how<br />

heritable traits <strong>in</strong>fluence cancer genotypes. Here, we report a complementary approach exam<strong>in</strong><strong>in</strong>g<br />

gene expression profiles <strong>in</strong> a naturally occurr<strong>in</strong>g soft tissue sarcoma (hemangiosarcoma) from Golden<br />

Retrievers compared to other dog breeds (non-Golden Retrievers). Gene expression signatures show<br />

naturally occurr<strong>in</strong>g Golden Retriever tumors clustered separately from non-Golden Retriever tumors<br />

with contributions from pro-<strong>in</strong>flammatory and angiogenic genes that were not due simply to breed<br />

haplotypes. Moreover, genes <strong>in</strong>volved <strong>in</strong> angiogenesis, immune response and survival, <strong>in</strong>clud<strong>in</strong>g<br />

VEGF receptors and acid ceramidase, showed expression patterns that segregated both by breed and<br />

tumor type. Biological responses mediated through VEGF receptors reflected the breed-dependent<br />

dichotomy <strong>in</strong> expression. Our results illustrate how heritable factors mold gene expression phenotypes<br />

<strong>in</strong> sporadic, naturally occurr<strong>in</strong>g tumors, and provide <strong>in</strong>sights <strong>in</strong>to the ontogeny and multipotential of<br />

tumor-<strong>in</strong>itiat<strong>in</strong>g cells.<br />

30


session three: hemangiosarComa <strong>in</strong>DuCeD by<br />

non-pharmaCeutiCals<br />

The third session will exam<strong>in</strong>e the <strong>in</strong>duction of hemangiosarcomas by non-pharmaceuticals agents.<br />

There are a number of examples <strong>in</strong> the scientific literature show<strong>in</strong>g the <strong>in</strong>duction of hemangiosarcomas<br />

by non-pharmaceuticals <strong>in</strong> rodent species. The mode-of-action by which these agents <strong>in</strong>duce<br />

hemangiosarcomas <strong>in</strong> rodents has proven to be through both DNA reactive as well as non-genotoxic<br />

mechanisms. V<strong>in</strong>yl chloride, an <strong>in</strong>ducer of hemangiosarcomas <strong>in</strong> rodents and humans, appears to function<br />

through genotoxic mechanisms. The mode-of-action <strong>in</strong>volves the formation of genomic DNA adducts. In<br />

rodent studies, molecular dosimetry of DNA adducts <strong>in</strong> the liver has been exam<strong>in</strong>ed after v<strong>in</strong>yl chloride<br />

treatment and showed that the 7-(2’-Oxoethyl)guan<strong>in</strong>e (7OEG) was the major DNA adduct detected (98%<br />

of adducts). The N 2 , 3-Ethenoguan<strong>in</strong>e and 3, N 4 -etheno-2’-deoxycytid<strong>in</strong>e were also found (1% of adducts).<br />

N 6 -etheno-2’-deoxyadenos<strong>in</strong>e was present at even lower concentrations. The persistence of these four<br />

adducts showed that while the 7OEG adduct had a ½ life of 62 h, the other three etheno adducts were highly<br />

persistent, hav<strong>in</strong>g ½ lives of 30 days or more. A number of nongenotoxic compounds have also been shown<br />

to <strong>in</strong>duce hemangiosarcomas particularly <strong>in</strong> the mouse. The compound 2-butoxyethanol has been reported<br />

to <strong>in</strong>crease hemangiosarcomas follow<strong>in</strong>g chronic <strong>in</strong>halation exposure. The <strong>in</strong>duction of hemangiosarcomas<br />

<strong>in</strong> mice appears to be related to the <strong>in</strong>duction of oxidative damage secondary to hemolysis <strong>in</strong>duced by the<br />

2-butoxyethanol. This compound is not DNA reactive and appears to be function<strong>in</strong>g through an <strong>in</strong>direct<br />

mechanism by which endothelial cells undergo cell proliferation <strong>in</strong> the mouse. In addition, more recent data<br />

suggests a possible role for HIF-1 alpha and VEGF <strong>in</strong> the carc<strong>in</strong>ogenesis process follow<strong>in</strong>g butoxyethanol<br />

exposure. In addition, Carbaryl which <strong>in</strong>duces hemangiosarcomas <strong>in</strong> the mouse <strong>in</strong> a dose responsive<br />

manner also appears to produce its carc<strong>in</strong>ogenic effects through nongenotoxic mechanisms. Studies have<br />

shown a demonstrable threshold for Carbaryl <strong>in</strong>duction of vascular tumors <strong>in</strong> the CD-1 mouse. Carbaryl<br />

failed to <strong>in</strong>duce hemangiosarcomas or other tumors at doses up 4000 ppm <strong>in</strong> heterozygous p53 knockout<br />

mice. This suggests a nongenotoxic mode-of-action for Carbaryl. Understand<strong>in</strong>g the mode-of-action by<br />

which hemangiosarcomas are produced <strong>in</strong> rodents is important <strong>in</strong> def<strong>in</strong><strong>in</strong>g the potential human health<br />

consequence of exposure to non-pharmaceuticals that can produce this vascular tumor. Us<strong>in</strong>g the weight of<br />

evidence approach for evaluat<strong>in</strong>g a mode-of-action of agents that produce hemangiosarcomas is extremely<br />

valuable to access species differences as well as to understand threshold and dose response characteristics<br />

of hemangiosarcoma formation.<br />

session three speaker abstracts:<br />

us<strong>in</strong>g mode-of-action Data to assess the human relevance of animal<br />

tumors<br />

Vicki L. Dellarco 1<br />

1 Office of Pesticide Programs, U.S. Environmental Protection Agency, Wash<strong>in</strong>gton, DC, United States<br />

Extrapolations or <strong>in</strong>ference methods are necessary when us<strong>in</strong>g the results of the rodent cancer bioassay<br />

to predict human health consequences. Unless there is evidence to the contrary, it is assumed, for<br />

example, that rodent data predict human effects and f<strong>in</strong>d<strong>in</strong>gs at high experimental doses predict effects<br />

at environmental exposure levels. The consideration of mode-of-action is extremely valuable to assess<br />

species differences and to <strong>in</strong>form dose-response extrapolation and model<strong>in</strong>g approaches. An approach<br />

will be presented that provides rigor and transparency to the weight of evidence evaluation of mode-ofaction<br />

has developed through collaborative efforts of the U.S. FDA, Health Canada, International<br />

Programme for Chemical Safety (IPCS), and the International Life Sciences Institute (ILSI).<br />

(This abstract represents the view of the author and does not necessarily represent the decisions or stated policies<br />

of the U.S. EPA. Mention of trade names does not imply endorsement.)<br />

31<br />

(Cont<strong>in</strong>ued on next page)


mode of action of V<strong>in</strong>yl Chloride-<strong>in</strong>duced hepatic hemangiosarcoma<br />

James A. Swenberg 1, 2, 3 , Esra Mutlu 1, 2 , L<strong>in</strong>a Gao 1 , Eric Mor<strong>in</strong>ello 2 , Amy Ham 3 , Leonard Coll<strong>in</strong>s 1 ,<br />

Gunnar Boysen 1 , Patricia Upton 1 , Darrell W<strong>in</strong>sett 4 , and Gary Hatch 4<br />

1 Department of Environmental Sciences, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

2 The Curriculum <strong>in</strong> Toxicology, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

3 Pathology and Laboratory Medic<strong>in</strong>e, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

4 U.S. EPA NHEERL, Research Triangle Park, NC, United States<br />

V<strong>in</strong>yl chloride (VC) is a known animal and human carc<strong>in</strong>ogen that <strong>in</strong>duces hemangiosarcoma of the<br />

liver as its hallmark cancer follow<strong>in</strong>g <strong>in</strong>halation exposure. In humans and animals, VC carc<strong>in</strong>ogenesis<br />

is associated with relatively high exposures (≥50 ppm). VC is metabolized primarily by CYP2E1 to<br />

the epoxide, chloroethylene oxide (CEO), a highly chemical unstable chemical that reacts with local<br />

cellular DNA and prote<strong>in</strong>s and is thought to have m<strong>in</strong>imal systemic exposure. Four DNA adducts<br />

have been identified and have had molecular dosimetry studies conducted. The major DNA adduct is<br />

7-(2-oxoethyl)guan<strong>in</strong>e (OEG), which comprises ~98% of the DNA adducts. This adduct is lost from<br />

DNA primarily by chemical depur<strong>in</strong>ation and is not considered to be promutagenic. Three etheno<br />

adducts are also formed and are thought to be promutagenic. All four of the DNA adducts of VC<br />

exhibit supral<strong>in</strong>ear exposure response curves due to saturation of metabolic activation. In addition, we<br />

have recently shown that all four adducts are formed endogenously from lipid peroxidation products.<br />

Thus, there is always a background of identical endogenous adducts present. By expos<strong>in</strong>g rats to<br />

[ 13 C 2 ]-VC, we have been able to accurately determ<strong>in</strong>e the half-lives of all four of the VC-<strong>in</strong>duced<br />

adducts. N 2 ,3-Ethenoguan<strong>in</strong>e (εG) has been shown to have a half-life of ~150 days, suggest<strong>in</strong>g<br />

that it is very poorly repaired, if at all. In contrast, 1,N 6 -ethenodeoxyadenos<strong>in</strong>e (εdA) and 3N 4<br />

ethenodeoxycytid<strong>in</strong>e are actively repaired and have half-lives of ~ 1 day, while OEG has a half-life<br />

of ~4 days as a result of chemical depur<strong>in</strong>ation. Hepatic hemangiosarcomas <strong>in</strong> animals and humans<br />

conta<strong>in</strong> mutations <strong>in</strong> the p53 tumor suppressor gene and ras oncogenes that are compatible with the<br />

mutations caused by the three etheno adducts. The Mode of Action for VC fits well with a mutagenic<br />

MOA. What is less clear is how risk extrapolates at low exposures, s<strong>in</strong>ce identical endogenous DNA<br />

adducts are present <strong>in</strong> unexposed animals and humans.<br />

Vascular tumor potential of Carbaryl <strong>in</strong> the heterozygous p53<br />

knockout mouse model<br />

Dom<strong>in</strong>ique R. Lasserre-Bigot 1 , Eric L. Debruyne 1 , and Neil G. Carmichael 2<br />

1 Bayer CropScience SA, Sophia-Antipolis Cedex France<br />

2 ECETOC AISBL, Brussels, Belgium<br />

In the mouse 2-year carc<strong>in</strong>ogenicity study where 80 CD-1 mice per group were given diets provid<strong>in</strong>g<br />

0, 100, 1000 and 8000 ppm carbaryl, there was an <strong>in</strong>crease of liver tumours <strong>in</strong> females and kidney<br />

tumors <strong>in</strong> males at 8000 ppm (dose exceed<strong>in</strong>g the Maximum Tolerated Dose) and also an <strong>in</strong>creased<br />

<strong>in</strong>cidence of vascular tumors <strong>in</strong> both sexes at 8000 ppm. An <strong>in</strong>crease of vascular tumors (statistically<br />

significant) was still observed <strong>in</strong> male mice at the mid-dose (1000 ppm). At the low dose of 100 ppm,<br />

the <strong>in</strong>cidence of vascular tumors <strong>in</strong> male mice was <strong>in</strong>creased compared to the control but without<br />

statistical significance; no clear dose-effect relationship was obta<strong>in</strong>ed. In previous studies to assess its<br />

genotoxic potential, carbaryl had shown potential clastogenic activity <strong>in</strong> an <strong>in</strong> vitro CHO assay with<br />

S9 activation, but was negative <strong>in</strong> an <strong>in</strong> vivo chromosomal aberration test as well as <strong>in</strong> all other tests<br />

of the genotoxicity battery. However, mouse metabolism data with carbaryl had suggested the possible<br />

32


formation of epoxide metabolites. To <strong>in</strong>vestigate the possible implication of a genotoxic mechanism<br />

<strong>in</strong> the <strong>in</strong>duction of the vascular tumors observed with carbaryl <strong>in</strong> CD-1 mice, carbaryl was tested <strong>in</strong><br />

the p53 heterozygous knockout mouse model. Carbaryl was adm<strong>in</strong>istered cont<strong>in</strong>uously via the diet<br />

to groups of 20 male heterozygous p53 knockout mice at concentrations of 0, 10, 30, 100, 300, 1000<br />

and 4000 ppm for 6 months. Histopathological exam<strong>in</strong>ations revealed no evidence of carbaryl <strong>in</strong>duced<br />

neoplasms of any type, especially no neoplastic or preneoplastic changes were noted <strong>in</strong> the vascular<br />

tissue of any of the organs exam<strong>in</strong>ed. Therefore, under the conditions of this study, the No Observed<br />

Effect Level of carbaryl was 4000 ppm (around 716 mg/kg/day) for neoplastic changes. These results<br />

were not due to the lack of sensitivity of the model, s<strong>in</strong>ce we demonstrated <strong>in</strong> a previous study the<br />

sensitivity of the model for the <strong>in</strong>duction of vascular tumors us<strong>in</strong>g urethane, as positive control. The<br />

results obta<strong>in</strong>ed so far with a number of chemicals <strong>in</strong>dicate a good correlation <strong>in</strong> the carc<strong>in</strong>ogenic<br />

response between the p53 knockout mouse model and the conventional mouse bioassay. It appears<br />

that most genotoxic chemicals can be detected <strong>in</strong> this p53 knockout mouse model. Furthermore, it<br />

was demonstrated <strong>in</strong> an <strong>in</strong> vivo mouse b<strong>in</strong>d<strong>in</strong>g assay to prote<strong>in</strong> and DNA that carbaryl did not <strong>in</strong>teract<br />

and b<strong>in</strong>d covalently to DNA or chromatid prote<strong>in</strong>s <strong>in</strong> the liver when adm<strong>in</strong>istered orally to mice.<br />

In conclusion, Carbaryl does not appear to be genotoxic <strong>in</strong> vivo and therefore the vascular tumors<br />

observed <strong>in</strong> CD-1 mice were not formed through a genotoxic mechanism.<br />

nongenotoxic agents: studies with 2-butoxyethanol<br />

James E. Klaunig 1 , Lisa M. Kamendulis 1 , and Stacey Corthals 1<br />

1<br />

Center for Environmental Health, Department of Pharmacology and Toxicology, Indiana University<br />

School of Medic<strong>in</strong>e, Indianapolis, IN, United States<br />

2-Butoxyethanol (BE) has been reported to <strong>in</strong>duce an <strong>in</strong>crease <strong>in</strong> liver hemangiosarcomas <strong>in</strong> male<br />

B6C3F1 mice follow<strong>in</strong>g chronic <strong>in</strong>halation, but not <strong>in</strong> female mice or either gender <strong>in</strong> rats. The<br />

mechanisms <strong>in</strong>volved <strong>in</strong> BE-<strong>in</strong>duced hemangiosarcoma formation are not clear, but data from our<br />

group and others suggests the <strong>in</strong>volvement of oxidative damage subsequent to red blood cell hemolysis<br />

and iron deposition, and activation of Kupffer cells <strong>in</strong> the liver, events that exhibit a threshold <strong>in</strong><br />

both animals and humans. In isolated mouse and rat hepatocytes, neither BE or its major metabolite,<br />

2-butoxyacetic acid (BAA), <strong>in</strong>creased oxidative DNA damage (OH8dG), lipid peroxidation (MDA<br />

formation) or decreased vitam<strong>in</strong> E concentrations, while both ferrous sulfate (iron) and hemolyzed<br />

RBCs produced dose-related changes <strong>in</strong> biomarkers of oxidative stress. Comparatively, mouse<br />

hepatocytes were more sensitive to oxidative stress by iron and hemolyzed RBC compared with the<br />

rat. In the Syrian Hamster Embryo (SHE) cell transformation assay, BE and BAA acid did not <strong>in</strong>duce<br />

cellular transformation. In contrast, iron produced dose-related <strong>in</strong>creases <strong>in</strong> cell transformation,<br />

OH8dG and DNA damage—effects that were prevented by co-exposure to antioxidants (vitam<strong>in</strong> E<br />

or EGCG). In cultured endothelial cells, BE, BAA and the aldehyde <strong>in</strong>termediate (BALD) did not<br />

<strong>in</strong>duce DNA damage (COMET). However, iron, hemolyzed RBC and hydrogen peroxide <strong>in</strong>creased<br />

DNA damage <strong>in</strong> endothelial cells. Additional studies showed that activated macrophages <strong>in</strong>creased<br />

both endothelial cell DNA damage and DNA synthesis, suggest<strong>in</strong>g a role for macrophage activation<br />

<strong>in</strong> <strong>in</strong>creased cell proliferation <strong>in</strong> endothelial cells. Complimentary <strong>in</strong> vivo studies have also been<br />

performed. In a sub-chronic study <strong>in</strong> mice and rats, BE <strong>in</strong>duced hemolysis <strong>in</strong> both species, at all<br />

timepo<strong>in</strong>ts (7, 14, 28, 90 days) and doses (225 and 450 mg/kg [rats]; 225, 450, and 900 mg/kg [mice]).<br />

Evidence of hemolysis was also shown by an <strong>in</strong>crease <strong>in</strong> iron-sta<strong>in</strong>ed Kupffer cells <strong>in</strong> both species,<br />

at the 2 highest doses of BE. Increases <strong>in</strong> OH8dG and MDA were biphasic and species selective;<br />

<strong>in</strong>creases were seen after 7 and 90 days, selectively <strong>in</strong> mouse liver. BE also produced a biphasic and<br />

species selective <strong>in</strong>duction of DNA synthesis <strong>in</strong> mouse liver; endothelial cell DNA synthesis <strong>in</strong>creased<br />

early after BE exposure (7 and 14 days) while DNA synthesis <strong>in</strong> hepatocytes <strong>in</strong>creased only at 90<br />

33<br />

(Cont<strong>in</strong>ued on next page)


days. To assess the role of Kupffer cells <strong>in</strong> BE-<strong>in</strong>duced endothelial cell DNA synthesis, mice were<br />

given BE <strong>in</strong> the presence or absence of Kupffer cell depletion (via clodronate-encapsulated liposomes;<br />

CL) for 7 days. BE <strong>in</strong>creased the number of Kupffer cells (F4/80+ cells), while CL decreased Kupffer<br />

cell number by >90%. BE produced the anticipated effects; <strong>in</strong>creased hemolysis, and iron-sta<strong>in</strong>ed<br />

nonparenchymal cells, and <strong>in</strong>creased endothelial cell DNA synthesis. However, all values were similar<br />

to control <strong>in</strong> Kupffer cell depleted mice, suggest<strong>in</strong>g that this cell type participates <strong>in</strong> BE-<strong>in</strong>duced<br />

endothelial cell DNA synthesis. Collectively, evidence from <strong>in</strong> vitro and <strong>in</strong> vivo show that BE does<br />

not elicit direct effects on the liver, and provides suggestive evidence that the mode-of-action for<br />

BE-<strong>in</strong>duced liver hemangiosarcomas <strong>in</strong>volves <strong>in</strong>direct effects <strong>in</strong>clud<strong>in</strong>g Kupffer cell modulation and<br />

oxidative stress.<br />

Day 2<br />

friDay, DeCember 5, 2008<br />

session four: hemangiosarComa <strong>in</strong>DuCeD by ppar<br />

agonists—hesi <strong>in</strong>itiatiVe<br />

A unify<strong>in</strong>g nongenotoxic mode of action (MOA) for the myriad of compounds that <strong>in</strong>duce<br />

hemangiosarcoma has not been articulated. To date, 2-butoxyethanol (2-BE) is the most comprehensive<br />

nongenotoxic MOA for hemangiosarcoma-<strong>in</strong>duction <strong>in</strong> mice whereby 2-BE produces hemolysis lead<strong>in</strong>g to<br />

iron accumulation with<strong>in</strong> Kupffer cells result<strong>in</strong>g <strong>in</strong> oxidative damage and release of growth factors from<br />

the Kupffer cells. However, this MOA does not appear to be universally applicable to other compounds.<br />

In 2005, the HESI PPAR Agonist Project Committee was formed by a group of pharmaceutical companies<br />

to advance research on and understand<strong>in</strong>g of the MOA and human relevance of PPAR-<strong>in</strong>duced tumors<br />

by gamma (γ) and dual gamma/alpha (γ/α) agonists: hemangiosarcoma <strong>in</strong> mice, fibrosarcoma <strong>in</strong> rats, and<br />

bladder lesions <strong>in</strong> animal models. A MOA framework has been developed for hemangiosarcoma based<br />

on collective data shar<strong>in</strong>g by the companies engaged <strong>in</strong> the Project Committee at a meet<strong>in</strong>g <strong>in</strong> August<br />

2007. Because this MOA framework identified several knowledge gaps and this tumor is seen with many<br />

important pharmaceutical and <strong>in</strong>dustrial compounds, this SOT CCT workshop was organized. Troglitazone<br />

is the prototypical low aff<strong>in</strong>ity (μM) PPARγ agonist developed to treat Type II Diabetes. In carc<strong>in</strong>ogenicity<br />

studies, it produced an <strong>in</strong>creased <strong>in</strong>cidence of hemangiosarcomas <strong>in</strong> mice but not rats. These tumors were<br />

primarily seen <strong>in</strong> adipose tissue, consistent with its pharmacology. The first speaker, Dr. March, will<br />

summarize angiogenesis and adipogenesis, as understand<strong>in</strong>g these processes are potentially central to the<br />

MOA. Adipocytes are a rich source of growth factors <strong>in</strong>clud<strong>in</strong>g those that stimulate angiogenesis. Studies<br />

with angiogenesis <strong>in</strong>hibitors have demonstrated that angiogenesis is a necessary requirement for fat pad<br />

growth. Hence, a key component of the MOA framework for PPARγ agonist <strong>in</strong>duction of hemangiosarcoma<br />

is that adipocytes release angiogenic growth factors which would stimulate endothelial cell proliferation.<br />

The second speaker, Dr. Jon Cook, will summarize the work conducted by Pfizer to understand the MOA<br />

for troglitazone as well as describe the HESI MOA framework for PPARγ-<strong>in</strong>duced hemangiosarcoma. The<br />

proposed HESI MOA <strong>in</strong>cludes the follow<strong>in</strong>g major components: (1) PPARγ agonists b<strong>in</strong>d to the PPARγ<br />

receptor <strong>in</strong> adipocytes and stimulate their proliferation lead<strong>in</strong>g to the release of angiogenic growth factors<br />

and/or a decrease <strong>in</strong> anti-angiogenic growth factors; (2) dysregulated angiogenesis occurs due to PPARγ<br />

agonists <strong>in</strong>itially <strong>in</strong>hibit<strong>in</strong>g endothelial cell growth result<strong>in</strong>g <strong>in</strong> local tissue hypoxia which occurs <strong>in</strong> the<br />

presence of a net angiogenic environment lead<strong>in</strong>g to selection of endothelial cells which can proliferate;<br />

and (3) a selective growth stimulus leads to clonal endothelial cell expansion and tumorigenesis. Accessory<br />

cells such as macrophages could also contribute to an angiogenic stimulus (e.g., IL-1, IL-6). An alternative<br />

34


hypothesis is that the target organ (i.e., fat pad) provides an angiogenic growth stimulus that recruits<br />

circulat<strong>in</strong>g endothelial stem cells from the bone marrow and these stems cells seed the target organs lead<strong>in</strong>g<br />

to the formation of hemangiosarcoma. In addition, gaps <strong>in</strong> our knowledge from the MOA framework will<br />

be highlighted to facilitate the discussions at the end of this workshop. The third speaker, Dr. Samuel<br />

Cohen, will review his work to assess the human relevance of mouse endothelial cell tumors <strong>in</strong>duced by<br />

troglitazone. To address this gap, endothelial cell proliferation was assessed us<strong>in</strong>g a dual label of Ki-67 (cell<br />

proliferation) and CD-31 (endothelial cell marker) <strong>in</strong> mice, rats, and humans. Mice were shown to have a<br />

higher basal endothelial cell proliferation rate than rats or humans. Mice have a relatively high background<br />

<strong>in</strong>cidence whereas such tumors are rare <strong>in</strong> rats and humans. In vivo, troglitazone <strong>in</strong>duces endothelial cell<br />

proliferation <strong>in</strong> mice after 4 weeks of dos<strong>in</strong>g. Us<strong>in</strong>g <strong>in</strong> vitro experiments with troglitzone, therapeutic levels<br />

were shown to be cytotoxic to human cells while mitogenic to mouse microvascular endothelial cells.<br />

This differential <strong>in</strong> vitro response suggests that troglitazone would not stimulate a mitogenic endothelial<br />

response <strong>in</strong> humans.<br />

session four speaker abstracts:<br />

angiogenesis and adipogenesis<br />

Keith L. March 1<br />

1 Indiana University School of Medic<strong>in</strong>e, Indianapolis, IN, United States<br />

The delivery of autologous cells to reverse or halt the progression of ischemic diseases of the heart<br />

and peripheral tissues, either by differentiation or secretion of growth factors, is emerg<strong>in</strong>g as a novel<br />

treatment option for patients with cardiovascular disease, but may be limited by the accessibility of<br />

sufficient numbers of certa<strong>in</strong> pluripotent cell types.<br />

Adipose stem cells (ASC) from human subcutaneous fat are a plentiful population of pluripotent<br />

cells which demonstrate activity <strong>in</strong> promot<strong>in</strong>g recovery from peripheral vascular disease (PVD) and<br />

acute myocardial <strong>in</strong>farction. Adipose stromal cells (ASCs) have been isolated from human, mur<strong>in</strong>e<br />

and rat subcutaneous adipose tissues and characterized by flow cytometry. Greater than 75% of the<br />

total population of adherent cells express the stem cell surface marker CD34 and are negative for<br />

the hematopoietic markers CD45 and c-kit. Studies of phenotypic differentiation have <strong>in</strong>dicated that<br />

ASCs possess the ability to participate as perivascular cells with characteristics of pericytes, as well<br />

as to adopt an alternative adipogenic fate. Complementary studies have been conducted to address<br />

the potential supportive function of ASCs <strong>in</strong> promot<strong>in</strong>g recruitment, protection and differentiation of<br />

relevant pluripotent cells. We have shown that ASCs secrete multiple angiogenic and anti-apoptotic<br />

growth factors at bioactive levels that can enhance endothelial cell growth and survival. We have also<br />

demonstrated therapeutic potential of ASCs <strong>in</strong> a mouse model of peripheral vascular disease as well<br />

as a rat model of acute myocardial <strong>in</strong>farction. In ischemic mouse h<strong>in</strong>dlimbs, ASCs have been found<br />

to survive <strong>in</strong> vivo for at least 1 week. Furthermore, ASCs delivered by <strong>in</strong>jection have been able to<br />

promote blood reperfusion and <strong>in</strong>creased capillary density <strong>in</strong> surgically <strong>in</strong>duced ischemic h<strong>in</strong>dlimbs<br />

of mice. Critical to the success of these approaches is the development of consistent and reasonably<br />

efficient cell delivery methodology to permit the ASCs to provide paracr<strong>in</strong>e stimulation to the organs<br />

of <strong>in</strong>terest.<br />

We expect progress <strong>in</strong> understand<strong>in</strong>g the paracr<strong>in</strong>e and vascular <strong>in</strong>teractions of ASCs to provide<br />

<strong>in</strong>sight <strong>in</strong>to a range of novel potential pharmacological targets. These encourag<strong>in</strong>g results have laid the<br />

foundation for the first cl<strong>in</strong>ical trials us<strong>in</strong>g ASCs to treat both cardiac and peripheral ischemic diseases<br />

<strong>in</strong> humans.<br />

35<br />

(Cont<strong>in</strong>ued on next page)


troglitazone: an illustration of the hesi mode-of-action framework<br />

for ppar gamma-<strong>in</strong>duced hemangiosarcoma<br />

Jon C. Cook 1<br />

1 Pfizer Inc., Groton, CT, United States<br />

Troglitazone is a low aff<strong>in</strong>ity (μM) PPARγ agonist developed to treat Type II Diabetes. In<br />

carc<strong>in</strong>ogenicity studies, it produced hemangiosarcomas <strong>in</strong> mice but not rats. These tumors were<br />

primarily seen <strong>in</strong> adipose tissue, consistent with its pharmacology. In July 2005, the HESI PPAR<br />

Agonist Project Committee was formed to address the carc<strong>in</strong>ogenicity concerns from the high aff<strong>in</strong>ity<br />

(nM) PPARγ and dual γ/α agonists. One of the HESI workstreams developed a MOA Framework<br />

for hemangiosarcoma based on data from the open literature as well as on-go<strong>in</strong>g research with high<br />

aff<strong>in</strong>ity agonists. This talk will summarize the <strong>in</strong>itial work conducted by Pfizer to understand the MOA<br />

as well as <strong>in</strong>tegrate these data <strong>in</strong>to the HESI MOA framework for PPARγ-<strong>in</strong>duced hemangiosarcoma.<br />

troglitazone effects on endothelial Cells In Vivo and In Vitro:<br />

Differences between mice and humans<br />

Samuel M. Cohen 1 , Satoko Kiyota 1 , Shugo Suzuki 1 , and Lora L. Arnold 1<br />

1<br />

Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE,<br />

United States<br />

PPARγ and dual agonists frequently <strong>in</strong>duce hemangiomas and hemangiosarcomas <strong>in</strong> mice but not<br />

rats. We have used troglitazone as a prototype drug to <strong>in</strong>vestigate the mode-of-action. Utiliz<strong>in</strong>g a<br />

dual label<strong>in</strong>g technique, we demonstrated that mice have a higher endothelial cell proliferation rate<br />

than either rats or humans. Mice have a relatively high spontaneous <strong>in</strong>cidence of hemangiosarcomas<br />

whereas rats and humans do not. Troglitazone is nongenotoxic. It <strong>in</strong>duces endothelial cell proliferation<br />

<strong>in</strong> vivo <strong>in</strong> the brown and white fat pads and <strong>in</strong> the liver at doses that are carc<strong>in</strong>ogenic <strong>in</strong> mice. Utiliz<strong>in</strong>g<br />

mouse and human microvascular endothelial cells <strong>in</strong> vitro, troglitazone is cytotoxic to each, but the<br />

human cells are more sensitive than the mouse. However, troglitazone is mitogenic to the mouse cells<br />

but not to human endothelial cells. This appears to be due to <strong>in</strong>creased cell proliferation comb<strong>in</strong>ed<br />

with decreased apoptosis. These data strongly suggest a direct mitogenic effect as a possible mode-ofaction<br />

for troglitazone-<strong>in</strong>duced endothelial cell tumorigenesis <strong>in</strong> mice, and suggests that the human is<br />

resistant to this effect. A possible paracr<strong>in</strong>e effect, however, is suggested by the presence of mammary<br />

ductular cell hyperplasia <strong>in</strong> the fat pad of troglitazone-treated mice.<br />

36


session fiVe: hemangiosarComa <strong>in</strong>DuCeD by other<br />

pharmaCeutiCals<br />

Hemanigosarcomas <strong>in</strong> rodents have become an <strong>in</strong>creas<strong>in</strong>gly common issue <strong>in</strong> carc<strong>in</strong>ogenicity studies<br />

of a variety of classes of pharmaceutical agents, <strong>in</strong> addition to the PPAR agonists. Based on a variety of<br />

standard <strong>in</strong> vitro and <strong>in</strong> vivo genotoxicity assays, the pharmaceutical agents produc<strong>in</strong>g these effects are not<br />

genotoxic, and specifically they are not DNA reactive. Thus, their mode-of-action must <strong>in</strong>volve an <strong>in</strong>crease<br />

<strong>in</strong> cell proliferation, either based on an <strong>in</strong>crease <strong>in</strong> cell births and/or a decrease <strong>in</strong> cell deaths. The specific<br />

mode-of-action for these agents has yet to be def<strong>in</strong>ed, although several promis<strong>in</strong>g avenues of research<br />

are be<strong>in</strong>g pursued. The evidence <strong>in</strong>creas<strong>in</strong>gly suggests that specific <strong>in</strong>teraction of the pharmaceutical<br />

agents with specific receptors is required. Our lack of understand<strong>in</strong>g of the MOA for the <strong>in</strong>duction of<br />

hemangiosarcomas by these chemicals poses significant uncerta<strong>in</strong>ty <strong>in</strong> the assessment of human risk,<br />

complicat<strong>in</strong>g regulatory decision mak<strong>in</strong>g.<br />

Several basic questions regard<strong>in</strong>g endothelial sarcoma <strong>in</strong>duction rema<strong>in</strong>. Of fundamental importance is<br />

determ<strong>in</strong>ation of the target cell that gives rise eventually to the sarcomas. Is it the endothelial cell itself<br />

or a precursor stem cell aris<strong>in</strong>g from the bone marrow? Why do the tumors localize <strong>in</strong> certa<strong>in</strong> tissues,<br />

predom<strong>in</strong>antly liver, spleen and bone marrow <strong>in</strong> mice, but subcutaneous adipose tissue <strong>in</strong> humans? Does<br />

the <strong>in</strong>duc<strong>in</strong>g chemical act directly on the cells that become transformed or do they act on other cell types<br />

produc<strong>in</strong>g a biologic effect lead<strong>in</strong>g to the release of growth factors or other substances which then affect<br />

the precursor cells? Are observations <strong>in</strong> studies on angiogenesis occurr<strong>in</strong>g <strong>in</strong> physiologic or pathologic<br />

processes, such as <strong>in</strong>flammation or tumorigenesis, relevant to the malignant transformation lead<strong>in</strong>g to<br />

hemangiosarcomas? Many of the <strong>in</strong>vestigations on angiogenesis and hemangiosarcoma <strong>in</strong>duction are<br />

performed us<strong>in</strong>g <strong>in</strong> vitro culture systems. Do these reflect the complex <strong>in</strong>teractions that are occurr<strong>in</strong>g<br />

between different cell types and tissues that may be required for sarcomagenesis? Are endothelial cells<br />

derived from large vessels, such as the umbilical ve<strong>in</strong> or vena cave, biologically the same <strong>in</strong> vitro as<br />

endothelial cells from microvascular sources? Are endothelial cells derived from one microvascular source,<br />

such as subcutaneous adipose tissue, biologically the same as from another source, such as the liver?<br />

Some of these issues have been addressed <strong>in</strong> previous sessions. Some have arisen <strong>in</strong> <strong>in</strong>vestigations on<br />

other classes of pharmaceuticals. In addition to the observations of PPAR agonists presented <strong>in</strong> Session 4,<br />

additional examples will be presented <strong>in</strong> this session; namely, fenret<strong>in</strong>ide which is a ret<strong>in</strong>oic acid receptor<br />

(RAR) receptor agonist and pregabal<strong>in</strong> which is an α2δ receptor agonist.<br />

In session four, the specific modes-of-action be<strong>in</strong>g <strong>in</strong>vestigated for PPAR agonists are presented. The PPAR<br />

nuclear receptor forms a heterodimer with the ret<strong>in</strong>oid receptor (RXR). Ret<strong>in</strong>oids have frequently been<br />

shown to <strong>in</strong>duce hemangiosarcomas <strong>in</strong> mice. Dr. Timothy E. Johnson will present the results of extensive<br />

<strong>in</strong>vestigations concern<strong>in</strong>g fenret<strong>in</strong>ide, which <strong>in</strong>duces a 100% <strong>in</strong>cidence of hemangiosarcomas at several<br />

sites <strong>in</strong> mice, but, like the PPAR agonists, most are <strong>in</strong> the adipose tissue, either subcutaneous or abdom<strong>in</strong>al<br />

cavity. These compounds b<strong>in</strong>d RAR, and evidence based on <strong>in</strong> vitro <strong>in</strong>vestigations demonstrates that they<br />

negatively regulate cell growth and angiogenesis, quite possibly through RAR-<strong>in</strong>dependent pathways. The<br />

issue of off-target receptor <strong>in</strong>teraction is another of several critical issues that needs to be addressed <strong>in</strong><br />

extrapolat<strong>in</strong>g the observations <strong>in</strong> mice to humans. Several possible factors have also been demonstrated to<br />

be <strong>in</strong>volved <strong>in</strong> the hemangiosarcomas <strong>in</strong>duced <strong>in</strong> mice by pregabal<strong>in</strong>, which will be presented by Dr. Kay<br />

A. Criswell. Most of these tumors occurred <strong>in</strong> the usual tissues <strong>in</strong>volved <strong>in</strong> mice, namely liver, spleen, and<br />

bone marrow. The data <strong>in</strong>dicate that the <strong>in</strong>itial event is the <strong>in</strong>duction of hypoxia at high doses, lead<strong>in</strong>g to a<br />

cascade of events <strong>in</strong>clud<strong>in</strong>g the overproduction of megakaryocytes and platelets, with consequent release<br />

of various endothelial growth factors, <strong>in</strong>clud<strong>in</strong>g vascular endothelial growth factor (VEGF) and plateletderived<br />

growth factor (PDGF).<br />

37<br />

(Cont<strong>in</strong>ued on next page)


Ultimately, the human relevance of these modes of action needs to be evaluated. The <strong>in</strong>creased<br />

<strong>in</strong>cidences of hemangiosarcomas consistently occur <strong>in</strong> mice, but not rats. Mice have a high background<br />

<strong>in</strong>cidence of hemangiosarcomas, whereas rats do not. These tumors are rare <strong>in</strong> humans. With respect to<br />

hemangiosarcoma <strong>in</strong>duction, the question rema<strong>in</strong>s as to whether humans behave like mice or rats.<br />

session five speaker abstracts:<br />

ret<strong>in</strong>oid-<strong>in</strong>duced hemangiosarcoma <strong>in</strong> mice: potential <strong>in</strong>sight from<br />

In Vivo and In Vitro studies<br />

Timothy E. Johnson 1<br />

1 Merck Research Laboratories, West Po<strong>in</strong>t, PA, United States<br />

The synthetic ret<strong>in</strong>oid, 4-hyroxyphenyl ret<strong>in</strong>amide/fenret<strong>in</strong>ide (4-HPR) causes hemangiosarcomas<br />

<strong>in</strong> mice, primarily <strong>in</strong> the subcutis and abdom<strong>in</strong>al soft tissues. Other ret<strong>in</strong>oids, ret<strong>in</strong>yl acetate and<br />

etret<strong>in</strong>ate, are also mouse hemangiosarcomagens, albeit much weaker than 4-HPR. The mechanism<br />

of ret<strong>in</strong>oid <strong>in</strong>duced hemangiosarcoma is poorly understood. 4-HPR and other ret<strong>in</strong>oids b<strong>in</strong>d to and<br />

transactivate ret<strong>in</strong>oic acid receptors (RARs) on target genes and regulate gene transcription. In cultured<br />

cells, 4-HPR treatment results <strong>in</strong> growth <strong>in</strong>hibition and the <strong>in</strong>duction of apoptosis. Although some of<br />

these actions might be through RAR dependent pathways, studies us<strong>in</strong>g cells conta<strong>in</strong><strong>in</strong>g non functional<br />

RARs suggest that much of the anti-proliferative effects are RAR <strong>in</strong>dependent. In bov<strong>in</strong>e endothelial<br />

cells, 4-HPR caused an <strong>in</strong>crease <strong>in</strong> apoptosis which correlated with elevated levels of ceramide.<br />

Interest<strong>in</strong>gly, fumonison, an <strong>in</strong>hibitor of ceramide synthesis, prevented 4-HPR <strong>in</strong>duced cell kill<strong>in</strong>g.<br />

Other data suggests that 4-HPR transactivates the peroxisome proliferator activated receptor-gamma<br />

(PPARg), at concentrations that cause growth <strong>in</strong>hibition <strong>in</strong> human oral carc<strong>in</strong>oma cell l<strong>in</strong>es. In addition<br />

to be<strong>in</strong>g a negative regulator of cell growth, 4-HPR also <strong>in</strong>hibits <strong>in</strong> vitro angiogenesis. Thus, the<br />

<strong>in</strong>duction of apoptosis and the anti-angiogenic effect of 4-HPR and other ret<strong>in</strong>oids might promote an<br />

environment that is conducive to hemangiosarcoma development <strong>in</strong> mice.<br />

epigenetic mode of action associated with <strong>in</strong>duction of<br />

hemangiosarcoma <strong>in</strong> mice treated with pregabal<strong>in</strong><br />

Kay A. Criswell 1<br />

1 Pfizer Global Research and Development, Groton, CT, United States<br />

In 2-year carc<strong>in</strong>ogenicity studies <strong>in</strong> mice, but not rats, pregabal<strong>in</strong> treatment selectively <strong>in</strong>creased the<br />

<strong>in</strong>cidence of hemangiosarcoma primarily <strong>in</strong> hematopoietic tissues (liver, spleen, bone marrow). An<br />

extensive body of work has empirically established a proposed mode-of-action for hemangiosarcoma<br />

<strong>in</strong>duction by pregabal<strong>in</strong> <strong>in</strong> mice. Early events <strong>in</strong> this process <strong>in</strong> mice are <strong>in</strong>creases <strong>in</strong> HCO 3 and blood<br />

pH and correspond<strong>in</strong>g decreases <strong>in</strong> respiratory rate and m<strong>in</strong>ute volume, which are all tightly l<strong>in</strong>ked<br />

physiological responses. Extensive changes <strong>in</strong> mouse bone marrow occur, and these are consistent<br />

with effects of hypoxia and/or alkalosis and are characterized by erythropoiesis, megakaryopoiesis,<br />

<strong>in</strong>creased macrophages with erythrophages, and clusters of hemosider<strong>in</strong>-laden macrophages suggestive<br />

of macrophage activation. Extramedullary hematopoiesis is also evident <strong>in</strong> the spleens of pregabal<strong>in</strong>treated<br />

mice. Peripheral erythrocyte and platelet counts are <strong>in</strong>creased <strong>in</strong> association with bone marrow<br />

changes and platelet activation is also <strong>in</strong>creased. Review of the National Toxicology Program database<br />

regard<strong>in</strong>g compounds that <strong>in</strong>duce mouse-specific hemangiosarcomas by a nongenotoxic mechanism<br />

38


suggest that accelerated hematopoiesis is a common feature among them. Together, the bone marrow<br />

and platelet changes create a growth factor-rich environment, which is trophic for endothelial cells.<br />

Indeed, platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), vascular<br />

endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) elevations are observed <strong>in</strong> plasma<br />

or <strong>in</strong> liver, spleen, or bone marrow (the three tissues with the highest <strong>in</strong>cidence of hemangiosarcomas),<br />

lead<strong>in</strong>g to proliferation of endothelial cells, the target cell type for hemangiosarcoma <strong>in</strong>duction.<br />

With the exception of respiratory parameters <strong>in</strong> rats, which were transiently decreased, none of the<br />

pregabal<strong>in</strong>-<strong>in</strong>duced factors identified <strong>in</strong> mice were detected <strong>in</strong> rats, monkeys, or humans. Therefore, the<br />

data <strong>in</strong>dicate that hemangiosarcoma <strong>in</strong>duction is species-specific to mice and that pregabal<strong>in</strong> does not<br />

present a carc<strong>in</strong>ogenic risk to humans.<br />

session six: regulatory perspeCtiVes on Data gaps<br />

The human relevance of treatment–related hemangiomas and hemangiosarcomas <strong>in</strong> laboratory animals<br />

has been the subject of much debate and various hypotheses have been put forth. These hypotheses need<br />

to be judged aga<strong>in</strong>st data <strong>in</strong> a rigorous, structured, and transparent manner <strong>in</strong> order to determ<strong>in</strong>e whether<br />

available data support a postulated mode of carc<strong>in</strong>ogenic action for any given chemical or chemical class<br />

and its relevance for humans. There may not be a s<strong>in</strong>gle pathway by which these tumors are <strong>in</strong>duced by<br />

various chemicals. Thus, alternative modes of action that logically present themselves must be considered.<br />

Weight-of-evidence approaches to understand<strong>in</strong>g the mode of action and human relevance of animal<br />

tumor responses have been <strong>in</strong>troduced (i.e., the IPCS and ILSI’s Human Cancer Relevance Framework).<br />

To further our understand<strong>in</strong>g of this carc<strong>in</strong>ogenic process, it is important to identify critical data gaps and<br />

<strong>in</strong>form the design of studies related to modes of action. This session will provide a regulatory perspective<br />

on the issues surround<strong>in</strong>g <strong>in</strong>duced hemangiomas and hemangiosarcomas responses <strong>in</strong> animal studies for<br />

pharmaceuticals, pesticides, and <strong>in</strong>dustrial chemicals.<br />

session six speaker abstracts:<br />

epa perspectives<br />

Vicki L. Dellarco 1<br />

1 Office of Pesticide Programs, U.S. Environmental Protection Agency, Wash<strong>in</strong>gton, DC, United States<br />

A prelim<strong>in</strong>ary exam<strong>in</strong>ation of chronic rodent bioassays on pesticides shows that hemangiosarcomas<br />

occur relatively <strong>in</strong>frequent (approximately 5% of those compounds produc<strong>in</strong>g a tumor response). This<br />

type of tumor response seems to be observed <strong>in</strong> the liver or spleen but has been found <strong>in</strong> other tissues<br />

(e.g., sk<strong>in</strong> or adipose). This response also appears to occur more <strong>in</strong> mice than rats. Mode-of-Action<br />

data are not typically available, thus an understand<strong>in</strong>g of the various pathways to hemangiosarcoma<br />

would be useful <strong>in</strong> determ<strong>in</strong><strong>in</strong>g the relevance of these f<strong>in</strong>d<strong>in</strong>gs to humans.<br />

(This abstract has not received formal Agency review and does not represent the views or policies of<br />

the U.S. Environmental Protection Agency.)<br />

39<br />

(Cont<strong>in</strong>ued on next page)


hemangiosarcomas and pharmaceuticals: an fDa perspective<br />

Abigail C. Jacobs 1<br />

1 Center for Drug Evaluation and Research, U.S. Food and Drug Adm<strong>in</strong>istration, Silver Spr<strong>in</strong>g, MD,<br />

United States<br />

The relevance of hemangiomas and hemangiosarcomas <strong>in</strong> mice to humans is not known. There<br />

appear to be several different pathways to hemangiosarcoma for pharmaceuticals <strong>in</strong> rats and mice. For<br />

pharmaceuticals, some effects may be pharmacologic exaggeration or off-target and relevant across<br />

species, and some modes of action may be species specific. Different classes of pharmaceuticals may<br />

have different modes of action for formation of hemangiosarcomas. F<strong>in</strong>d<strong>in</strong>gs with pharmaceuticals<br />

complement what has been seen for other chemicals. Hemangiosarcoma, primarily <strong>in</strong> mice, have been<br />

seen with many PPAR dual agonists, some but not all calcium channel blockers, antipychotics, PDE-5<br />

<strong>in</strong>hibitors, DPP-4 <strong>in</strong>hibitors, antiarrythmics, antisense compounds, nitric oxide releasers, hemolytic<br />

compounds, and VEGF <strong>in</strong>ducers. Understand<strong>in</strong>g of the various pathways to hemangiosarcoma is<br />

needed <strong>in</strong> order to determ<strong>in</strong>e relevance of these f<strong>in</strong>d<strong>in</strong>gs to humans.<br />

european perspectives on Carc<strong>in</strong>ogenicity test<strong>in</strong>g<br />

Jan Willem van der Laan 1<br />

1 National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands<br />

When prepar<strong>in</strong>g ICH discussions <strong>in</strong> the EU the noncl<strong>in</strong>ical assessors from Germany and the<br />

Netherlands prepared a survey of all carc<strong>in</strong>ogenicity studies carried out for human pharmaceuticals<br />

(Van Oosterhout et al, 1997). The Safety Work<strong>in</strong>g Party of the CPMP shared the conclusion of the<br />

authors that, based on this experience the carc<strong>in</strong>ogenic risk will be identified sufficiently with a s<strong>in</strong>gle<br />

long-term carc<strong>in</strong>ogenicity study <strong>in</strong> rats. Mechanistic studies are more important to expla<strong>in</strong> the action,<br />

than to confirm it <strong>in</strong> a mouse study. Despite this EU position the Guidel<strong>in</strong>e S1B still requires two<br />

studies, one preferably with rats, while the other study can be either a short-term study <strong>in</strong> transgenic<br />

mice or a long-term study <strong>in</strong> normal mice (<strong>in</strong> that sequence). An update of the carc<strong>in</strong>ogenicity database<br />

will be presented. After completion of this guidel<strong>in</strong>e the PPARγ-agonists showed a relatively specific<br />

pattern of tumors <strong>in</strong> rats (bladder tumors) and mice (hemangiosarcoma). S<strong>in</strong>ce no adequate explanation<br />

could be found expla<strong>in</strong><strong>in</strong>g the rise of the hemangiosarcoma, the appearance of these tumors lead to<br />

regulatory measures related to this class of compounds, i.e. restrict<strong>in</strong>g cl<strong>in</strong>ical trials up to 6 months,<br />

challeng<strong>in</strong>g the conclusion of the earlier study that mouse-derived data are not relevant for the human<br />

situation.<br />

40


session seVen: moDe(s) of aCtion for hemangiosarComa<br />

<strong>in</strong>DuCtion<br />

Our lack of understand<strong>in</strong>g of the MOA for the <strong>in</strong>duction of hemangiosarcomas by nongenotoxic chemicals<br />

poses significant uncerta<strong>in</strong>ty <strong>in</strong> the assessment of human risk, complicat<strong>in</strong>g regulatory decision mak<strong>in</strong>g.<br />

The goal of this panel-led forum is to discuss whether there is a unify<strong>in</strong>g MOA framework for the<br />

nongenotoxic agents that were discussed at this workshop. The workshop organizers will modify the MOA<br />

framework developed for troglitzone based on the discussions at this workshop and use this framework to<br />

stimulate the forum discussion. A key outcome from the panel discussion is to highlight research gaps <strong>in</strong><br />

the MOA and identify key experiments to address these gaps.<br />

The follow<strong>in</strong>g list of questions are meant to stimulate and guide the panel discussion:<br />

Of fundamental importance is determ<strong>in</strong>ation of the target cell that gives rise eventually to the sarcomas.<br />

Is it the endothelial cell itself or a precursor stem cell aris<strong>in</strong>g from the bone marrow?<br />

Why do the tumors localize <strong>in</strong> certa<strong>in</strong> tissues, predom<strong>in</strong>antly liver, spleen and bone marrow <strong>in</strong> mice, but<br />

subcutaneous adipose tissue <strong>in</strong> humans?<br />

Spontaneous hemangiosarcomas occur commonly <strong>in</strong> mice but rarely <strong>in</strong> rats or humans. What is the basis<br />

for this <strong>in</strong> mice, and are mice an acceptable model for predict<strong>in</strong>g this tumor type <strong>in</strong> humans?<br />

Does the <strong>in</strong>duc<strong>in</strong>g chemical act directly on the cells that become transformed or do they act on other cell<br />

types produc<strong>in</strong>g a biologic effect lead<strong>in</strong>g to the release of growth factors or other substances which then<br />

affect the precursor cells?<br />

Are observations <strong>in</strong> studies on angiogenesis occurr<strong>in</strong>g <strong>in</strong> physiologic or pathologic processes, such as<br />

<strong>in</strong>flammation or tumorigenesis, relevant to the malignant transformation lead<strong>in</strong>g to hemangiosarcomas?<br />

Many of the <strong>in</strong>vestigations on angiogenesis and hemangiosarcoma <strong>in</strong>duction are performed us<strong>in</strong>g<br />

<strong>in</strong> vitro culture systems. Do these reflect the complex <strong>in</strong>teractions that are occurr<strong>in</strong>g between different cell<br />

types and tissues that may be required for sarcomagenesis?<br />

Are endothelial cells derived from large vessels, such as the umbilical ve<strong>in</strong> or vena cava, biologically the<br />

same <strong>in</strong> vitro as endothelial cells from microvascular sources?<br />

Are endothelial cells derived from one microvascular source, such as subcutaneous adipose tissue,<br />

biologically the same as from another source, such as the liver?<br />

41


notes<br />

42


poster abstraCts list<strong>in</strong>g by author, title, and number<br />

abstract author abstract title abstract #<br />

Matt Cave et al. Biomarkers for V<strong>in</strong>yl Chloride-Induced Hepatic<br />

Hemangiosarcoma<br />

Matt Cave et al. V<strong>in</strong>yl Chloride-Induced Hepatic Hemangiosarcoma:<br />

The Louisville Experience<br />

Tim Coskran et al. Evidence for Hypoxia <strong>in</strong> Tissues Associated with<br />

Hemangiosarcoma <strong>in</strong> 2-BE-Treated Mice<br />

L<strong>in</strong>a Gao et al. 1N 6 -Etheno-2’Deoxyadenos<strong>in</strong>e (εdA) <strong>in</strong> Weanl<strong>in</strong>g and<br />

Adult Rats Exposed to 13 C 2-V<strong>in</strong>yl Chloride by Inhalation<br />

Mark M. Gos<strong>in</strong>k et al. Systems Biology Analysis Suggests Insul<strong>in</strong>/AKT<br />

Signal<strong>in</strong>g <strong>in</strong> Compound-Induced Hemangiosarcoma<br />

Satoko Kakiuchi-Kiyota<br />

et al.<br />

Lisa M. Kamendulis<br />

et al.<br />

Evaluation of the Effects of the PPAR-Gamma Agonist<br />

Troglitazone on Cytotoxicity and Mitogenesis of<br />

Endothelial Cells: Differences between Human and<br />

Mouse<br />

Mechanistic Studies of Polyhexamethylene Biguanide<br />

(PHMB) Carc<strong>in</strong>ogenicity<br />

Daphna Laifenfeld et al. Causal Network Model<strong>in</strong>g Identifies Hypoxia-<br />

Induced Inflammation as a Potential Contributor to<br />

2-Butoxyethanol-Induced Hemangiosarcomas<br />

Esra Mutlu et al. Molecular Dosimetry of the V<strong>in</strong>yl Chloride-Induced<br />

DNA Adduct, 7-Oxoethylguan<strong>in</strong>e<br />

Esra Mutlu et al. A New LC-MS/MS Method for Detection of the V<strong>in</strong>yl<br />

Chloride-Induced DNA Adduct N 2 ,3-Ethenoguan<strong>in</strong>e<br />

Chunhua Q<strong>in</strong> et al. Mutations <strong>in</strong> the PTEN, Tek, VHLH and Kdr/Vegfr2<br />

Genes Are Not Susceptibility Factors for Spontaneous<br />

Hemangiosarcoma Development <strong>in</strong> Mice<br />

Chunhua Q<strong>in</strong> et al. DNA Methylation Changes Identified <strong>in</strong> Spontaneous<br />

Hemangiosarcomas and PPARγ Agonist-Treated Mouse<br />

Endothelial Cells <strong>in</strong> a Genome-Wide Methylation<br />

Profil<strong>in</strong>g Study<br />

Chunhua Q<strong>in</strong> et al. Potential Bone Marrow Stem Cell Orig<strong>in</strong> of<br />

Hemangiosarcoma and the Role of TGFβ and BMP<br />

Signal<strong>in</strong>g Pathways<br />

Sharon Sokolowski et al. Development of Flow Cytometry and Laser Scann<strong>in</strong>g<br />

Cytometry Methods to Assess Endothelial Cell<br />

Proliferation <strong>in</strong> Liver, Spleen, and Bone Marrow of<br />

2-Butoxyethanol-Treated Mice<br />

43<br />

1<br />

2<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

13<br />

14<br />

poster abstraCts


notes<br />

44


poster abstracts<br />

1. biomarkers for V<strong>in</strong>yl Chloride-<strong>in</strong>duced hepatic hemangiosarcoma<br />

Matt Cave 1 , Keith Falkner 1 , Mihir Patel 1 , Lark Reynolds 1 , Shirish Barve 1 , and Craig McCla<strong>in</strong> 1<br />

1 Department of Medic<strong>in</strong>e, University of Louisville, Louisville, KY, United States<br />

Purpose: V<strong>in</strong>yl chloride (VC) exposure is a well-documented risk factor for hepatic hemangiosarcoma.<br />

This association was first made <strong>in</strong> 1974 at a Kentucky chemical plant. To date, 25 employees from this<br />

plant have developed hemangiosarcoma, and this is the largest s<strong>in</strong>gle site cluster <strong>in</strong> the United States.<br />

Nationally, 80,000 chemical workers have been exposed to VC. Due to a long latency period, many are<br />

still at risk for hemangiosarcoma. A biomarker is needed for the early detection of this deadly tumor.<br />

Hemangiosarcoma arises from s<strong>in</strong>usoidal endothelial cells which also metabolize hyaluronic acid<br />

(HA). Here, we test the hypothesis that serum HA is elevated <strong>in</strong> VC workers with, or who subsequently<br />

developed, hemangiosarcoma.<br />

Materials and Methods: A cohort of 103 workers with high VC exposure has been followed cl<strong>in</strong>ically<br />

s<strong>in</strong>ce 1974 for the development of hemangiosarcoma. Us<strong>in</strong>g a nested case control approach, serum HA<br />

levels were measured <strong>in</strong> archived samples from workers with hemangiosarcoma or from those who<br />

subsequently developed hemangiosarcoma, and <strong>in</strong> control groups from the same plant with reduced VC<br />

exposures that did not develop hemangiosarcoma.<br />

Results: The average values of rout<strong>in</strong>e liver chemistries were surpris<strong>in</strong>gly normal at the time of diagnosis<br />

of hemangiosarcoma: ALT 24 (±14) U/L, AST 34 (±19) U/L, alkal<strong>in</strong>e phosphatase 66 (±36) U/L, total<br />

bilirub<strong>in</strong> 1.2 (±2.2) mg/dL, and album<strong>in</strong> 4.3 (±0.31) g/dL. The mean HA level <strong>in</strong> 2 patients at the time<br />

of diagnosis of hemangiosarcoma was markedly elevated (464 μg/L). The mean HA level <strong>in</strong> 11 workers<br />

at 11±6.9 years prior to the diagnosis of hemangiosarcoma was 71.6 μg/L (±58.1). This was greater than<br />

the mean HA levels <strong>in</strong> both the high VC exposure control group (30.0+24.9 μg/L, p = 5.57 x 10 -5 ) and the<br />

low VC exposure control group (32.0±26.7 μg/L, p = 0.00703). No differences <strong>in</strong> rout<strong>in</strong>e liver chemistries<br />

were noted between groups. A l<strong>in</strong>ear regression analysis revealed no relationship between cumulative VC<br />

exposure and HA level (r 2 =0.03). Thus, future hemangiosarcoma was associated with elevated HA which<br />

was <strong>in</strong>dependent of cumulative VC exposure.<br />

Conclusions: Rout<strong>in</strong>e liver chemistries were <strong>in</strong>effective biomarkers to detect v<strong>in</strong>yl chloride-<strong>in</strong>duced<br />

hepatic hemangiosarcoma. In contrast, serum hyaluronic acid was elevated <strong>in</strong> subjects with, or who<br />

subsequently developed, this deadly liver cancer. We are work<strong>in</strong>g on identify<strong>in</strong>g additional biomarkers for<br />

this problem.<br />

45<br />

(Cont<strong>in</strong>ued on next page)


2. V<strong>in</strong>yl Chloride-<strong>in</strong>duced hepatic hemangiosarcoma: the louisville<br />

experience<br />

Matt Cave 1 , Keith Falkner 1 , Mihir Patel 1 , Lark Reynolds 1 , Shirish Barve 1 , and Craig McCla<strong>in</strong> 1<br />

1 Department of Medic<strong>in</strong>e, University of Louisville, Louisville, KY, United States<br />

Purpose: V<strong>in</strong>yl chloride (VC) exposure is a well-documented risk factor for hepatic hemangiosarcoma.<br />

This association was first made <strong>in</strong> 1974 at a Kentucky chemical plant. To date, 25 employees from this<br />

plant have developed hemangiosarcoma, and this is the largest s<strong>in</strong>gle site cluster <strong>in</strong> the United States.<br />

Nationally, 80,000 chemical workers have been exposed to VC. Due to a long latency period, many are still<br />

at risk for this deadly cancer.<br />

Materials and Methods: A cohort of 103 chemical workers with high cumulative VC exposure has been<br />

followed cl<strong>in</strong>ically over 30 years as part of a cancer screen<strong>in</strong>g program <strong>in</strong>stituted by their employer <strong>in</strong><br />

collaboration with the University of Louisville. Cl<strong>in</strong>ical data are subsequently analyzed.<br />

Results: All subjects were white males. The mean age at the time of diagnosis of hemangiosarcoma<br />

was 58.6 (±12, standard deviation) years old. The average BMI was 25.0 (±3.12). Approximately ¾ of<br />

the subjects neither smoked cigarettes nor drank alcohol. The mean cumulative v<strong>in</strong>yl chloride exposure<br />

was 17,200 (±6820) PPM-Yr. Hemangiosarcoma presented at an average of 30.7 (±12.2) years after first<br />

exposure and 22.2 (±11.3) years after reach<strong>in</strong>g a threshold exposure of 7000 PPM-Yr. No employee with<br />

a cumulative exposure below this value has developed hemangiosarcoma to date. Common present<strong>in</strong>g<br />

symptoms <strong>in</strong>cluded: abdom<strong>in</strong>al pa<strong>in</strong> (82%), nausea (63%), weight loss (63%), fatigue (45%), fever (41%),<br />

and jaundice (14%). The average values of rout<strong>in</strong>e liver function tests were surpris<strong>in</strong>gly normal at the time<br />

of diagnosis of hemangiosarcoma: ALT 24 (±14) U/L, AST 34 (±19) IU/L, alkal<strong>in</strong>e phosphatase 66 (±36)<br />

IU/L, total bilirub<strong>in</strong> 1.2 (±2.2) mg/dL, and album<strong>in</strong> 4.3 (±0.31) g/dL. Liver biopsy slides were available <strong>in</strong><br />

6 cases and were over-read by a s<strong>in</strong>gle expert liver pathologist. In addition to hemangiosarcoma, 5 subjects<br />

had co-exist<strong>in</strong>g nonalcoholic steatohepatitis, 3 had s<strong>in</strong>usoidal dilation, and 1 had peliosis hepatitis. All 25<br />

subjects died of hemangiosarcoma despite aggressive treatment <strong>in</strong> the majority of cases. One employee<br />

lived 14.5 years after surgical resection. The mean survival for the other 24 was only 1.24 (± 1.38) years<br />

after diagnosis.<br />

Conclusions: Hepatic hemangiosarcoma develops after a long period of high occupational VC exposure<br />

and has been uniformly fatal. At the time of diagnosis of hemangiosarcoma, present<strong>in</strong>g symptoms were<br />

nonspecific and rout<strong>in</strong>e liver chemistries were surpris<strong>in</strong>gly normal.<br />

46


3. evidence for hypoxia <strong>in</strong> tissues associated with hemangiosarcoma <strong>in</strong><br />

2-be-treated mice<br />

Tim Coskran 1 , Xiul<strong>in</strong>g Guo 1 , Ch<strong>in</strong>-Hu Huang 1 , Petra Koza-Taylor 1 , Damir Simic 1 , Leslie Obert 1 ,<br />

Michael Lawton 1 , Mart<strong>in</strong> Sanders 1 , and Chris Somps 1<br />

1 Pfizer Global Research and Development, Groton, CT, United States<br />

2-Butoxyethanol (2-BE) causes hemolysis <strong>in</strong> mice and rats and is associated with an <strong>in</strong>creased <strong>in</strong>cidence of<br />

hemangiosarcoma <strong>in</strong> mouse, but not rat, liver. A non-genotoxic mode of action (MOA) has been proposed<br />

<strong>in</strong> which hemolysis leads to hepatic iron accumulation, activation of local macrophages (Kupffer cells),<br />

oxidative damage and release of growth factors. Together, these responses could produce an environment<br />

that stimulates clonal endothelial cell expansion and tumorigenesis or stimulates recruitment of circulat<strong>in</strong>g<br />

endothelial precursor cells from other hematopoietic tissues, such as bone marrow or spleen. An alternative<br />

MOA that may also be important is that hemolysis produces tissue hypoxia, a stimulus known to play<br />

a key role <strong>in</strong> endothelial cell tumor development. Hypoxia activates the HIF (hypoxia <strong>in</strong>ducible factor)<br />

transcription factor, which regulates a variety of signal<strong>in</strong>g pathways that promote cellular proliferation and<br />

angiogenesis. Furthermore, HIF is associated with proliferation of endothelial cells and has been l<strong>in</strong>ked<br />

with <strong>in</strong>creased <strong>in</strong>cidence of hemangiosarcoma <strong>in</strong> mouse. The work described here evaluates methods for<br />

detect<strong>in</strong>g tissue hypoxia and explores the potential role of hypoxia <strong>in</strong> 2-BE <strong>in</strong>duced hemangiosarcoma<br />

<strong>in</strong> mouse liver. B6C3F1 mice were subjected to either 6-8% whole-body hypoxia for ~2 hrs or to daily<br />

doses of 900 mg/kg 2-BE for 1 or 7 days. Serum EPO and local tissue hypoxia endpo<strong>in</strong>ts (Hypoxyprobe,<br />

stabilized HIF-1α, gene expression) were measured and compared. Elevations <strong>in</strong> serum EPO were<br />

observed follow<strong>in</strong>g both acute hypoxia and 2-BE treatment. Slight <strong>in</strong>creases <strong>in</strong> Hypoxyprobe sta<strong>in</strong><strong>in</strong>g and<br />

stabilized nuclear HIF-1α were detected <strong>in</strong> tissues from both hypoxia and 2-BE treated mice. In addition, a<br />

comparison of hepatic gene expression changes for the two treatments showed a large overlap <strong>in</strong> expression<br />

profiles among the hypoxia and 2-BE treated mice. We conclude that 2-BE causes local hypoxia <strong>in</strong> tissues<br />

associated with hemangiosarcoma <strong>in</strong> mouse. This local tissue hypoxia may contribute to a growth factor<br />

environment coupled with the macrophage effects previously reported that stimulates endothelial cell<br />

proliferation and hemangiosarcoma.<br />

4. 1N 6 -etheno-2’-Deoxyadenos<strong>in</strong>e (εda) <strong>in</strong> Weanl<strong>in</strong>g and adult rats<br />

exposed to 13 C 2 -V<strong>in</strong>yl Chloride by <strong>in</strong>halation<br />

L<strong>in</strong>a Gao 1 , Patricia Upton 1 , Gunnar Boysen 1 , Darrell W<strong>in</strong>sett 2 , Gary Hatch 2 , and James Swenberg 1<br />

1<br />

Department of Environmental Sciences and Eng<strong>in</strong>eer<strong>in</strong>g, University of North Carol<strong>in</strong>a, Chapel Hill, NC,<br />

United States<br />

2<br />

U.S. EPA, NHEERL, Research Triangle Park, NC, United States<br />

V<strong>in</strong>yl chloride (VC) is classified as a human carc<strong>in</strong>ogen by IARC, which can <strong>in</strong>duce angiosarcoma of<br />

the liver, a rare cancer <strong>in</strong> humans. As a mutagen <strong>in</strong> a number of organisms, VC ma<strong>in</strong>ly <strong>in</strong>duces base-pair<br />

substitution mutations by form<strong>in</strong>g DNA adduct. Through metabolic activation by CYP2E1, chloroethylene<br />

oxide, the ultimate carc<strong>in</strong>ogenic metabolite of VC, is formed which can react with DNA directly. εdA is one<br />

of the DNA adducts that formed follow<strong>in</strong>g VC exposure; however, it is also formed endogenously by lipid<br />

peroxidation. As one of the promutagenic etheno nucleosides, it can <strong>in</strong>duce εA→G transitions, as well as<br />

εA→T and εA→C tranversions. In order to accurately determ<strong>in</strong>e the formation and repair of εdA <strong>in</strong>duced<br />

by VC, adult and weanl<strong>in</strong>g Sprague-Dawley rats were exposed to 1100 ppm [ 13 C 2 ]-VC for 5d (6h/day). The<br />

weanl<strong>in</strong>g animals were sacrificed 2h post exposure. The adults were sacrificed 2h, 2, 4, and 8 weeks post<br />

47<br />

(Cont<strong>in</strong>ued on next page)


exposure. A sensitive nanoUPLC-MS/MS assay was developed to detect both endogenous [ 12 C 2 ]-εdA and<br />

exogenous [ 13 C 2 ]-εdA <strong>in</strong> DNA. The results show that [ 13 C 2 ]-εdA is detected <strong>in</strong> the exposed adult group and<br />

the exposed weanl<strong>in</strong>g group at the end of exposure, but that it was below detection levels <strong>in</strong> the exposed<br />

adult recovery groups. εdA is therefore rapidly repaired, which agrees with previous studies on the half-life<br />

of εdA (


6. evaluation of the effects of the ppar-gamma agonist troglitazone on<br />

Cytotoxicity and mitogenesis of endothelial Cells: Differences between<br />

human and mouse<br />

Satoko Kakiuchi-Kiyota 1 , Rakesh K. S<strong>in</strong>gh 1 , Joseph A. Vetro 2 , Michelle L. Varney 1 , Huai-Yun Han 2 ,<br />

Shugo Suzuki 1 , Karen L. Penn<strong>in</strong>gton 1 , Lora L. Arnold 1 , and Samuel M. Cohen 1<br />

1<br />

Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United<br />

States<br />

2<br />

Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center,<br />

Omaha, NE, United States<br />

Long-term treatment with high doses of troglitazone, a thiazolid<strong>in</strong>edione that b<strong>in</strong>ds and activates PPARγ,<br />

<strong>in</strong>creased the <strong>in</strong>cidence of hemangiosarcomas <strong>in</strong> various tissues <strong>in</strong> mice. We observed <strong>in</strong>creased endothelial<br />

cell (EC) proliferation <strong>in</strong> the liver and brown and white adipose tissues <strong>in</strong> female B6C3F1 mice treated<br />

with 400 and 800 mg/kg for 4 weeks. The purpose of this study was to determ<strong>in</strong>e if troglitazone directly<br />

<strong>in</strong>creases EC proliferation of an immortalized human microvascular endothelial cell l<strong>in</strong>e (HMEC1) and<br />

conditionally immortalized mouse microvascular endothelial cells isolated from the mammary fat pad of<br />

Immortomice (MFP MVEC). Cytotoxicity was observed <strong>in</strong> a dose dependent manner <strong>in</strong> both HMEC1 and<br />

MFP MVEC treated with high doses of troglitazone. The LC50 on day 3 was 17.4 μM <strong>in</strong> HMEC1 and<br />

92.2 μM <strong>in</strong> MFP MVEC. No changes of viability were observed when HMEC1, cultured with reduced<br />

concentrations of growth factors, were treated with low doses of troglitazone (0.001, 0.1, 1, 10, and 20<br />

μM) for 1, 3 and 6 days. In contrast, when MFP MVEC, cultured with a reduced concentration (1%) of<br />

FBS, were treated with troglitazone (0.1, 1, 5, 10, 25, and 50 μM) for 1, 3, and 6 days, the viability of cells<br />

treated with low doses of troglitazone was significantly <strong>in</strong>creased compared with cells cultured without<br />

troglitazone. To determ<strong>in</strong>e the effect of troglitazone on DNA synthesis <strong>in</strong> MFP MVEC cultured with 1%<br />

FBS, a tritiated thymid<strong>in</strong>e <strong>in</strong>corporation assay was performed. Cell proliferation was reduced from day 1<br />

when MFP MVEC were cultured without troglitazone. In contrast, low doses of troglitazone ma<strong>in</strong>ta<strong>in</strong>ed<br />

DNA synthesis on day 3 even with reduced growth factors. To <strong>in</strong>vestigate the effect on apoptosis, MFP<br />

MVEC cultured with 1% FBS were treated with low doses of troglitazone for 3 days, and the percentage<br />

of apoptotic cells was determ<strong>in</strong>ed by flow cytometry us<strong>in</strong>g Annex<strong>in</strong> V and PI sta<strong>in</strong><strong>in</strong>g. Compared with<br />

non-treated cells, low doses of troglitazone reduced the number of apoptotic cells, especially late apoptotic<br />

cells. In summary, 1) mouse microvascular EC may be more resistant to high doses of troglitazone<br />

compared with human microvascular EC; 2) under the condition of reduced growth factors, troglitazone<br />

may <strong>in</strong>crease the viability of mouse microvascular EC by <strong>in</strong>creased cell proliferation and suppression of<br />

apoptosis. In conclusion, troglitazone may <strong>in</strong>crease EC proliferation directly <strong>in</strong> mice.<br />

49<br />

(Cont<strong>in</strong>ued on next page)


7. mechanistic studies of polyhexamethylene biguanide (phmb)<br />

Carc<strong>in</strong>ogenicity<br />

Lisa M. Kamendulis 1 , X<strong>in</strong>zhu Pu 1 , Steven Barbee 2 , and Zem<strong>in</strong> Wang 1<br />

1 Department of Pharmacology and Toxicology, Center for Environmental Health Indiana University School<br />

of Medic<strong>in</strong>e, Indianapolis, IN, United States<br />

2 Arch Chemicals Inc, Cheshire, CT, United States<br />

Chronic dietary exposure to polyhexamethylene biguanide (PHMB) resulted <strong>in</strong> an <strong>in</strong>creased <strong>in</strong>cidence of<br />

liver hemangiosarcomas <strong>in</strong> C57/Bl6 mice (4/55 <strong>in</strong> control, 20/55 <strong>in</strong> 4000 ppm group). The mechanism for<br />

liver hemangiosarcoma <strong>in</strong>duction is not understood, but s<strong>in</strong>ce PHMB does not <strong>in</strong>duce mutations or produce<br />

genotoxicity, these tumors are produced through non-DNA reactive mechanisms. The present studies<br />

evaluated the dose-response profile of PHMB on endothelial cell growth. C57/Bl6 mice were exposed to<br />

0, 100, 200, 400, 1200, and 4000 ppm PHMB for 7, 14, and 28 days. PHMB did not <strong>in</strong>duce hepatotoxicity<br />

(evidenced by ALT and AST) at any dose or time evaluated. Rates of DNA synthesis, quantified us<strong>in</strong>g BrdU<br />

immunohistochemistry, showed that 4000 ppm PHMB decreased DNA synthesis after 7 and 14 days, a<br />

f<strong>in</strong>d<strong>in</strong>g that may be related to the decreased bodyweight, food utilization, and th<strong>in</strong>n<strong>in</strong>g of the <strong>in</strong>test<strong>in</strong>es seen<br />

at this dose. At 28 days, PHMB <strong>in</strong>creased DNA synthesis <strong>in</strong> a dose-responsive manner (~1.5- and 1.6-fold<br />

over control at 1200 and 4000 ppm), with no <strong>in</strong>creases <strong>in</strong> this endpo<strong>in</strong>t at or below 400 ppm. In vitro, non<br />

cytotoxic concentrations of PHMB (0-1ppm) did not <strong>in</strong>crease endothelial cell DNA synthesis. Concomitant<br />

with the <strong>in</strong>creased liver tumors <strong>in</strong> the 2-year study was an <strong>in</strong>crease <strong>in</strong> Kupffer cell pigmentation,<br />

suggest<strong>in</strong>g that this cell type may be activated and participate <strong>in</strong> PHMBs carc<strong>in</strong>ogenic response. To test<br />

this, DNA synthesis was measured <strong>in</strong> endothelial cells co-cultured with macrophages. PHMB (0-1ppm)<br />

did not <strong>in</strong>crease DNA synthesis <strong>in</strong> endothelial cells co-cultured with macrophages. Accompany<strong>in</strong>g the<br />

gastro<strong>in</strong>test<strong>in</strong>al effects caused by PHMB <strong>in</strong> vivo was an <strong>in</strong>crease <strong>in</strong> plasma endotox<strong>in</strong> by 1200 and 4000<br />

ppm PHMB after 28 days. S<strong>in</strong>ce endotox<strong>in</strong> is a known activator of macrophages, and PHMB did not<br />

produce direct effects on endothelial cell growth <strong>in</strong> vitro, these results collectively suggest that PHMB<br />

elicits effects on endothelial cell growth through an <strong>in</strong>direct effect, perhaps <strong>in</strong>volv<strong>in</strong>g endotox<strong>in</strong>-mediated<br />

activation of macrophages.<br />

8. Causal Network Model<strong>in</strong>g Identifies Hypoxia-Induced Inflammation as a<br />

potential Contributor to 2-butoxyethanol-<strong>in</strong>duced hemangiosarcomas<br />

Daphna Laifenfeld 1 , Annalyn Gilchrist 1 , Sean Eddy 1 , Sloane Furniss 1 , Keith Elliston 1 ,<br />

Petra Koza-Taylor 2 , Chris Somps 2 , Kay Criswell 2 , Jon Cook 2 , and Michael Lawton 2<br />

1 Genstruct Inc., Cambridge, MA, United States<br />

2 Pfizer Global Research and Development, Groton, CT, United States<br />

Hemangiosarcomas, tumors of endothelial cells (ECs), are rare <strong>in</strong> humans, but occur <strong>in</strong> mice spontaneously<br />

and <strong>in</strong> response to multiple therapeutic compounds, and can therefore impact drug approval processes.<br />

In order to understand the relevance of compound-<strong>in</strong>duced hemangiosarcomas <strong>in</strong> mice to human risk,<br />

a better understand<strong>in</strong>g of the molecular mechanisms lead<strong>in</strong>g to hemangiosarcomas, and their species<br />

specificity, is required. The present study identified molecular networks affected by hypoxia and capable<br />

of caus<strong>in</strong>g EC proliferation, which can ultimately result <strong>in</strong> hemangiosarcomas. We assessed the molecular<br />

response to three treatments <strong>in</strong> mice: 1. baclofen, which <strong>in</strong>duces respiratory suppression that can result<br />

<strong>in</strong> hypoxia, 2. hypoxia treatment, and 3. 2-Butoxyethanol (2BE), an <strong>in</strong>dustrial solvent that <strong>in</strong>creases<br />

50


liver hemangiosarcomas <strong>in</strong> mice. The molecular response to each was characterized by Causal Network<br />

Model<strong>in</strong>g, a scalable, computable framework for model<strong>in</strong>g biological networks us<strong>in</strong>g causality that<br />

<strong>in</strong>tegrates “omic” data with new and exist<strong>in</strong>g scientific knowledge. Causal Network Model<strong>in</strong>g us<strong>in</strong>g gene<br />

expression data from each treatment was used together with the Human Knowledge Assembly model to<br />

def<strong>in</strong>e networks and mechanisms that can lead to EC proliferation and hemangiosarcoma. The results of this<br />

study confirm that respiratory suppression <strong>in</strong>duced by baclofen leads to hypoxia and downstream effects<br />

<strong>in</strong> the liver. In addition, an <strong>in</strong>flammatory response was shared between all three models: the two hypoxiacaus<strong>in</strong>g<br />

treatments, hypoxia and baclofen, and the hemangiosarcoma caus<strong>in</strong>g agent, 2BE, implicat<strong>in</strong>g<br />

<strong>in</strong>flammation as a key component of hypoxia lead<strong>in</strong>g to EC proliferation and hemangiosarcoma.<br />

Inflammation leads to EC proliferation via <strong>in</strong>duction of growth factors and ROS, and Causal Network<br />

Model<strong>in</strong>g supports early prote<strong>in</strong> <strong>in</strong>duction of Fgf2 (fibroblast growth factor 2) and Egf (epidermal growth<br />

factor) <strong>in</strong> response to both baclofen and hypoxia treatments. In addition, Causal Network Model<strong>in</strong>g<br />

po<strong>in</strong>ts to macrophage activation <strong>in</strong> the three treatments, which can result <strong>in</strong> <strong>in</strong>creased EC proliferation.<br />

Macrophage activation lead<strong>in</strong>g to release of ROS has been previously implicated <strong>in</strong> the 2BE-<strong>in</strong>duction<br />

of hemangiosarcoma. Overall, the results implicate an <strong>in</strong>flammatory response <strong>in</strong> the <strong>in</strong>duction of EC<br />

proliferation and hemangiosarcoma <strong>in</strong> response to 2BE <strong>in</strong> the liver, and suggest that this mechanism may be<br />

downstream of RBC hemolysis-<strong>in</strong>duced hypoxia. Increased vulnerability of mice to hypoxia due to vitam<strong>in</strong><br />

E deficiency which impairs the ability of the mouse to protect aga<strong>in</strong>st hypoxic sequelae, such as ROS, and<br />

may therefore contribute to the species specificity of hemangiosarcomas.<br />

9. molecular Dosimetry of the V<strong>in</strong>yl Chloride-<strong>in</strong>duced Dna adduct,<br />

7-oxoethylguan<strong>in</strong>e<br />

Esra Mutlu 1,2 , Yo-Chan Jeong 1 , Leonard Coll<strong>in</strong>s 2 , Patricia B. Upton 2 , Darrell W<strong>in</strong>sett 3 , Gary Hatch 3 ,<br />

and James A. Swenberg 1,2<br />

1 Department of Environmental Sciences, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

2 The Curriculum <strong>in</strong> Toxicology, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

3 U.S. EPA NHEERL, Research Triangle Park, NC, United States<br />

V<strong>in</strong>yl Chloride (VC) is an <strong>in</strong>dustrial chemical and known genotoxic carc<strong>in</strong>ogen to animals and humans,<br />

which results <strong>in</strong> liver hemangiosarcoma with high exposures (>50 ppm). VC is also found <strong>in</strong> superfund<br />

sites as a result of microbial metabolism of TCE and PCE. Our research developed a new, more sensitive<br />

LC-MS/MS method to analyze the major DNA adduct of VC, 7-oxoethylguan<strong>in</strong>e (7-OEG). We applied<br />

this method to analyze tissues from both adult and weanl<strong>in</strong>g rats exposed to 1100 ppm [ 13 C 2 ]-VC for 5<br />

days. After neutral thermal hydrolysis the 7-OEG was derivatized with O- t butylhydroxylam<strong>in</strong>e to an oxime<br />

adduct followed by LC-MS/MS analysis with the limit of detection of 11.4±7.1 adducts/10 8 guan<strong>in</strong>e. In this<br />

study we determ<strong>in</strong>ed both endogenous and exogenous amounts of 7-OEG <strong>in</strong> liver, lung, kidney, spleen and<br />

testis DNA from adult and weanl<strong>in</strong>g rats exposed to [ 13 C 2 ]-VC. The presence of endogenous 7-OEG was<br />

demonstrated for the first time. The data from liver DNA was 0.2±0.1 adducts/10 5 guan<strong>in</strong>e of endogenous<br />

7-OEG and 10.4±2.3 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG; from lung 0.02±0.01 adducts/10 5 guan<strong>in</strong>e<br />

of endogenous 7-OEG and 13.9±0.2 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG; from kidney 0.07±0.04<br />

adducts/10 5 guan<strong>in</strong>e of endogenous 7-OEG and 2.8±0.7 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG;<br />

from testis 1.44±1.14 adducts/10 5 guan<strong>in</strong>e of endogenous 7-OEG and 0.03±0.01 adducts/10 5 guan<strong>in</strong>e of<br />

exogenous 7-OEG; from spleen 0.5±0.1 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG for adult rats (n=4). The<br />

data from weanl<strong>in</strong>g rats exposed to [ 13 C 2 ]-VC <strong>in</strong> liver was 0.08±0.05 adducts/10 5 guan<strong>in</strong>e of endogenous<br />

7-OEG and 29.9±12.9 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG; <strong>in</strong> lung 0.04±0.01 adducts/10 5 guan<strong>in</strong>e of<br />

51<br />

(Cont<strong>in</strong>ued on next page)


endogenous 7-OEG and 8.3±1.9 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG; <strong>in</strong> testis 0.32±0.11 adducts/10 5<br />

guan<strong>in</strong>e of endogenous 7-OEG and 0.06±0.02 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG; <strong>in</strong> kidney<br />

6.6±1.2 adducts/10 5 guan<strong>in</strong>e of exogenous 7-OEG; <strong>in</strong> spleen 0.8±0.1 adducts/10 5 guan<strong>in</strong>e of exogenous<br />

7-OEG (n=8). Studies on the persistence of 7-OEG have been analyzed from adult rats sacrificed 2, 4, and<br />

8 wks post exposure to [ 13 C 2 ]-VC to establish the half life of 7-OEG <strong>in</strong> liver (4.1 days) and lung (4 days).<br />

Prelim<strong>in</strong>ary data from bra<strong>in</strong> DNA demonstrated small amounts of exogenous 7-OEG <strong>in</strong> adult rats exposed<br />

to [ 13 C 2 ]-VC (0.15±0.06 adducts/10 5 guan<strong>in</strong>e, n=5).<br />

10. a new lC-ms/ms method for the Detection of the V<strong>in</strong>yl Chloride<strong>in</strong>duced<br />

Dna adduct N 2 ,3-ethenoguan<strong>in</strong>e<br />

Esra Mutlu 1,2 , Leonard Coll<strong>in</strong>s 2 , Patricia B. Upton 2 , Matthew D. Stout 1 , Darrell W<strong>in</strong>sett 3 ,<br />

Gary Hatch 3 , and James A. Swenberg 1,2<br />

1 Department of Environmental Sciences, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

2 The Curriculum <strong>in</strong> Toxicology, University of North Carol<strong>in</strong>a, Chapel Hill, NC, United States<br />

3 U.S. EPA, NHEERL, Research Triangle Park, NC, United States<br />

In the 1970s it was shown that high level exposure to v<strong>in</strong>yl chloride (VC) caused liver angiosarcoma<br />

<strong>in</strong> VC workers. Early reports also suggested that exposure to VC may be associated with bra<strong>in</strong> tumors.<br />

However, we had demonstrated that N 2 ,3-ethenoguan<strong>in</strong>e (εG) was not detectable <strong>in</strong> the bra<strong>in</strong>s of adult<br />

rats exposed to 1100 ppm VC. This research cont<strong>in</strong>ues with development of a new LC-MS/MS method<br />

for analyz<strong>in</strong>g εG <strong>in</strong> tissues from both adult and weanl<strong>in</strong>g rats exposed to 1100 ppm [ 13 C 2 ]-VC for 5 days<br />

and 1100 ppm VC for 1 day. Our new εG assay utilized an HPLC enrichment step after neutral thermal<br />

hydrolysis, followed by quantitation by LC-HESI + -MS/MS. Separation was performed on a 10mm i.d. C18<br />

column while monitor<strong>in</strong>g 176→176 m/z for endogenous εG, 178→178 m/z for exogenous [ 13 C 2 ]-εG, and<br />

182→182 m/z for the <strong>in</strong>ternal standard <strong>in</strong> SRM mode. Prelim<strong>in</strong>ary data us<strong>in</strong>g the HPLC-LC-MS/MS assay<br />

demonstrated 2.3±0.7 adducts/10 8 guan<strong>in</strong>e of endogenous εG <strong>in</strong> calf thymus DNA, which is consistent<br />

with previous studies. Cont<strong>in</strong>u<strong>in</strong>g data analysis has determ<strong>in</strong>ed both endogenous and exogenous amounts<br />

of the promutagenic DNA adduct εG <strong>in</strong> liver, lung, and kidney DNA from rats exposed to [ 13 C 2 ]-VC<br />

for 5 days. The data from liver DNA was 4.1±2.8 adducts/10 8 guan<strong>in</strong>e of endogenous and 19.0±4.9<br />

adducts/10 8 guan<strong>in</strong>e of exogenous εG; from lungs 8.4±2.8 adducts/10 8 guan<strong>in</strong>e of endogenous and 7.4±0.5<br />

adducts/10 8 guan<strong>in</strong>e of exogenous εG; from kidneys 5.9±3.3 adducts/10 8 guan<strong>in</strong>e of endogenous and<br />

5.7±2.1 adducts/10 8 guan<strong>in</strong>e of exogenous εG for adult rats (n=4). Additionally, the data from weanl<strong>in</strong>g<br />

rats demonstrated higher adduct levels due to greater metabolism; from lung 5.3±2.4 adducts/10 8 guan<strong>in</strong>e<br />

of endogenous and 15.8±3.6 adducts/10 8 guan<strong>in</strong>e of exogenous εG, from kidney 4.1±0.9 adducts/10 8<br />

guan<strong>in</strong>e of endogenous and 12.9±0.4 adducts/10 8 guan<strong>in</strong>e of exogenous εG was determ<strong>in</strong>ed (n=8). The half<br />

life of εG was calculated based on post exposure data: <strong>in</strong> liver and lung 150 days and <strong>in</strong> kidney 75 days.<br />

Additionally, we will determ<strong>in</strong>e the formation εG <strong>in</strong> the bra<strong>in</strong>s and testis of [ 13 C 2 ]-VC exposed weanl<strong>in</strong>g<br />

rats to establish whether or not the exogenous DNA adduct is formed <strong>in</strong> those tissues follow<strong>in</strong>g exposure<br />

to VC.<br />

52


11. mutations <strong>in</strong> the pten, tek, Vhlh and kdr/Vegfr2 genes are not<br />

susceptibility factors for spontaneous hemangiosarcoma Development <strong>in</strong><br />

mice<br />

Chunhua Q<strong>in</strong> 1 , Alessandra Tosol<strong>in</strong>i 1 , and Richard D. Storer 1<br />

1 Safety Assessment, Merck Research Laboratories, West Po<strong>in</strong>t, PA, United States<br />

Hemangiosarcoma is one of the most frequent spontaneous malignancies <strong>in</strong> mice, with a historical<br />

<strong>in</strong>cidence of up to 12% <strong>in</strong> B6C3F1 mice. Previous studies showed that mutations <strong>in</strong> the p53 and ras<br />

genes, which play a role <strong>in</strong> human hemangiosarcoma development, are not essential for vascular tumor<br />

development <strong>in</strong> mice (Duddy, S.K., et al., Toxicology and Applied Pharmacology 156 (2), 106-112, 1999;<br />

Duddy, S.K., et al., Toxicology and Applied Pharmacology 160 (2): 133-140, 1999). The cause of the<br />

<strong>in</strong>creased susceptibility <strong>in</strong> mice rema<strong>in</strong>s elusive. Dysregulation of the molecules <strong>in</strong> at least 3 of the key<br />

signal<strong>in</strong>g pathways <strong>in</strong>volved <strong>in</strong> vasculogenesis and angiogenesis are associated with vascular tumors;<br />

these <strong>in</strong>clude (1) the Von Hippel-L<strong>in</strong>dau gene (VHL or VHLH for VHL homolog) and hypoxia-<strong>in</strong>ducible<br />

factor (HIF) pathway, (2) the vascular endothelial growth factor receptor 2 (VEGFR-2 or KDR) and<br />

phosphatase and tes<strong>in</strong> homolog (PTEN) pathway, and (3) the Tie2/Tek signal<strong>in</strong>g pathway. We hypothesized<br />

that polymorphisms and/or spontaneous mutations <strong>in</strong> these genes may play a role <strong>in</strong> species, stra<strong>in</strong>, and<br />

or <strong>in</strong>dividual animal susceptibility <strong>in</strong> mice to the development of hemangiosarcoma. In this study, we<br />

sequenced 20 outbred CD1 mice, 1 C3H mouse, 1 C57BL mouse, 1 129 stem cell l<strong>in</strong>e, and 3 spontaneous<br />

hemangiosarcoma tumors derived from p53 -/- tumors for mutations or polymorphisms <strong>in</strong> critical doma<strong>in</strong>s/<br />

exons of these genes. Our results showed that there were no mutations for these genes <strong>in</strong> the animals<br />

tested albeit some non-cod<strong>in</strong>g or silent nucleotide base polymorphisms were observed. We thus conclude<br />

that mutations <strong>in</strong> critical doma<strong>in</strong>s of the PTEN, Tek, Vhlh and Kdr/Vegfr2 are not associated with the<br />

development of spontaneous hemangiosarcoma <strong>in</strong> mice.<br />

12. DNA Methylation Changes Identified <strong>in</strong> Spontaneous<br />

hemangiosarcomas and pparγ agonist-treated mouse endothelial Cells<br />

<strong>in</strong> a Genome-Wide Methylation Profil<strong>in</strong>g Study<br />

Chunhua Q<strong>in</strong> 1 , Kathy A. McNulty 1 , Thomas L. Fare 1 , Alan Ng 1 , and Richard D. Storer 1<br />

1 Safety Assessment, Merck Research Laboratories, West Po<strong>in</strong>t, PA, United States<br />

Ten out of 12 peroxisome proliferator activated receptor (PPAR) agonists developed for treat<strong>in</strong>g Type II<br />

Diabetes <strong>in</strong>duced hemangiosarcomas <strong>in</strong> mice. Most of these agonists did not exhibit genotoxicity <strong>in</strong> the<br />

genotoxicity battery test<strong>in</strong>g, and the tumorigenic effects of this class are considered to be act<strong>in</strong>g through<br />

a non-genotoxic mode-of-action. DNA methylation change is one form of epigenetic change that has<br />

recently been characterized as a hallmark of cancer development and an important mode-of-action for<br />

non-genotoxic rodent carc<strong>in</strong>ogens such as phenobarbital. To explore the effect of PPARγ agonists on DNA<br />

methylation, we evaluated the Epigenomics Differential Methylation Hybridization (DMH) microarray<br />

platform for genome-wide DNA methylation studies <strong>in</strong> mice. We determ<strong>in</strong>ed the DNA methylation<br />

changes <strong>in</strong> MS1 mouse endothelial cells treated with PPARγ agonists troglitazone and rosiglitazone. In<br />

MS1 cells, the Epigenomics DMH platform identified 1127 regions with significant (p


liver necrosis/cell death). We also analysed the methylation changes <strong>in</strong> 3 spontaneous hemangiosarcomas<br />

(control: kidney) from p53 -/- mice and identified 63 candidate biomarkers with significant (p


14. Development of flow Cytometry and laser scann<strong>in</strong>g Cytometry<br />

methods to assess endothelial Cell proliferation <strong>in</strong> liver, spleen, and bone<br />

marrow of 2-butoxyethanol-treated mice<br />

Sharon Sokolowski 1 , Amy Shen 1 , Shuou Zhao 1 , Chunli Huang 1 , Susan Eddy 1 , Marc Roy 1 ,<br />

Amy Jakowski 1 , Cather<strong>in</strong>e Tabor 1 , and Leslie Obert 1<br />

1 Pfizer Global Research and Development, Groton, CT, United States<br />

The purpose of this work was to develop automated methods to evaluate endothelial cell proliferation <strong>in</strong><br />

the bone marrow, spleen, and liver of mice after treatment with 2-butoxyethanol (2-BE). While chronic<br />

exposure to 2-BE is known to <strong>in</strong>duce liver hemangiosarcoma formation <strong>in</strong> male mice, acute exposure<br />

for 7-14 days has been shown to <strong>in</strong>crease DNA synthesis <strong>in</strong> liver s<strong>in</strong>usoidal endothelial cells. Increased<br />

endothelial cell proliferation may be one essential component contribut<strong>in</strong>g to the <strong>in</strong>creased <strong>in</strong>cidence of<br />

hemangiosarcoma formation <strong>in</strong> the liver of mice after treatment with 2-BE. Sensitive automated methods<br />

capable of detect<strong>in</strong>g alterations <strong>in</strong> the proliferation rate of endothelial cells <strong>in</strong> target organs may provide an<br />

early surrogate biomarker for tumor formation. Us<strong>in</strong>g 2-BE as a positive control compound, male B6C3F1<br />

mice were dosed by oral gavage once daily for 7 consecutive days with either 900 mg/kg 2-BE or vehicle.<br />

To assess cellular proliferation, mice were implanted with osmotic pumps conta<strong>in</strong><strong>in</strong>g either 20 mg/mL<br />

5-bromo-2'-deoxyurid<strong>in</strong>e (BrdU) or 5-ethynyl-2'-deoxyurid<strong>in</strong>e (EdU). At study term<strong>in</strong>ation, bone marrow,<br />

spleen, and liver were harvested and processed accord<strong>in</strong>gly for either flow cytometric and/or morphometric<br />

evaluation. Endothelial cells were labeled us<strong>in</strong>g a s<strong>in</strong>gle marker or comb<strong>in</strong>ations of markers (CD31,<br />

CD105, CD45, VEGFR2). Proliferat<strong>in</strong>g cells were identified via EdU Click-IT ® chemistry, anti-BrdU<br />

immunohistochemistry (IHC), or anti-Ki67 IHC. Flow cytometric analysis of cell populations isolated from<br />

bone marrow, spleen and liver revealed fold <strong>in</strong>creases of approximately 1.6, 7.3, and 1.2, respectively, <strong>in</strong><br />

proliferat<strong>in</strong>g endothelial cells <strong>in</strong> 2-BE-treated mice when compared to controls. Morphometric analysis of<br />

liver s<strong>in</strong>usoid endothelial cells us<strong>in</strong>g laser scann<strong>in</strong>g cytometry measured a 1.2 fold <strong>in</strong>crease <strong>in</strong> proliferat<strong>in</strong>g<br />

endothelial cells <strong>in</strong> 2-BE-treated mice compared to controls. Although the fold <strong>in</strong>crease <strong>in</strong> liver s<strong>in</strong>usoidal<br />

endothelial cell proliferation for the current study was less than the 2-fold <strong>in</strong>crease previously reported, the<br />

similar fold changes <strong>in</strong> liver s<strong>in</strong>usoidal endothelial proliferation <strong>in</strong> the current experiments suggests that<br />

these two <strong>in</strong>dependent automated methods of analysis can be used to assess endothelial cell proliferation.<br />

In addition, this study demonstrates that 2-BE treatment of mice <strong>in</strong>duces <strong>in</strong>creased endothelial proliferation<br />

<strong>in</strong> bone marrow and spleen. In conclusion, automated methods to assess endothelial cell proliferation have<br />

been established to evaluate drug- and/or chemical-<strong>in</strong>duced alterations <strong>in</strong> endothelial cell proliferation rate<br />

<strong>in</strong> the bone marrow, spleen and liver of mice us<strong>in</strong>g the positive control, 2-BE.<br />

55


notes<br />

56


57<br />

attenDees


The Society of Toxicology is<br />

committed to honor<strong>in</strong>g the privacy<br />

of those who attend our meet<strong>in</strong>gs.<br />

This <strong>in</strong>formation is <strong>in</strong>tended<br />

for the personal use of the SOT<br />

CCT registrants and should not<br />

be reproduced or used for any<br />

commercial purpose.<br />

58


a<br />

Mart<strong>in</strong>s Oluwadare Adeyemo<br />

Daiichi Sankyo Company Limited<br />

399 Thornall Street<br />

Edison, NJ 08837<br />

United States<br />

madeyemo@dsus.com<br />

(732) 590-4883<br />

Krishna P. Allamneni<br />

Genentech Inc<br />

1 DNA Way<br />

MS 240B<br />

South San Francisco, CA 94080<br />

United States<br />

allamneni.krishna@gene.com<br />

(650) 467-3794<br />

Ayaad W. Assaad<br />

U.S. EPA/Office of Pesticide Programs<br />

1200 Pennsylvania Avenue, NW<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

assaad.ayaad@epa.gov<br />

(703) 305-0314<br />

b<br />

Steven J. Barbee<br />

Arch Chemicals Inc<br />

350 Knotter Drive<br />

Cheshire, CT 06410<br />

United States<br />

sjbarbee@archchemicals.com<br />

(203) 271-4308<br />

Stanley Barone, Jr.,<br />

U.S. EPA/Office of Research and Development<br />

1200 Pennsylvania Avenue, NW<br />

MD8601P<br />

Wash<strong>in</strong>gton, DC 20006<br />

United States<br />

barone.stan@epa.gov<br />

(703) 347-8555<br />

attendees<br />

59<br />

Col<strong>in</strong> Berry<br />

Queen Mary, University of London<br />

1 College Gardens<br />

Dulwich, London SE21 7BE<br />

United K<strong>in</strong>gdom<br />

berry@dulwich98.freeserve.co.uk<br />

44 2082 990066<br />

Mart<strong>in</strong> Bopst<br />

Hoffmann La Roche Inc<br />

Non Cl<strong>in</strong>ical Drug Safety<br />

Build<strong>in</strong>g 73 101 B<br />

Basel 4070<br />

Switzerland<br />

mart<strong>in</strong>.bopst@roche.com<br />

61 6874 337<br />

Kathryn Bowenkamp<br />

Novartis Institutes for BioMedical Research<br />

One Health Plaza<br />

East Hanover, NJ 07936-1080<br />

United States<br />

kathryn.bowenkamp@novartis.com<br />

(862) 778-7189<br />

C<br />

Neil G. Carmichael<br />

ECETOC AISBL<br />

4 Avenue Van Nieuwenhuyse<br />

Box 6<br />

Brussels 1160<br />

Belgium<br />

neil.carmichael@ecetoc.org<br />

32 2675 3600<br />

Matthew Cave<br />

University of Louisville<br />

511 S. Floyd Street, Room 306<br />

Louisville, KY 40202<br />

United States<br />

drmattcave@aol.com<br />

(502) 852-6189


Samuel M. Cohen<br />

University of Nebraska Medical Center<br />

983135 Nebraska Medical Center<br />

Omaha, NE 68198-3135<br />

United States<br />

scohen@unmc.edu<br />

(402) 559-6388<br />

Jon C. Cook<br />

Pfizer Inc<br />

Eastern Po<strong>in</strong>t Road<br />

MS 8274 1222<br />

Groton, CT 06340<br />

United States<br />

jon.c.cook@pfizer.com<br />

(860) 715-2693<br />

George B. Corcoran<br />

Wayne State University<br />

259 Mack Avenue<br />

Eugene Applebaum College of Pharmacy Health<br />

Sciences<br />

Detroit, MI 48201<br />

United States<br />

corcoran@wayne.edu<br />

(313) 577-1737<br />

Kay A. Criswell<br />

Pfizer Global Research and Development<br />

2400 Eastern Po<strong>in</strong>t Road<br />

MS 8274-1424<br />

Groton, CT 06340<br />

United States<br />

kay.criswell@pfizer.com<br />

(860) 686-9430<br />

D<br />

Charles A. Dangler<br />

sanofi-aventis<br />

9 Great Valley Parkway<br />

Malyern, PA 19355<br />

United States<br />

charles.dangler@sanofi-aventis.com<br />

(610) 889-8907<br />

60<br />

Vicki L. Dellarco<br />

U.S. EPA<br />

1200 Pennsylvania Avenue, NW<br />

7509P<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

dellarco.vicki@epa.gov<br />

(703) 305-1803<br />

Gerard Descotes<br />

Fournier/Solvay<br />

42 rue Rouget-de-Lisle<br />

Suresnes 92150<br />

France<br />

gerard.descotes@solvay.com<br />

33 1462 58598<br />

Sanjivani B. Diwan<br />

U.S. EPA ORD/NCEA-W<br />

1200 Pennsylvania Avenue, NW<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

diwan.sanjivani@epa.gov<br />

(703) 305-7188<br />

Nancy G. Doerrer<br />

ILSI Health and Environmental Sciences Institute<br />

1156 Fifteenth Street, NW<br />

Second Floor<br />

Wash<strong>in</strong>gton, DC 20005<br />

United States<br />

ndoerrer@hesiglobal.org<br />

(202) 659-3306<br />

John Douglas Doherty<br />

U.S. EPA<br />

1200 Pennsylvania Avenue, NW<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

doherty.john@epa.gov<br />

(703) 620-3473


e<br />

Sean Eddy<br />

Genstruct<br />

One Alewife Center<br />

Cambridge, MA 02140<br />

United States<br />

seddy@genstruct.com<br />

(617) 547-5421<br />

Jeri El-Hage<br />

Aclairo Pharmaceutical Development Group<br />

1950 Old Gallows Road, Suite 300<br />

Vienna, VA 22182<br />

United States<br />

jelhage@aclairo.com<br />

(703) 506-6760 (310)<br />

Ronald Eydelloth<br />

3 Autumn Meadow Lane<br />

Malvern, PA 19355<br />

United States<br />

eydell@comcast.net<br />

(484) 320-8059<br />

f<br />

Susan P. Felter<br />

Procter & Gamble Company<br />

11810 E Miami River Road<br />

C<strong>in</strong>c<strong>in</strong>nati, OH 45252<br />

United States<br />

felter.sp@pg.com<br />

(513) 627-1958<br />

Lynn Flowers<br />

U.S. EPA<br />

1200 Pennsylvania Avenue, NW<br />

8601P<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

flowers.lynn@epa.gov<br />

(703) 347-8537<br />

Sab<strong>in</strong>e Francke-Carroll<br />

U.S. FDA/CFSAN HFS205<br />

5100 Pa<strong>in</strong>t Branch Parkway<br />

College Park, MD 20740<br />

United States<br />

sab<strong>in</strong>e.francke@fda.hhs.gov<br />

(301) 436-1308<br />

61<br />

Lois Freed<br />

U.S. FDA/CDER<br />

10903 New Hampshire Avenue<br />

W022 Rm 4380<br />

Silver Spr<strong>in</strong>g, MD 20993<br />

United States<br />

lois.freed@fda.hhs.gov<br />

(301) 796-1070<br />

Re<strong>in</strong>a Fuji<br />

Genentech<br />

1 DNA Way<br />

Mailstop 59<br />

South San Francisco, CA 94080<br />

United States<br />

fuji.re<strong>in</strong>a@gene.com<br />

(650) 467-1227<br />

g<br />

James Michael Gerhart<br />

Merial Limited<br />

3239 Satellite Boulevard<br />

Duluth, GA 30096-4640<br />

United States<br />

james.gerhart@merial.com<br />

(678) 638-3617<br />

Mark Gos<strong>in</strong>k<br />

Pfizer Inc.<br />

Eastern Po<strong>in</strong>t Road., MS 8274-1246<br />

Groton, CT 06340<br />

United States<br />

mark.m.gos<strong>in</strong>k@pfizer.com<br />

(860) 686-4468<br />

Maureen R. Gw<strong>in</strong>n<br />

U.S. EPA/EICG<br />

1200 Pennsylvania Avenue, NW<br />

Mailcode 8623P<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

gw<strong>in</strong>n.maureen@epa.gov<br />

(703) 347-8565


h<br />

Patricia Harlow<br />

U.S. FDA /CDER<br />

10903 New Hampshire Avenue<br />

Silver Spr<strong>in</strong>g, MD 20993<br />

United States<br />

patricia.harlow@fda.hhs.gov<br />

(301) 796-1082<br />

Olivia Harris<br />

NCEH/ATSDR/CDC<br />

4770 Buford Highway NE, F-61<br />

Atlanta, GA 30341-3717<br />

United States<br />

oharris@cdc.gov<br />

(770) 488-0597<br />

Kenneth L. Hast<strong>in</strong>gs<br />

sanofi-aventis<br />

4520 East West Highway<br />

Suite 210<br />

Bethesda, MD 20814<br />

United States<br />

kenneth.hast<strong>in</strong>gs@sanofi-aventis.com<br />

(301) 771-4267<br />

Heike Hellmold<br />

AstraZeneca R&D Sweden<br />

151 85 SE<br />

Sodertalje<br />

Sweden<br />

heike.hellmold@astrazeneca.com<br />

46 8553 2432<br />

Elizabeth Hofmann<br />

U.S. EPA<br />

1300 Pennsylvania Avenue, NW<br />

Wash<strong>in</strong>gton, DC 20004<br />

United States<br />

hofmann.lee@epa.gov<br />

(202) 564-6811<br />

Michael P. Holsapple<br />

ILSI Health and Environmental Sciences Institute<br />

1156 Fifteenth Street, NW<br />

Second Floor<br />

Wash<strong>in</strong>gton, DC 20005<br />

United States<br />

mholsapple@hesiglobal.org<br />

(202) 659-3306<br />

62<br />

j<br />

Abigail C. Jacobs<br />

U.S. FDA<br />

10903 New Hampshire Avenue<br />

Build<strong>in</strong>g 22 Room 6484<br />

Silver Spr<strong>in</strong>g, MD 20993<br />

United States<br />

abigail.jacobs@fda.hhs.gov<br />

(301) 796-0174<br />

Timothy Johnson<br />

Merck Research Laboratories<br />

WP45-319<br />

West Po<strong>in</strong>t, PA 19486<br />

United States<br />

timothy_johnson@merck.com<br />

(215) 652-4323<br />

k<br />

Satoko Kakiuchi-Kiyota<br />

University of Nebraska Medical Center<br />

983135 Nebraska Medical Center<br />

Omaha, NE 68918-3135<br />

United States<br />

skiyota@unmc.edu<br />

(402) 559-6157<br />

Lisa M. Kamendulis<br />

Indiana University School of Medic<strong>in</strong>e<br />

635 Barnhill Drive<br />

MS A507<br />

Indianapolis, IN 46202<br />

United States<br />

lkamendu@iupui.edu<br />

(317) 278-7823<br />

Gerald L. Kennedy, Jr.<br />

DuPont Haskell Laboratories<br />

1090 Elkton Road<br />

P.O. Box 50<br />

Newark, DE 19714-0050<br />

United States<br />

gerald.l.kennedy@usa.dupont.com<br />

(302) 366-5259


James E. Klaunig<br />

Indiana University School of Medic<strong>in</strong>e<br />

635 Barnhill Drive<br />

MS A 503<br />

Indianapolis, IN 46202<br />

United States<br />

jklauni@iupui.edu<br />

(317) 274-7824<br />

Mami Kouchi<br />

Da<strong>in</strong>ippon Sumitomo Pharma Co., Ltd.<br />

33-94 Enoki-cho<br />

Suita 564-0053<br />

Japan<br />

mami-kochi@ds-pharma.co.jp<br />

81 6633 75923<br />

Melissa Kramer<br />

U.S. EPA<br />

1200 Pennsylvania Avenue, NW<br />

MC 8105R<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

kramer.melissa@epa.gov<br />

(202) 564-8497<br />

l<br />

Dom<strong>in</strong>ique Lasserre-Bigot<br />

Bayer CropScience<br />

BP 153 335, Rue Dostoievski<br />

6903 Sophia Antipolis<br />

France<br />

dom<strong>in</strong>ique.lasserre-bigot@bayercropscience.com<br />

Michael P. Lawton<br />

Pfizer Inc<br />

Eastern Po<strong>in</strong>t Road<br />

Groton, CT 06340<br />

United States<br />

michael.lawton@pfizer.com<br />

(860) 441-1568<br />

63<br />

John Leighton<br />

U.S. FDA<br />

10903 New Hampshire Avenue<br />

Silver Spr<strong>in</strong>g, MD 20993<br />

United States<br />

John.Leighton@fda.hhs.gov<br />

(301) 796-2340<br />

Stuart Lev<strong>in</strong><br />

Takeda Global R & D<br />

675 N Field Drive<br />

Lake Forest, IL 60045<br />

United States<br />

stuart.lev<strong>in</strong>@tgrd.com<br />

(847) 582-4978<br />

Gerald Long<br />

Lilly Research Laboratories<br />

Drop Code 0434<br />

Lilly Corporate Center<br />

Indianapolis, IN 46285<br />

United States<br />

g.long@lilly.com<br />

(317) 277-4711<br />

m<br />

David Malarkey<br />

National Institute of Environmental Health Sciences<br />

Maildrop B3-06 P.O. Box 12233<br />

111 Alexander Drive<br />

RTP, NC 27709<br />

United States<br />

malarkey@niehs.nih.gov<br />

(919) 541-1745<br />

Mary K. Manibusan<br />

U.S. EPA<br />

1200 Pennsylvania Avenue, NW<br />

Mail Stop 7509P<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

manibusan.mary@epa.gov<br />

(703) 308-0025


Keith L. March<br />

Cryptic Masons Medical Research Foundation<br />

Director, Indiana Center for Vascular Biology<br />

& Medic<strong>in</strong>e<br />

Medical Research & Library, Room 442<br />

975 W. Walnut Street<br />

Indianapolis, IN 46202-5121<br />

United States<br />

kmarch@iupui.edu<br />

(317) 278-0130<br />

Craig McCla<strong>in</strong><br />

University of Louisville<br />

511 South Floyd St., Room 305<br />

Louisville, KY 40202<br />

United States<br />

craig.mccla<strong>in</strong>@louisville.edu<br />

(502) 852-6189<br />

Jos Mertens<br />

WIL Research Laboratories LLC<br />

1407 George Road<br />

Ashland, OH 44805<br />

United States<br />

jmertens@wilresearch.com<br />

(419) 289-8700<br />

Jaime F. Modiano<br />

College of Veter<strong>in</strong>ary Medic<strong>in</strong>e and<br />

Masonic Cancer<br />

455 VMC<br />

MMC6194<br />

1365 Gortner Avenue<br />

St. Paul, MN 55108<br />

United States<br />

modiano@umn.edu<br />

(612) 625-7436<br />

Mike Moore<br />

Covance Inc<br />

9200 Leesburg Pike<br />

Vienna, VA 22182-1699<br />

United States<br />

michael.moore@covance.com<br />

(703) 245-2200<br />

64<br />

Esra Mutlu<br />

University of North Carol<strong>in</strong>a Chapel Hill<br />

346C Rosenau Hall<br />

CB 7431<br />

Chapel Hill, NC 27599<br />

United States<br />

esra_mutlu@unc.edu<br />

(919) 966-6141<br />

o<br />

Leslie Ann Obert<br />

Pfizer<br />

P.O. Box 702<br />

Old Mystic, CT 06372<br />

United States<br />

leslie.obert@pfizer.com<br />

(860) 686-9443<br />

p<br />

Sang-ki Park<br />

U.S. Food and Drug Adm<strong>in</strong>istration<br />

5100 Pa<strong>in</strong>t Branch Parkway<br />

College Park, MD 20740<br />

United States<br />

Sang-ki.Park@fda.hhs.gov<br />

(301) 436-1287<br />

Terry S. Peters<br />

U.S. FDA/CDER<br />

10903 New Hampshire Avenue<br />

W022 Room 4376<br />

Silver Spr<strong>in</strong>g, MD 20993<br />

United States<br />

terry.peters@fda.hhs.gov<br />

(301) 796-0785<br />

Mart<strong>in</strong> A. Philbert<br />

University of Michigan<br />

School of Public Health<br />

1420 Wash<strong>in</strong>gton Heights<br />

Ann Arbor, MI 48109-2029<br />

United States<br />

philbert@umich.edu<br />

(734) 763-4523


John Mart<strong>in</strong> Pletcher<br />

Charles River Labs<br />

15 Worman’s Mill Court, Suite I<br />

Frederick, MD 21701<br />

United States<br />

jpletcher@us.crl.com<br />

(301) 624-2008<br />

Christopher Joseph Powell<br />

GlaxoSmithKl<strong>in</strong>e<br />

Park Road<br />

Herts SG12 0DP<br />

United K<strong>in</strong>gdom<br />

christopher.j.powell@gsk.com<br />

44 1920 469469<br />

Resha Mae Putzrath<br />

U.S. EPA<br />

1025 F Street, NW 3rd Floor<br />

MC1400F<br />

Wash<strong>in</strong>gton, DC 20004<br />

United States<br />

putzrath.resha@epa.gov<br />

(202) 343-9978<br />

r<br />

Kathleen C. Raffaele<br />

U.S. EPA<br />

1200 Pennsylvania Avenue, NW<br />

MC 8105R<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

raffaele.kathleen@epa.gov<br />

(703) 305-5664<br />

Lynnda L. Reid<br />

U.S. FDA/CDER<br />

Center for Drug Evaluation and Research<br />

10903 New Hampshire Avenue<br />

White Oak Build<strong>in</strong>g 22 Room 5388<br />

Silver Spr<strong>in</strong>g, MD 20993<br />

United States<br />

lynnda.reid@fda.hhs.gov<br />

(301) 796-0984<br />

65<br />

Jerry M. Rice<br />

Georgetown University Medical Center<br />

3800 Reservoir Road, NW<br />

Wash<strong>in</strong>gton, DC 20057-1465<br />

United States<br />

jr332@georgetown.edu<br />

(301) 986-0659<br />

Michael S. Rogers<br />

Harvard Medical School<br />

Karp Research Build<strong>in</strong>g 11.006H<br />

Boston, MA 02115<br />

United States<br />

michael.rogers@childrens.harvard.edu<br />

(617) 919-2252<br />

Nigel Oliver Roome<br />

sanofi-avantis<br />

2-8 Rue de Rouen<br />

Gargenville 93230<br />

France<br />

nigel.roome@sanofi-aventis.com<br />

33 1347 95840<br />

s<br />

Fumiko Sano<br />

Mitsubishi Tanabe Pharma Corporation<br />

1-1-1, Kazusakamatari<br />

Kisarazu 292-0818<br />

Japan<br />

sano.fumiko@mg.mt-pharma.co.jp<br />

(814) 385-2353 7<br />

Keiichiro Sato<br />

Takedo Pharmaceutical Company<br />

17-85 Jusohonmachi 2-chome<br />

Osaka 532-8686<br />

Japan<br />

sato_keiichiro@takeda.co.jp<br />

81 6630 06930<br />

Sh<strong>in</strong>ya Sehata<br />

Daiichi Sankyo Inc.<br />

399 Thormal Street<br />

Edison, NJ 08837<br />

United States<br />

ssehata@dsus.com<br />

(732) 590-4329


Leslie Sharkey<br />

University of M<strong>in</strong>nesota<br />

C339 VMC 1352 Boyd Ave<br />

St Paul, MN 55108<br />

United States<br />

shark009@umn.edu<br />

(651) 402-1261<br />

Chris John Somps<br />

Pfizer Inc<br />

Eastern Po<strong>in</strong>t Road<br />

Bldg 274<br />

Groton, CT 06340<br />

United States<br />

christopher.j.somps@pfizer.com<br />

(860) 715-2841<br />

Richard Davis Storer<br />

Merck Research Laboratories<br />

Sumneytown Pike<br />

P.O. Box 4<br />

WP45 311<br />

West Po<strong>in</strong>t, PA 19486-0004<br />

United States<br />

richard_storer@merck.com<br />

(215) 652-5872<br />

Tadaki Sugawara<br />

Daiichi Sankyo Company Limited<br />

399 Thornall Street<br />

Edison, NJ 08837<br />

United States<br />

tsugawara@dsus.com<br />

(732) 590-4317<br />

James A. Swenberg<br />

University of North Carol<strong>in</strong>a Chapel Hill<br />

CB 7431 Room 253C<br />

Rosenau Hall<br />

Chapel Hill, NC 27599-0001<br />

United States<br />

james_swenberg@unc.edu<br />

(919) 966-6139<br />

66<br />

t<br />

Thio Tanja<br />

RCC Ltd.<br />

Zelgliweg 1<br />

It<strong>in</strong>gen 4452<br />

Switzerland<br />

thio.tanja@rcc.ch<br />

(416) 197-5153 9<br />

V<br />

Jan Willem van der Laan<br />

National Institute of Public Health and Environment<br />

(RIVM)<br />

Centre for Biological Medic<strong>in</strong>es and Medical<br />

Technology<br />

P.O. Box 1<br />

Bilthoven 3720 BA<br />

Netherlands<br />

jan-Willem.van.der.Laan@rivm.nl<br />

(313) 027-4290 4<br />

Suryanarayana V. Vulimiri<br />

U.S. EPA<br />

1200 Pennsylvania Avenue NW<br />

Wash<strong>in</strong>gton, DC 20460<br />

United States<br />

vulimiri.sury@epa.gov<br />

(703) 308-7949<br />

W<br />

Zem<strong>in</strong> Wang<br />

Indiana University School of Medic<strong>in</strong>e<br />

635 Barnhill Drive MS A401<br />

Indianapolis, IN 46202<br />

United States<br />

zemwang@iupui.edu<br />

(317) 278-3094<br />

Jayne Wright<br />

Syngenta<br />

Jealott’s Hill International Research Centre<br />

Bracknell RG42 6EY<br />

United K<strong>in</strong>gdom<br />

jayne.wright@syngenta.com<br />

(013) 444-1447 4

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

Saved successfully!

Ooh no, something went wrong!