35th Annual Meeting Program - American College of Toxicology
Transcription
35th Annual Meeting Program - American College of Toxicology
American College of Toxicology 35 Annual Meeting Program th Orlando, Florida November 9–12, 2014 Hyatt Regency Grand Cypress www.actox.org Some Orlando photos are courtesy of Visit Orlando unless otherwise noted. Some photos by Harvey Smith Photography. AMERICANCOLLEGE COLLEGEOF OFTOXICOLOGY TOXICOLOGY AMERICAN Welcome! COLLEGE OF TOXICOLOGY AMERICAN Dear Colleagues and Guests, 2013–2014 COUNCIL 2013–2014 COUNCIL PRESIDENT PRESIDENT 2013–2014 COUNCIL Drew A. Badger, PhD, DABT Drew A. Badger, PhD, DABT PRESIDENT PRESIDENT-ELECT DrewMary A. Badger, DABT PRESIDENT-ELECT Ellen PhD, Cosenza, PhD, Mary Ellen Cosenza, PhD, DABT, RAC DABT, RAC PRESIDENT-ELECT Mary Ellen Cosenza, PhD, VICE PRESIDENT DABT, PRESIDENT Hanan N.VICE Ghantous, PhD,RAC DABT Hanan N. Ghantous, PhD, DABT VICE PRESIDENT SECRETARY Hanan N. Ghantous, PhD, DABT SECRETARY Grace M. Furman, PhD, DABT Grace M. Furman, PhD, DABT SECRETARY TREASURER Grace M. Furman, PhD, DABT Alan M.TREASURER Hoberman, PhD, Alan M. Hoberman, PhD, DABT, ATS DABT, ATS TREASURER Alan M. Hoberman, PhD, COUNCILORS DABT, COUNCILORS Nancy R. Bordelon, PhD, ATS DABT Nancy R. Bordelon, PhD, DABT Alan P. Brown, PhD, DABT COUNCILORS Alan P. Brown, PhD, DABT Nancy R. R. Bordelon, PhD, DABT David Compton, PhD, DABT David R. Compton, PhD, DABT Alan P.Jerry Brown, PhD, DABT F. Hardisty, DVM Jerry F. Hardisty, DVM David R.Anthony Compton, DABT L. PhD, Kiorpes, PhD, Anthony L. Kiorpes, DVM,PhD, DABT Jerry F. Hardisty, DVM DVM, DABT Timothy J. McGovern, PhD Anthony Kiorpes, PhD, Timothy J. L. McGovern, PhD DABT Sandra L.DVM, Morseth, PhD Sandra L. Morseth, PhD Timothy McGovern, Melissa C.J. Rhodes, PhD, PhD DABT Melissa C. Rhodes, PhD, DABT Sandra L. Morseth, PhD Patricia C. Ryan, PhD Patricia C. Ryan, PhD Melissa C. Rhodes, PhD, DABT PAST PRESIDENT On behalf of the American College of Toxicology, I would like to welcome you to ACT’s 35th Annual Meeting at the Hyatt Regency Grand Cypress in sunny Orlando, Florida. I am proud of this year’s meeting that includes a variety of applied and regulatory toxicology sessions, as well as a venue that offers numerous opportunities for fun and professional networking. I encourage you to review the schedule of scientific sessions and special events in this Program so that you can make the most of the days ahead. Sunday offers a variety of half-day Continuing Education (CE) courses as well as an extended Study Director Short Course. If you have not already registered in advance and would like to register for a course on-site, stop by the ACT Registration Desk outside of the Exhibit Hall and we’ll be happy to assist you. A detailed description for each course can be found in this Program. We are pleased to present Monday morning’s Plenary lecturer, Deborah Blum, Pulitzer-Prize Winner and Author of The Poisoner’s Handbook: Murder and the Birth of Forensic Medicine in Jazz Age New York. Wednesday morning’s Plenary lecturer will be Dr. Alison Van Eenennaam of University of California, Davis speaking on “Food and Feed Safety of Genetically Modified Organisms: The Hype and the Facts.” ACT’s Annual Meetings are known for being collegial, with many opportunities for professional networking and catching up with friends and colleagues. We officially kick off the meeting on Saturday with a low stress and fun golf outing followed by a reception for all the golfers. “Jazzicology,” our own home-grown band comprised of ACT members, is back this year to entertain us during Sunday’s Welcome Reception, which is a ticketed event that is not included in the price of the meeting registration. Don’t miss Monday’s Awards Luncheon and evening Poster Reception, both of which are included in your registration fee. New this year is the ToxTrot on Tuesday morning on a trail around the grounds of the Hyatt. Whether you walk or run, it’s sure to be fun! Participation in the ToxTrot is free, but advance registration is required. We’ve also added a breakfast reception for all meeting attendees in the Exhibit Hall following the ToxTrot event and a wine tasting with incoming President Mary Ellen Cosenza on Wednesday evening for ACT members only. PAST PRESIDENT During your daily breaks, I encourage you to visit the exhibitors at their booths in the Exhibit Hall as they are a vital part of the meeting. You also will have the opportunity to peruse over 100 posters on display in the Exhibit Hall. EDITOR-IN-CHIEF I would like to extend a special thank you to Program Committee Chair Mary Ellen Cosenza and Education Committee Chair Jerry Hardisty, and their committees, for planning an outstanding program. Patricia C. Ryan, PAST PRESIDENT Robin C. Guy, MS, PhD DABT, Robin C. Guy, MS, DABT, RQAP-GLP RQAP-GLP Robin C. Guy, MS, DABT, EDITOR-IN-CHIEF EDITOR-IN-CHIEF Mary BethRQAP-GLP Genter, PhD, Mary Beth Genter, PhD, DABT, ATS DABT, ATS Mary Beth Genter, PhD, EXECUTIVE DIRECTOR DABT, ATS EXECUTIVE DIRECTOR Nancy Rollman Nancy Rollman Let’s have a great week! EXECUTIVE DIRECTOR Nancy Rollman Drew A. Badger, PhD, DABT 2013–2014 ACT President 1821 Michael Faraday Drive, Suite 300, Reston, Virginia 20190 1821 Michael Faraday Drive, Suite 300,[email protected] Reston, Virginia 20190 Telephone: 703.547.0875 Fax: 703.438.3113 Email: Website: www.actox.org Telephone: 703.547.0875 Fax: 703.438.3113 Email: [email protected] Website: www.actox.org 1821 Michael Faraday Drive, Suite 300, Reston, Virginia 20190 Telephone: 703.547.0875 Fax: 703.438.3113 Email: [email protected] Website: www.actox.org th 35 American College of Toxicology Annual Meeting 2014 Schedule of Events Overview Saturday, November 8 12:00 PM–5:30 PM ACT Golf Outing (Ticketed Event) Grand Cypress Golf Course 3:00 PM–5:00 PM Registration Open Registration Area #1 4:00 PM–6:00 PM Speaker Ready Room Open Poinciana B 5:00 PM–7:00 PM ACT Council Meeting Magnolia Sunday, November 9 CE Continental Breakfast Regency Hall 7:00 AM–9:00 AM Speaker Ready Room Open Poinciana B 7:00 AM–5:00 PM Registration Open Registration Area #1 8:00 AM–11:30 AM CE 1: Best Practices in Toxicologic Pathology Regency Hall CE 2: Regulatory Toxicology—In the FDA and Beyond Palm CE 3: Drug Line Extensions: Nonclinical Testing and Solutions Regency Hall CE 4: Interpreting Adverse Clinical and Anatomic Pathology Results: Putting It All Together Regency Hall 8:00 AM–3:15 PM Study Director Short Course Regency Hall 9:15 AM–10:30 AM CE Refreshment Break (See Course Details for Break Times) Regency Hall 11:30 AM–1:00 PM Lunch On Your Own 12:00 Noon–12:55 PM Exhibitor-Hosted Program: Submitting Standardized SEND to the FDA: Data Fitment and Review Considerations Presented by: PointCross Life Sciences, Inc. Regency Hall 12:00 Noon–4:30 PM Exhibits and Poster Setup Grand Cypress Ballroom 12:00 Noon–1:00 PM New Member Lunch (By Invitation Only) Hydrangea In the Event of Rain: Portico West 12:00 Noon–2:00 PM Speaker Ready Room Open Poinciana B 1:00 PM–4:30 PM CE 5: Toxicology and Pathology of the Respiratory System Regency Hall CE 6: (Q)SAR—A Tool for the Toxicologist Regency Hall CE 7: Managing Anti-Drug Antibody Responses during Biologics Drug Development Palm CE 8: Metabolites: Guidance and Considerations in Drug Development Regency Hall 2:30 PM–3:10 PM CE Refreshment Break (See Course Details for Break Times) Regency Hall 4:00 PM–4:45 PM Student Poster Setup Grand Cypress Ballroom 5:00 PM–6:30 PM Student Poster Judging Grand Cypress Ballroom 6:30 PM–8:30 PM Welcome Reception Featuring Jazzicology (Ticketed Event) The Wilderness In the Event of Rain: Portico West 1 OVERVIEW 7:00 AM–8:00 AM th 35 American College of Toxicology Annual Meeting 2014 Schedule of Events Overview (continued) OVERVIEW Monday, November 10 6:45 AM–8:00 AM Past Presidents' Breakfast (By Invitation Only) Gardenia 7:00 AM–8:00 AM Continental Breakfast Grand Cypress Ballroom Prefunction Area 7:00 AM–9:00 AM Speaker Ready Room Open Poinciana B 7:00 AM–5:00 PM Registration Open Registration Area #1 8:00 AM–8:55 AM Plenary Lecture: The Poisoner's Guide to Life Speaker: Deborah Blum Regency Hall 9:00 AM–12:00 Noon Symposium 1: From Mice to Men, Development of Human Drugs and Biologics under the Animal Rule Palm Symposium 2: What the AEL is the NOAEL? Grand Cypress Ballroom A Symposium 3: Systems Toxicology: The Future of Risk Assessment Magnolia 9:30 AM–12:00 Noon Exhibits and Posters Open Grand Cypress Ballroom 10:20 AM–10:55 AM Refreshment Break (Exhibit Hall: See Session Details for Break Times) Grand Cypress Ballroom 10:20 AM–10:55 AM Book Signing by Plenary Lecturer Deborah Blum ACT Member Lounge in the Grand Cypress Ballroom 11:00 AM–2:00 PM Speaker Ready Room Open Poinciana B 12:00 Noon–2:00 PM Awards Ceremony and Luncheon (Open to all Registrants) Speaker: Anthony Dayan, Distinguished Scientist Awardee Regency Hall 2:00 PM–5:00 PM Symposium 4: Antidrug Antibody-Independent Immune Responses to Biologics in Rodent Studies—Regulatory Success in Early Drug Development Palm Symposium 5: Use of Humanized Mouse Models in DMPK and Safety Testing of Compounds Magnolia Symposium 6: Targeted Cancer Therapeutics: Concepts and Strategies to Improve Oncology Drug Development Grand Cypress Ballroom A 2:00 PM–7:00 PM Exhibits and Posters Open Grand Cypress Ballroom 3:20 PM–3:55 PM Refreshment Break (Exhibit Hall: See Session Details for Break Times) Grand Cypress Ballroom 3:20 PM–3:55 PM Meet Distinguished Scientist Awardee Anthony Dayan ACT Member Lounge in the Grand Cypress Ballroom 5:30 PM–7:00 PM Poster Session and Reception (Exhibit Hall) Grand Cypress Ballroom 2 th 35 American College of Toxicology Annual Meeting 2014 Schedule of Events Overview (continued) Tuesday, November 11 ACT ToxTrot (Registration Required by October 30) Portico West Patio 8:00 AM–9:00 AM Breakfast Reception (Exhibit Hall) Grand Cypress Ballroom 8:00 AM–4:30 PM Exhibits and Posters Open Grand Cypress Ballroom 8:00 AM–5:00 PM Registration Open Registration Area #1 8:00 AM–10:00 AM Speaker Ready Room Open Poinciana B 9:00 AM–12:00 Noon Symposium 7: Breathe In, Breathe Out, It's Easy: What You Need to Know about Developing Inhaled Drugs Regency Hall Symposium 8: Cardiovascular Safety Evaluation—In Vitro to Humans Grand Cypress Ballroom A Symposium 9: Centrally Administered Compounds in Small and Large Species—Dose Routes, Study Considerations, and Data Interpretation Palm 10:10 AM–10:55 AM Refreshment Break (Exhibit Hall: See Session Details for Break Times) Grand Cypress Ballroom 12:00 Noon–1:30 PM IJT Editorial Board Meeting Magnolia 12:00 Noon–2:00 PM Lunch On Your Own 12:00 Noon–1:30 PM 2015 Program Planning Meeting (All ACT Members Invited, Box Lunch Provided. Sign Up at Registration Desk.) Regency Hall 2:00 PM–5:00 PM Symposium 10: Cytokines: The Good, the Bad, and the Deadly Grand Cypress Ballroom A Symposium 11: Differentiating Adverse from Adaptive Changes in Toxicology Palm Symposium 12: In Silico Methods for Mutagenicity Prediction vis-à-vis ICH M7 Guidance Regency Hall 3:20 PM–3:55 PM Meet IJT Editor Mary Beth Genter Booth 304 in the Grand Cypress Ballroom 3:20 PM–3:55 PM Refreshment Break (Exhibit Hall: See Session Details for Break Times) Grand Cypress Ballroom 4:30 PM–7:30 PM Exhibit and Poster Teardown (Exhibit Hall) Grand Cypress Ballroom 5:00 PM–6:30 PM ACT Members’ Meeting (All ACT Members Invited) Regency Hall 3 OVERVIEW 6:30 AM–7:30 AM th 35 American College of Toxicology Annual Meeting 2014 Schedule of Events Overview (continued) Wednesday, November 12 OVERVIEW 7:00 AM–7:55 AM Exhibitor-Hosted Program: Maintaining Quality and Regulatory Compliance in a Global Setting Presented by: WuXi AppTec Grand Cypress Ballroom H Exhibitor-Hosted Program: Practicalities of Dermal Administration Presented by: Huntingdon Life Sciences Grand Cypress Ballroom G Exhibitor-Hosted Program: Zebrafish: Reshaping Toxicity Testing Presented by: Charles River Grand Cypress Ballroom I 7:00 AM–8:00 AM Continental Breakfast Grand Cypress Ballroom Prefunction Area 7:00 AM–8:00 AM Council Orientation Magnolia 7:30 AM–1:30 PM Registration Open Registration Area #1 8:00 AM–12:00 Noon Speaker Ready Room Open Poinciana B 8:00 AM–8:55 AM Plenary Lecture: Food and Feed Safety of Genetically Modified Organisms: The Hype and the Facts: Speaker: Alison Van Eenennaam, PhD Grand Cypress Ballroom D 9:00 AM–12:00 Noon Symposium 13: Novel Therapeutics in Oncology: Recent Advances and Considerations for Nonclinical Safety Evaluation Palm Symposium 14: Applied Nanotoxicology Grand Cypress Ballroom A Symposium 15: Stem Cells Research Grand Cypress Ballroom B Symposium 16: Safety Assessment Updates for Preventive Vaccines and Vaccine Adjuvants Grand Cypress Ballroom C 10:20 AM–11:00 AM Refreshment Break (See Course Details for Break Times) Grand Cypress Ballroom Prefunction Area 12:00 Noon–12:55 PM Exhibitor-Hosted Program: Demonstration of New Expert Alert System for In Silico Assessment of Impurities under ICH M7 Guidelines Presented by: Leadscope Inc. Grand Cypress Ballroom H Exhibitor-Hosted Program: Evaluation of Electronic Cigarettes in Testing Tobacco-Related Products Presented by: Battelle Grand Cypress Ballroom G Exhibitor-Hosted Program: Seizure Liability, qEEG and Sleep Quantification in Nonclinical Drug Development: Regulatory and Scientific Considerations Presented by: CiToxLAB Grand Cypress Ballroom I 12:00 Noon–1:30 PM ACT Council Meeting Magnolia 12:00 Noon–1:30 PM Lunch On Your Own 1:30 PM–4:30 PM Symposium 17: Hot Topics Grand Cypress Ballroom D 3:00 PM–3:20 PM Refreshment Break Grand Cypress Ballroom Prefunction Area 5:00 PM–6:30 PM Wine Tasting: Meet the 2015 ACT President, Mary Ellen Cosenza (Open to All ACT Members, Pre-Registration Required) Grand View Terrace 4 American College of Toxicology 35th Annual Meeting Program Content President’s Welcome �������������������������������������������������������������������������������������������� Inside Front Cover Schedule of Events Overview ������������������������������������������������������������������������������������������������������������� 1 General Information General Information Venue ������������������������������������������������������������������������������������������������������������������������������������������������������ 6 Internet Access ������������������������������������������������������������������������������������������������������������������������������������ 6 Room Reservations ���������������������������������������������������������������������������������������������������������������������������� 6 Parking ��������������������������������������������������������������������������������������������������������������������������������������������������� 6 Transportation ������������������������������������������������������������������������������������������������������������������������������������� 6 Climate ��������������������������������������������������������������������������������������������������������������������������������������������������� 6 Attire ������������������������������������������������������������������������������������������������������������������������������������������������������� 6 Registration Information ����������������������������������������������������������������������������������������������������������������� 6 Restaurants ������������������������������������������������������������������������������������������������������������������������������������������� 6 Photography Policy ��������������������������������������������������������������������������������������������������������������������������� 6 Hotel Map ���������������������������������������������������������������������������������������������������������������������������������������������� 8 Resort Map ������������������������������������������������������������������������������������������������������������������������������������������������� 9 Special Events������������������������������������������������������������������������������������������������������������������������������������������ 10 2014 Awards ��������������������������������������������������������������������������������������������������������������������������������������������� 12 Program Agenda Continuing Education Courses Study Director Short Course��������������������������������������������������������������������������������������������������������� 17 Morning CE Courses ������������������������������������������������������������������������������������������������������������������������ 18 Afternoon CE Courses ������������������������������������������������������������������������������������������������������������������� 22 Scientific Sessions Monday Morning Sessions ����������������������������������������������������������������������������������������������������������� 27 Monday Afternoon Sessions ������������������������������������������������������������������������������������������������������� 34 Tuesday Morning Sessions ���������������������������������������������������������������������������������������������������������� 40 Tuesday Afternoon Sessions ������������������������������������������������������������������������������������������������������� 46 Wednesday Morning Sessions ���������������������������������������������������������������������������������������������������� 53 Wednesday Afternoon Session ��������������������������������������������������������������������������������������������������� 62 Abstracts Orlando November 9–12, 2014 2014 Poster Sessions ACT Poster Presentations ������������������������������������������������������������������������������������������������������������������� 63 100 Series—General Toxicology ����������������������������������������������������������������������������������������������� 64 200 Series—Regulatory ����������������������������������������������������������������������������������������������������������������� 70 300 Series—Safety Evaluation Nonpharmaceuticals���������������������������������������������������������� 73 400 Series—Toxicology Methods ��������������������������������������������������������������������������������������������� 80 500 Series—Safety Evaluation Pharmaceuticals ������������������������������������������������������������������ 93 Author Index ����������������������������������������������������������������������������������������������������������������������������������������� 102 Exhibit Information Exhibitor Directory ����������������������������������������������������������������������������������������������������������������������������� 108 Exhibit Hall Map ���������������������������������������������������������������������������������������������������������������������������������� 136 Exhibitor-Hosted Programs ������������������������������������������������������������������������������������������������������������ 137 Reference Council Listing ������������������������������������������������������������������������������������������������������������������������������������� Committee Listings ���������������������������������������������������������������������������������������������������������������������������� Headquarters Staff ����������������������������������������������������������������������������������������������������������������������������� Past Presidents ������������������������������������������������������������������������������������������������������������������������������������� ACT Award Recipients ����������������������������������������������������������������������������������������������������������������������� Corporate Members �������������������������������������������������������������������������������������������������������������������������� Welcome New Members ������������������������������������������������������������������������������������������������������������������ ACT Charter Members ����������������������������������������������������������������������������������������������������������������������� ACT Distinguished Fellows ������������������������������������������������������������������������������������������������������������� 5 138 140 145 146 147 148 149 149 149 GENERAL INFO th 35 American College of Toxicology Annual Meeting 2014 General Information Venue Transportation Hyatt Regency Grand Cypress Resort Theme Park Transportation The Hyatt Regency Grand Cypress provides complimentary transportation to and from Walt Disney World,® Universal Orlando® and Sea World. Please contact the Concierge directly for scheduled times. One Grand Cypress Boulevard, Orlando, FL, 32836 Telephone: 407.239.1234 | Fax: 407.239.3800 ACT 35th Annual Meeting Registrants will enjoy the Hyatt Regency Grand Cypress where they will be secluded in the heart of it all. The Hyatt Regency Grand Cypress is set in Orlando theme park country near Lake Buena Vista, while being surrounded by 21 acres of shimmering lakes and lush landscaping. Resort Transportation A complimentary shuttle runs continuously around the resort from 6:30 am–11:00 pm daily. Hertz Options during downtime are numerous. The Hyatt Regency Grand Cypress is located just one mile from Walt Disney World® Resort and five miles from Sea World®, and offers a complimentary shuttle to both. The Hyatt Regency Grand Cypress also offers an impressive collection of on-site activities. From world-class championship golf and tennis to luxury spa services and leisurely afternoons sailing on the private lake; you may have to stay longer just to experience it all! The Hyatt Regency Grand Cypress offers an ideal destination for the stimulating scientific exchange at this year’s ACT Annual Meeting. Internet Access There will not be Wi-Fi Internet access available in the meeting rooms or Exhibit Hall. ACT will have an Internet Café located near the registration area, which will be available during registration hours. The café will include several computers with Internet access. If you are staying at the hotel, the room rate includes basic Internet access (wireless and wired) in your guest room. Room Reservations Meeting Rate • $189 single/double occupancy • Check-in time is 4:00 pm and check-out time is 11:00 am. Waived Resort Fee For all attendees staying at the hotel the $10 Hotel Program Fee has been waived. The Hotel Program Fee provides use of hotel activities and amenities including: hard-wire and in-room basic wireless Internet (WiFi will not be accessible in the session rooms or Exhibit Hall), in-room laptop safe, intra-property transportation, unlimited use of the Health Club, 9-hole Pitch and Put course (balls and clubs included), golf driving range access, court time at the Racquet Club (racquet and balls included), basketball, volleyball, rock climbing wall, non-motorized boats, as well as resort bicycles. Complimentary theme park shuttle to Disney, SeaWorld, Universal Studios/Island of Adventure (please check with the hotel concierge regarding shuttle times). Parking • Self-Parking, Fee: $11 with in/out privileges • Valet Parking, Fee: $23 with in/out privileges • Event Only (day) parking, Fee: $6 Located on property at the Hyatt Regency Grand Cypress, Hertz is offering special meeting rates to ACT meeting attendees. Reservations can be made online or by calling Hertz directly at 1.800.654.2240 or 405.749.4434. Please provide CV#022L4090. To talk to the Hertz desk at the Hyatt Regency Grand Cypress, please call 407.238.5309. Climate Orlando’s average temperature in November is a high of 79 degrees and a low of 59 degrees. November is one of the driest months of the year. Attire Meeting attire is business casual. The meeting rooms may be cool, so be sure to dress with a lightweight layer. Additionally, comfortable shoes for walking are advisable. Registration Information The registration fee includes the American College of Toxicology Awards Luncheon on Monday, continental breakfast Monday through Wednesday, all refreshment breaks, and a Monday evening poster session and reception. Additional registration fees are required for Continuing Education courses, for the Sunday evening Welcome Reception, and for the Golf Outing. If you have registered prior to October 15, you will receive your name badge, course ticket(s), and qualifying ribbons in the mail. You will not need to stand in the registration line if you received these in advance. You may pick up the Program and your badge holder on-site. Restaurants The Hyatt Regency Grand Cypress offers a variety of dining options in addition to in-room dining. The hotel’s restaurants offer spectacular settings and present inspiring backdrops for the delectable culinary creations from their talented chefs. Should you require assistance with reservations at any of their dining outlets before or during your stay, we encourage you to use Hyatt’s dedicated E-Concierge service online. Photography Policy ACT meeting attendees are asked to strictly adhere to the photography policy while participating in sessions and while in the Exhibit Hall. As a courtesy to our speakers, poster presenters, and exhibitors, ACT asks that participants refrain from electronic capture of any and all presented material unless given written permission from the proprietary. Any person who does not comply with the policy may be asked to leave the event. 6 The network for ACT members only! Access interACT from the ACT homepage to: •Build your MyPage profile •Upload your CV •Find an ACT member •Access your communities •Read the ACTalks blog and stay in touch! Access the interACT community today! Visit www.actox.org 7 T +1 407 239 1234 F +1 407 239 3800 grandcypress.hyatt.com th 35 American College of Toxicology Annual Meeting 2014 Hotel Map FLOOR PLAN Ground Level HYDRANGEA GENERAL INFO One Grand Cypress Boulevard Orlando, FL 32836 USA FEDEX OFFICE HYDRANGEA PATIO LOBBY / SALON ELEVATOR ORCHID PARKING ToxTrot Meeting Area GARDENIA SERVICE / LOADING AREA MEN G PATIO LOBBY LEVEL BOARD ROOMS HIBISCUS I H OFFICE 1 Exhibit Hall CAMELLIA GRAND CYPRESS BALLROOM MEN WOMEN TO MAIN LOBBY PORTICO WEST PREFUNCTION AREA PORTICO EAST WOMEN D E F LOBBY LEVEL MEETING ROOMS MEN A REG 2 SOUTH BUS ARRIVAL / DEPARTURE B DRESSING ROOM REG 1 C Registration B REG 4 REG 3 4 3 2 JACARANDA WOMEN REGENCY HALL 6 7 E 8 B A B POINCIANA C MAGNOLIA A 5 14th Floor D OUTDOOR PATIO ATRIUM ROOM 1 MEN Awards Luncheon A PALM UP TO LOBBY F SERVICE LOADING C C Speaker Ready Room RESTROOM FOYER D 9 SERVICE CORRIDOR LA COQUINA* Note: * La Coquina Entrance on lobby level 07.13 8 9 WETLANDS OPENSPACE SOUTH 9 RACQUET CLUB WESTERN STABLE RIDING TRAILS WETLANDS OPENSPACE SR 535 NORTH JACARANDA GOLF ACADEMY RECEPTION CENTER EAST 9 VILLAS OF GRAND CYPRESS CLUB HOUSE DRIVING RANGE AREA HIGHLIGHTED ON THIS MAP EXECUTIVE MEETING CENTER NORTH 9 VILLAS EQUESTRIAN CENTER SR 535 NEW COURSE 18 HYATT REGENCY GRAND CYPRESS WETLANDS OPENSPACE I-4WEST WEST I-4 SR 535 I-4 Western Trail Ride Camellia Gardenia Orchid Hibiscus Grand View Terrace 29 29 21 22 23 24 25 26 27 28 29 Racquet Ball Basketball Court Lakside Marina Ground Floor Level Entry Cascade La Coquina Palm Cafe and General Store On The Rocks Health Club Game Room Pitch ‘n Putt Racquet Club Ground Floor Level 16 17 18 19 20 Atrium/Lobby Level Meeting Space One Grand Cypress Blvd. • Orlando, FL 32836 USA 407-239-1234 • www.hyattgrandcypress.com Parking Tennis Pro Shop Tennis Courts Trellises Lounge La Coquina Entry Cascade Entry White Horse Sports Bar & Grill Hemingway’s Hurricane Lounge Lamont’s Shop Accents Shop Concierge/Guest Services Guest Elevators Front Desk/Registration Hemingways/Recreation Area Exit Camp Hyatt Child Care Center Business Center Grand Cypress Salon Entry TODISNEY DISNEY TO THEMEPARKS PARKS THEME WETLANDS OPENSPACE GRAND CYPRESS RESORT PROPERTY 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 Atrium/Lobby Level 30 31 32 33 34 35 36 37 38 39 40 Pitch ‘n Putt Course 28 28 Palm Poinciana Magnolia Regency Hall Grand Cypress Ballroom Pre-Function Area Portico Area Jacaranda Cypress Point Pavilion Wilderness Area Theme Park Bus Depot Jogging Trails Stables/Barn N Spa Riding Trails Grand Cypress Blvd. Ballroom and Hotel Self Parking Pitch ‘n Putt Check in Lower Level Meeting Space 21 21 Pool 27 27 Spa Meeting Space Spa 55 25 26 25 66 26 Pool Firepit Towel Hut Boat and Bike Rental Heated Pool Meeting Rooms Lake Windsong Beach 21 21 34 34 A B C A B C 30 30 D E F 37 37 A C 33 33 A 32 B 32 C 31 31 B D 11 15 15 19 18 19 18 Rooms Atrium 88 16 16 Meeting 14 14 17 17 77 22 22 Entrance to Bus Parking Theme Park Bus Depot 17 40 13 13 12 12 24 24 33 99 22 10 10 20 23 20 23 Hyatt Regency Grand Cypress Lower Level Meeting Space 36 36 35 35 D G 21 21 F E I H Service Only Hotel Arrival and Valet Parking 44 Exit to Grand Cypress Blvd. Hotel Main Entrance 11 11 38 38 Wilderness Area Heliopad Grand Cypress Blvd. Main Gate Welcome Reception To I-4 39 39 Jogging Trails SR 535 To Villas of Gra nd Cypress, Gra Golf Club and Equestrian Ce nd Cypress nter, (1.5 mil es) 2014 Resort Map GENERAL INFO Hyatt Regency Grand Cypress th 35 American College of Toxicology Annual Meeting GENERAL INFO th 35 American College of Toxicology Annual Meeting 2014 Special Events Golf Outing Poster Session and Reception Saturday, November 8 12:00 Noon–5:30 PM Grand Cypress Golf Course $125 Sponsored by HistoTox Labs and WIL Research (Advance registration required and space is limited) Monday, November 10 5:30 PM–7:00 PM Grand Cypress Ballroom Sponsored by SAGE (Open event, all registrants invited) This is the opportunity to discuss the latest research findings and methodology with poster presenters and visit with exhibitors during a reception with wine and light fare. Reception is included in the Annual Meeting registration fee. Arrive in Orlando early and enjoy a day on the golf course with other ACT Annual Meeting attendees. This is a great way to build relationships in a relaxed atmosphere while enjoying the beautiful Florida weather on a classic golf course. This event is played in a fun format that accommodates all levels of golfers. ACT’s Inaugural ToxTrot The event will take place on the Jack Nicklaus Signature New Course at the Grand Cypress Golf Club. Set in the midst of an open meadow, the New Course is Jack Nicklaus’ tribute and homage to the famed Old Course at St. Andrews, Scotland. The $125 fee per player includes 18 holes with cart, practice balls, club storage and cleaning, a light reception, and participant door prizes. Club Rentals are available for $50 per set and shoe rentals are available for $20 per pair. Welcome Reception Sunday, November 9 6:30 PM–8:30 PM The Wilderness In the event of rain: Portico West $50 (Ticketed event) Tuesday, November 11 6:30 AM–7:30 AM Portico West Patio (Free event, registration by October 30 required) ACT is holding its first ToxTrot at this year’s 35th Annual Meeting! Join members from ACT’s Council as they “trot” through the property of the Hyatt as the sun rises. This event is a 3.1-mile loop where you can walk, trot, or run. There is no fee for this event and you will receive a free t-shirt while supplies last to wear on the day of the ToxTrot. There will be Hyatt staff at the finish line to give you your trot time, if you are interested. You will receive your bib Tuesday morning, so please arrive early to the event. This is a great opportunity to meet fellow ACT members and network, all while burning a few calories. We will also be raffling off a complimentary 2015 ACT Annual Meeting registration after the event for participants who complete the Trot! ACT Members’ Meeting After a stimulating day of Continuing Education, mingle with your colleagues at a special dinner in the Wilderness area under the stars. You will relax to the smooth sounds of “Jazzicology,” a combo of ACT members appearing again by popular demand. Be sure to purchase your $50 ticket in advance as space is limited. Your ticket includes a fabulous barbeque dinner and two drink tickets. Tuesday, November 11 5:00 PM–6:30 PM Regency Hall (All ACT members invited) Awards Ceremony and Luncheon Wine Tasting with 2015 ACT President Mary Ellen Cosenza Monday, November 10 12:00 Noon–2:00 PM Regency Hall (Open event, all registrants invited) The Distinguished Scientist Award recipient Dr. Anthony Dayan will provide the keynote presentation at this lunch, which is included in the Annual Meeting registration. Other award recipients will be recognized and the Furst Award for the best student poster at the meeting will be announced. See page 12 for award recipients. Come to the meeting to hear the latest ACT business and provide feedback for the future. Wednesday, November 12 5:00 PM–6:30 PM Grand View Terrace In the event of rain: Grand Cypress Ballroom G (Free event for ACT members only, advance registration required) Come meet the incoming 2015 ACT President Mary Ellen Cosenza and share your thoughts on the ACT Annual Meeting over a glass of wine! This wine tasting event is a new opportunity to network with fellow ACT members while getting to know the incoming ACT President. Wine and light fare will be provided. This is a free event for all ACT members and advance registration is required. 10 Don’t Miss the Welcome Reception Sunday, November 9 The Wilderness In the Event of Rain: Portico West 6:30 PM–8:30 PM $50 Ticket After a stimulating day of Continuing Education courses, mingle with your colleagues at a delicious barbecue dinner under the stars. You will relax to the smooth sounds of Jazzicology, a combo of ACT members appearing again by popular demand. www.actox.org 11 GENERAL INFO th 35 American College of Toxicology Annual Meeting 2014 2014 Awards 2014 Awards will be presented at the Awards Ceremony and Luncheon on Monday, November 10 at 12:00 noon–2:00 pm in the Regency Hall. This event is open to all attendees (members, nonmembers, and exhibitors, etc.). For a historical listing of award recipients, see page 147 or visit www.actox.org/aboutact/pastrecipients.asp. committee), and longstanding support of the College's activities. The recipient must be a member of ACT. ACT Distinguished Scientist Award ACT recognizes Norman N. Kim, MS, DABT as the 2014 Service Awardee. The ACT Distinguished Scientist Award recognizes an individual (not necessarily a member of ACT) who has made outstanding contributions to toxicology, its relationship to the regulation of chemicals, and/or the improvement of public health. The DSA winner becomes the Keynote Awards Luncheon Speaker at the Annual Meeting. ACT recognizes Anthony Dayan, Emeritus Professor, MBBS (Hons) MD as the 2014 Distinguished Scientist Awardee. After gaining a degree in Pharmacology and Physiology, Dr. Dayan qualified as a physician in 1959 and specialized in pathology and neuropathology after various internships. He was a member of staff of the Hospital for Sick Children and National Hospitals for Nervous Diseases, London, from 1965–1973 when he joined the Wellcome Foundation Research Laboratories as a toxicologist, later becoming Head of Toxicology. In 1984, Dr. Dayan moved to become the first Professor of Toxicology and Head of the DH Department at Barts Hospital until his first retirement in 1998. Whilst at Barts and subsequently he was a member of many UK, EC, WHO and other international scientific advisory committees concerned with all aspects of the safety of human and veterinary drugs, medical devices, foods, air, and pesticides as well as advanced therapies, such as the products of biotechnology and gene therapy. He has also consulted widely in those fields for companies and research bodies. Dr. Dayan has been Chairman or President of the British Toxicology Society, HESI and ILSI Europe, and of the Comparative Medicine Section of the Royal Society of Medicine and a member of its Board of Directors. Dr. Dayan's academic career included directing and examining PhD and MD candidates in several universities in Britain and Europe, organizing an international collaborative study of immunotoxicity test methods and running an annual training course for WHO. ACT Service Award The ACT Service Award recognizes an individual for their long-term dedication to ACT including but not limited to service to the College (e.g., Councilor, officer, committee member), frequent participation and contribution to the Annual Meeting (speaker, chairperson, organizing Norman Kim has been an active member of the American College of Toxicology (ACT) since 2001. He has served in numerous capacities for ACT, which have included positions on the Nominating Committee (2005), Councilor (2007–2009), Membership Committee (Chair, 2008–2009), Treasurer (2011–2013), Finance Committee (Chair, 2011–2013), Publications Committee (2011–2013), Resource Committee (2013), and Program Committee (2014). As the Treasurer and Chair of the Finance Committee, the College’s finances were well managed. The College’s assets and investments were stabilized during the turbulent economic times, appropriately allocated to provide a more diverse investment portfolio, and greatly enhanced to better support the future needs of the College and to further benefit the ACT members. Many of the financial functions occurred during the challenging and critical stages of growth for the College when numerous organizational changes and new initiatives took place. Norman has supported ACT in many other roles that included organizing and chairing various Symposia and Continuing Education courses at the ACT Annual Meetings. He has also served on the Organizing Committees of the ACT-hosted toxicology training courses in the US (2010–2013) and in the UK (2013–2014) on behalf of ACT. Mr. Kim is currently a Director of Preclinical Safety at Biogen Idec, covering multi-functional roles since 2011. He has managed several toxicology programs, and currently is the head of the Discovery Toxicology group and the interim-lead of the Preclinical Safety department. He has over 25 years of experience in the field of toxicology. He was previously employed at Inotek Pharmaceuticals as the head of preclinical development working on ophthalmology programs, at Sepracor as a director of toxicology coordinating pulmonary and CNS programs, at Alkermes where he provided preclinical research and development support on drug delivery of proteins and small molecules, and at Arthur D. Little where he was a study director in genetic toxicology. He has worked on numerous INDs in the US, and CTAs and CTNs internationally. He has successfully been involved in several NDA and MAA processes, including approvals of drugs for multiple sclerosis (Tecfidera), asthma (Xopenex HFA MDI), chronic obstructive pulmonary disease (Brovana), insomnia (Lunesta), and growth hormone deficiency (Nutropin Depot). He has a number of patents issued. He received his MS degree in biomedical 12 science specializing in toxicology from the Northeastern University and BA degree in biology from the University of Rhode Island. He is a member of the Society of Toxicology, and participates on various external preclinical subcommittees including Efpia’s Preclinical Development Committee, PhRMA’s Clinical and Preclinical Development Committee, and HESI’s Neurotoxicity Biomarkers and Cardiotoxicity Committees. He is certified by the American Board of Toxicology (ABT), and currently serves on the Board of Directors at ABT (2014–2018). ACT Carol C. Lemire Unsung Hero Award 2014 ACT Young Professional Award The ACT Young Professional Award recognizes an ACT member with no more than 10 years of full-time employment experience since completing the highest degree (not including postdoctoral training). Nominee must have demonstrated outstanding service to the College, including serving as an officer, councilor, committee member, and/or frequent participation and contribution at the Annual Meeting or any other College activity (i.e., speaker, chairperson, organizing committee, etc.). ACT recognizes Ilona G. Bebenek, PhD as the 2014 Young Professional Awardee. The ACT Carol C. Lemire Unsung Hero Award recognizes an ACT member or staff who has made substantive unrecognized contributions to ACT. The nominee should have demonstrated on-going willingness to help in ACT activities generally behind the scenes. ACT recognizes Kok-Wah Hew, PhD, DABT as the 2014 Carol C. Lemire Unsung Hero Awardee. Dr. Hew is a Scientific Director at the Global Drug Safety Research and Evaluation Department of Takeda Pharmaceutical Company. He received his PhD in Anatomy and Cell Biology from the University of Michigan, Ann Arbor, Michigan. After graduation, he worked as a Study Director in Reproductive Toxicology at Ciba-Geigy Corporation and Novartis Pharmaceuticals Corporation. He also served as the Manager of Reproductive Toxicology at Springborn Laboratories; Senior Manager, and subsequently Associate Director in the Nonclinical Drug Safety Evaluation Department of Purdue Pharma; and Director of Toxicology at Emisphere Technologies, Inc. He has more than 20 years of experience in drug safety assessment, with special interests in reproductive toxicology and juvenile animal toxicity studies. In addition to authoring numerous toxicology reports and regulatory documents, he is also the lead or co-author of a number of peer-reviewed journal articles and book chapters. Dr. Hew was a member of the Editorial Board in the Reproductive Toxicology journal, and is currently a member of the Editorial Board of the International Journal of Toxicology as well as the Journal of Toxicological Sciences. Since 2010, he is the Course Director and a speaker of the annual weeklong course Toxicology for Industrial and Regulatory Scientists, hosted by the American College of Toxicology (ACT). Dr. Hew is a Diplomate of the American Board of Toxicology. He is also a member of ACT, Society of Toxicology (SOT), Teratology Society, Japanese Teratology Society, Middle Atlantic Reproduction and Teratology Association (MARTA), and the Developmental and Reproductive Toxicology Technical Committee in Health and Environmental Sciences Institute of the International Life Sciences Institute. In the SOT and Teratology Society, he has served on various committees, including the Chair of the Continuing Education Committee in both societies. He is also a past president of MARTA. Dr. Ilona Bebenek is a toxicologist at the U.S. Food and Drug Administration, in the Division of Transplant and Ophthalmology Products, Center for Drug Evaluation and Research. She was previously employed at ChemRisk/Cardno as a scientific consultant. She completed her PhD in Molecular Toxicology at University of California, Los Angeles. Her principal areas of training and expertise include mechanisms of inflammation and regulatory pathways in asthma, radiation biology, gene regulation, mechanisms of carcinogenesis and regulatory toxicology. Ilona won ACT’s Furst Best Student Poster Award in 2011 and was invited to participate in ACT’s strategic meeting in June 2011 and 2012, which included service on the ad hoc rebranding committee. She is currently chair of ACT’s webinar subcommittee and a member of the Education Committee. 13 GENERAL INFO th 35 American College of Toxicology Annual Meeting GENERAL INFO th 35 American College of Toxicology Annual Meeting ACT President’s Award for Best Paper Published in the International Journal of Toxicology ACT International Travel Grants The ACT President's Award recognizes the authors of the best paper published in International Journal of Toxicology issues 5 and 6 of the previous year through and including IJT issue 4 (July–August) of the current year. The Publications Committee carefully considered nominations for this award. TOXICOLOGY Volume 33 | Number 1 | January/February 2014 Authors: Jessica L. Price, T. Scott Manetz, Jeffry D. Shearer, Robert V. House Official Journal of the American College of Toxicology IJT33_1_Covers.indd 1 2014������� Solange Costa, Portugal Giorgia Del Favero, Italy Deepak Dhakal, Nepal Maria Elisa Peichoto, Argentina Yeshvandra Verma, India ACT North American Graduate Fellowships 2014������� Julie Castaneda, University of California, Los Angeles Theresa Lansdell, Michigan State Caroline Moore, University of California, Davis 2014.........International Journal of Toxicology: Volume: 32(5): Pages 327–335, September/October 2013 INTERNATIONAL JOURNAL OF 2014 ACT Student Travel Awards 2/3/14 1:01 PM A limited number of graduate Student Travel Awards for current students will be awarded based on merit of abstracts and completed applications. Paper Title: “Preclinical Safety Assessment of a Recombinant Plague Vaccine (rF1V)” 2013������� International Journal of Toxicology: Volume: 31(5): Pages 423–429, September/October 2012 Authors: Sudhir A. Shah, Madhav G. Paranjpe, Phillip I. Atkins, Eias A. Zahalka 2014������� Winners to be announced at the Awards Luncheon on Monday, November 10. 2013������� Shailender Singh Chauhan, Jamia Millia Islamia, India Chand Basha Davuljigari, Sri Venkateswara University, India Heidi Hsieh, University of Cincinnati, United States Chitrada Kaweeteerawat, University of California, Los Angeles, United States Paper Title: “Reduction in the Number of Animals and the Evaluation Period for the Positive Control Group in Tg.rasH2 Short-Term Carcinogenicity Studies.” ACT Furst Award The Furst Award, established through a generous contribution from the late Dr. Arthur Furst, is given to the best student poster presentation at the Annual Meeting. The Furst Award is determined by a panel of judges during a dedicated judging session at the Annual Meeting but must participate in the separate judging poster session. 2014�������� Winner to be announced at the Awards Luncheon on Monday, November 10. 2013������� Julie Castaneda Institution: University of California, Los Angeles Poster Title: Distribution Patterns for the CB2 Receptor Vary between Human B-Cells, T-Cells, and Malignant Cell Lines 14 Call for 2015 Awards ACT Award Nominations are Now Open Each year, the American College of Toxicology awards well-deserving individuals who have contributed to the field of toxicology and/or to ACT. Visit the ACT website for the Awards Nomination Form and the full list of awards offered. Send completed application, and CV if applicable, to [email protected] attention “Awards Nomination.” Deadline is March 31, 2015 15 2014 Awards Ceremony and Luncheon Monday, November 10 12:00 Noon–2:00 PM Regency Hall (Open Event, All Registrants Invited) Join us as we celebrate this year’s outstanding awardees for their efforts to advance the College and the field of toxicology. ACT’s Distinguished Scientist Award recipient Dr. Anthony Dayan will provide the keynote presentation. Poster Session and Reception Monday, November 10 5:30 PM–7:00 PM Grand Cypress Ballroom Sponsored by SAGE Publications (Open Event, All Registrants Invited) Discuss the latest findings and methodology with poster presenters and network with exhibitors and fellow colleagues during a reception with wine and light fare. 16 th 35 American College of Toxicology Annual Meeting 2014 PROGRAM AGENDA Continuing Education (CE) courses are 3.5 hours each and are held either Sunday morning (AM) or Sunday afternoon (PM) with the exception of the Study Director Short Course, which runs 7.25 hours (Study Director Short Course fee includes a boxed lunch). Preregistration is required and seating is limited. CONTINUING EDUCATION COURSE Study Director Short Course 8:00 AM–3:15 PM Regency Hall SESSION CHAIRS: Barbara J. Mounho-Zamora, ToxStrategies, Inc., Bend, OR, and Heather Dale, Covance Laboratories Inc., Madison, WI This Continuing Education course will review the essential aspects of the role and responsibilities of the study director. For the first half of the course, the presentations will include topics relevant for study directors including a review and history of the GLP regulations, managing and addressing the challenges of multisite studies, and selection of relevant animal models for toxicology studies. The second half of the course will include more detailed review of specific toxicology studies, including immunotoxicology, reproductive and developmental toxicology, and safety pharmacology. This course is an excellent introduction for the new study director, as well as a refresher course for experienced study directors. All students will receive a certificate for course participation that can be used for continuing education credits. 8:00 AM–8:10 AM Introduction: The Study Director Barbara J. Mounho-Zamora, ToxStrategies Inc., Bend, OR 8:10 AM–8:50 AM GLP Regulations: History and Roles of the Study Director Barbara Randolph, Biotechnical Services, Inc., Chattaroy, WA 8:50 AM–9:30 AM Animal Models for Toxicology Studies Scott E. Boley, MPI Research, Mattawan, MI 9:30 AM–10:10 AM Managing Multisite Studies Tom Beck, SNBL USA, Ltd., Everett, WA 10:10 AM–10:30 AM Break 10:30 AM–11:10 AM Immunotoxicity Assessment in Preclinical Safety Studies Greg Bannish, Huntingdon Life Sciences, East Millstone, NJ 11:10 AM–11:50 AM Safety Pharmacology in Human Pharmaceutical Development R. Dusty Sarazan, Data Science International, St. Paul, MN 11:50 AM–12:50 PM Lunch (boxed lunch provided) 12:50 PM–1:40 PM Reproductive Toxicology Alan M. Hoberman, Charles River Laboratories, Horsham, PA 1:40 PM–2:20 PM Clinical and Anatomic Pathology for Standard Preclinical Toxicology Studies: Data Integration and Interpretation Kevin McDorman, Charles River Laboratories, Frederick, MD 2:20 PM–3:00 PM What It Means to Be a Study Director—Managing the Issues and the Unexpected Heather Dale, Covance Laboratories Inc., Madison, WI 3:00 PM–3:15 PM Wrap Up 17 SUNDAY Supported by an educational donation provided by: Charles River Laboratories th 35 American College of Toxicology Annual Meeting 2014 Morning CE Courses CONTINUING EDUCATION COURSE— CE 1 Best Practices in Toxicologic Pathology 8:00 AM–11:30 AM Regency Hall SESSION CHAIRS: Kenneth A. Schafer, Vet Path Services, Inc., and Robert C. Sills, NTP/NIEHS, Research Triangle Park, NC SUNDAY Sponsored by the Society of Toxicologic Pathology and Supported by educational donations provided by: Experimental Pathology Laboratories, and Vet Path Services, Inc. During this session, Society of Toxicologic Pathology (STP) members will present information from a few of the key guidance documents. The guidance documents that have been selected are those that deal with basic issues in study design and interpretation that toxicologists and pathologists can use in their conduct and reporting of toxicity studies. STP has provided a number of best practice guidance documents, STP recommendations, and points to consider in the “Regulatory Forum” section of the society's journal, Toxicologic Pathology. These publications are intended to provide guidance to toxicologic pathologists and other interested scientists on issues that involve Data Interpretation/Management, Study Design, Study Reports/Peer Review and several guidance documents dealing with organ systems and tissues that require unique approaches for evaluation. The STP strives to collaborate with allied global organizations for many of these position papers. 8:00 AM–8:10 AM Introduction: Regulatory Forum: Best Practices, Points to Consider and STP Recommendations Robert C. Sills, NTP/NIEHS, Research Triangle Park, NC 8:10 AM–8:50 AM Use of Animal Models of Human Disease for Nonclinical Safety Assessment of Novel Pharmaceuticals Ronnie L. Yeager, AbbVie, Inc., North Chicago, IL 8:50 AM–9:35 AM Assessment of Circulating Hormones in Nonclinical Toxicity Studies: General Concepts and Considerations Dinesh Stanislaus, GlaxoSmithKline, King of Prussia, PA 9:35 AM–10:10 AM Break 10:10 AM–10:50 AM Evaluation of Organ Weights for Rodent and Non-Rodent Toxicity Studies Kenneth A. Schafer, Vet Path Services, Inc., Mason, OH 10:50 AM–11:30 AM Pathology Peer Review in GLP Toxicology Studies Rani Sellers, Albert Einstein College of Medicine, Bronx, NY 18 th 35 American College of Toxicology Annual Meeting 2014 CONTINUING EDUCATION COURSE—CE 2 Regulatory Toxicology—In the FDA and Beyond 8:00 AM–11:30 AM Palm SESSION CHAIRS: Patricia Frank, Patricia Frank & Associates, Inc., Evanston, IL and Shayne C. Gad, Gad Consulting Services, Cary, NC Supported by educational donations provided by: Gad Consulting Services and Patricia Frank & Associates, Inc. 8:00 AM–8:10 AM Introduction: Regulatory Toxicology Patricia Frank, Patricia Frank & Associates, Inc., Evanston, IL 8:10 AM–8:50 AM Interacting with FDA—A Regulatory Specialist's Viewpoint Stacy N. Suberg, RBR, Ltd., Woodbury, MN 8:50 AM–9:35 AM Interacting with CDER Patricia Frank, Patricia Frank & Associates, Inc., Evanston, IL 9:35 AM–10:05 AM Break 10:05 AM–10:45 AM Interactions with CBER Lauren Black, Charles River Laboratories, Reno, NV 10:45 AM–11:25 AM Interacting with CDRH—The Devices Division of the FDA Shayne C. Gad, Gad Consulting Services, Cary, NC 19 SUNDAY Many toxicologists are intimately involved in the design, conduct, and reporting of nonclinical studies. However, many of these professionals have a limited understanding of what happens to these reports after the studies have concluded. The aim of this course is to discuss the details of regulatory toxicology both from the perspective of regulatory affairs specialists and regulatory toxicology with an emphasis on drugs and devices. A discussion of the role that toxicologists play in the regulatory environment will be presented considering both US and ex-US regulations. A regulatory affairs specialist will explain what regulators expect from toxicologists and regulatory toxicologists will explain their role in interactions with drug, biologic and device regulatory bodies. A panel discussion of all the presenters will answer specific questions from the participants. th 35 American College of Toxicology Annual Meeting 2014 CONTINUING EDUCATION COURSE—CE 3 Drug Line Extensions: Nonclinical Testing and Solutions 8:00 AM–11:30 AM Regency Hall SESSION CHAIRS: David R. Jones, MHRA, London, UK and Paul Baldrick, Covance Laboratories Ltd., Harrogate, UK Supported by an educational donation provided by: the American College of Toxicology SUNDAY This Continuing Education course will examine what nonclinical testing needs to be considered for drug line extensions and/or new formulations. Talks will include EU and US regulatory agency experiences in this area along with a perspective on nonclinical strategy and solutions for line extension development. In addition, case examples will be given for which additional nonclinical testing was needed for a change to the original drug. The session will appeal to toxicologists working in the pharmaceutical arena and also possibly consumer healthcare products. 8:00 AM–8:05 AM Introduction Paul Baldrick, Covance Laboratories Ltd, Harrogate, UK, and David R. Jones, MHRA, London, UK 8:05 AM–8:40 AM EU Experiences with Line Extensions: From Initial Regulatory Contact to Marketing Application David R. Jones, MHRA, London, UK 8:40 AM–9:15 AM US Experiences with Line Extensions: From Initial Regulatory Contact to Marketing Application Timothy J. McGovern, US Food and Drug Administration, Silver Spring, MD 9:15 AM–9:45 AM Break 9:45 AM–10:20 AM Nonclinical Strategy and Solutions for Line Extension Development Paul Baldrick, Covance Laboratories Ltd, Harrogate, UK 10:20 AM–10:55 AM Nonclinical Program to Support Modification to an Existing Well-known Formulation Alan Christensen, Novo Nordisk A/S, Denmark 10:55 AM–11:30 AM Nonclinical Program to Support Change of Formulation and Administration Route of a GLP-1 Analogue Charlotte Weimann, Novo Nordisk A/S, Denmark 20 th 35 American College of Toxicology Annual Meeting 2014 CONTINUING EDUCATION COURSE—CE 4 Interpreting Adverse Clinical and Anatomic Pathology Results: Putting It All Together 8:00 AM–11:30 AM Regency Hall SESSION CHAIRS: Mary Jane Hinrichs, MedImmune, Gaithersburg, MD and Lila Ramaiah, Huntingdon Life Sciences, East Millstone, NJ Supported by educational donations provided by: MedImmune and the American College of Toxicology 8:00 AM–8:10 AM Introduction Mary Jane Hinrichs, MedImmune, Gaithersburg, MD 8:10 AM–9:00 AM Relations, Associations, Interactions and Causations: Principles for Correlating Anatomic Pathology (AP) and Clinical Pathology (CP) Findings in Toxicology Studies Lila Ramaiah, Huntingdon Life Sciences, East Millstone, NJ 9:00 AM–9:40 AM Interpretation of Toxicity Findings through the Combination of Clinical and Anatomic Pathology Data: Inflammation/Immunotoxicology William Iverson, MedImmune, Gaithersburg, MD, and Elizabeth Skuba, Novartis Institutes for Biomedical Research, East Hanover, NJ 9:40 AM–9:55 AM Break 9:55 AM–10:40 AM Interpretation of Toxicity Findings through the Combination of Clinical and Anatomic Pathology Data: Peripheral Blood Cytopenias William Iverson, MedImmune, Gaithersburg, MD, and Elizabeth Skuba, Novartis Institutes for Biomedical Research, East Hanover, NJ 10:40 AM–11:30 AM Diagnostic Utility and Interpretation of Traditional and Nontraditional Biomarkers of Kidney and Liver Injury Daniela Ennulat, GlaxoSmithKline, King of Prussia, PA 21 SUNDAY This course will discuss the integrative approach required to interpret safety findings in toxicology studies. Specific focus will be placed on the need to evaluate the data in its entirety, as neither clinical nor anatomic pathology can be relied upon in isolation to fully understand the relationship between study findings and the test article. As such, the course content will involve a collaborative effort between clinical and anatomical pathologists, who will discuss how they work together to interpret and draw correlations between clinical pathology and anatomic pathology safety findings. The approaches used to interpret organspecific findings and diagnose common systemic physiologic processes will be described using a combination of general principles and case examples. This course is intended for those interested in expanding their understanding of the approaches used to interpret test-article related findings in nonclinical safety studies. th 35 American College of Toxicology Annual Meeting 2014 EXHIBITOR-HOSTED PROGRAM 12:00 Noon–12:55 PM Regency Hall See page 137 for more information. 12:00 Noon–1:00 PM New Member Luncheon (By invitation only) Hydrangea Afternoon CE Courses SUNDAY CONTINUING EDUCATION COURSE—CE 5 Toxicology and Pathology of the Respiratory System 1:00 PM–4:30 PM Regency Hall SESSION CHAIRS: Daniel T. Kirkpatrick, WIL Research, Ashland, OH and Torrie A. Crabbs, Experimental Pathology Laboratories, Inc., Research Triangle Park, NC Supported by educational donations provided by: Experimental Pathology Laboratories, Inc. and WIL Research The respiratory tract is a common target organ in toxicology studies. In order to determine the toxicological significance of effects in the nose, larynx, trachea, and lung, it is important to understand the basic structure and function of these tissues. Designing protocols for inhalation toxicology studies also requires knowledge of the current methods for inhalation exposure of laboratory animals and evaluating respiratory structures and function. The first presentation will cover inhalation exposure systems and administration techniques, and methods for generation and characterization of exposure atmospheres. The remainder of the session will include detailed reviews of nose, larynx, trachea, and lung of common laboratory species. There will also be a discussion on biomarkers, different tools used to determine related effects, and spontaneous and induced changes observed in each part of the respiratory tract. 1:00 PM–1:10 PM Introduction Daniel T. Kirkpatrick, WIL Research, Ashland, OH 1:10 PM–1:50 PM Considerations in Assessing the Impact of the Aerosolization of Drugs and Toxicants in Respiratory Toxicology: It Is All About the Dose Matthew D. Reed, Lovelace Respiratory Research Institute, Albuquerque, NM 1:50 PM–2:35 PM Biomarkers for Respiratory Tract Toxicity Evaluation James Michael Randazzo, WIL Research, Ashland, OH 2:35 PM–3:10 PM Break 3:10 PM–3:50 PM Anatomy and Toxicologic Pathology of the Upper and Lower Respiratory Tract Torrie A. Crabbs, Experimental Pathology Laboratories, Inc., Research Triangle Park, NC 3:50 PM–4:30 PM Immunohistochemical, Morphometric, and Stereological Evaluation of the Respiratory Tract Danielle L. Brown, WIL Research, Hillsborough, NC 22 th 35 American College of Toxicology Annual Meeting 2014 CONTINUING EDUCATION COURSE—CE 6 (Q)SAR—A Tool for the Toxicologist 1:00 PM–4:30 PM Regency Hall SESSION CHAIRS: Samantha Gad-McDonald, Gad Consulting Services, Raleigh, NC and Thomas J. Steinbach, Experimental Pathology Laboratories, Inc., Raleigh, NC Supported by educational donations provided by: Experimental Pathology Laboratories, Inc. and Gad Consulting Services 1:00 PM–1:45 PM (Q)SAR Basics: Understanding (Q)SAR Models and their Predictions Naomi L. Kruhlak, US Food and Drug Administration, Silver Spring, MD 1:45 PM–2:30 PM Practical Applications of (Q)SAR in Toxicology Samantha Gad-McDonald, Gad Consulting Services, Raleigh, NC 2:30 PM–3:00 PM Break 3:00 PM–3:45 PM (Q)SAR and the Regulator Mark W. Powley, US Food and Drug Administration, Silver Spring, MD 3:45 PM–4:30 PM Applications and Pitfalls of (Q)SAR in Safety Assessments Nigel Greene, Pfizer Inc., Groton, CT 23 SUNDAY (Quantitative) Structure-Activity Relationships (Q)SAR methodologies use predictive computer modeling based on pre-defined rules or statistical tools to correlate biologic activity with the molecular structure or properties of a compound. (Q)SAR has applications in risk assessment, drug discovery, and regulatory decision making. Pressure within industry to reduce the cost of drug development and societal pressure for government regulatory agencies to produce more accurate and timely risk assessment of drugs and chemicals has necessitated the use of (Q)SAR. Producing a high-quality (Q)SAR model depends on many factors including the choice of statistical methods and descriptors, and the quality of the data input into the model. Understanding how a (Q)SAR model is developed and applied is critical to the successful use of such a powerful tool. This session will cover, from a toxicologist’s perspective, how to interpret (Q)SAR results; how regulatory agencies use and interpret (Q)SAR; practical applications of (Q)SAR in toxicology; and potential future application of (Q)SAR. th 35 American College of Toxicology Annual Meeting 2014 CONTINUING EDUCATION COURSE—CE 7 Managing Anti-Drug Antibody Responses during Biologics Drug Development 1:00 PM–4:30 PM Palm SESSION CHAIRS: Joerg Bluemel, Genentech, Inc., South San Francisco, CA, and Suzanne Wolford, Covance Laboratories Inc., Madison, WI SUNDAY Supported by educational donations provided by: Covance Laboratories Inc. and MedImmune Immunogenicity of biotechnology-derived pharmaceuticals is frequently observed in nonclinical studies but can also affect clinical development. Anti-Drug antibodies (ADA) are endogenous antibodies directed against a therapeutic protein, which can also cross-react to other endogenous epitopes. The presence of ADA can have various biological consequences ranging from no biological relevance, to impact on pharmacokinetics or pharmacodynamic or even the occurrence of toxicities due to ADA. The impact of the occurrence of ADA on development strategies, their relevance for risk assessment and the necessity of mitigation strategies is highly dependent on their functional consequences. 1:00 PM–1:45 PM Biology, Relevance, and Impact of Anti-Drug Antibodies (ADA) during Development of Biopharmaceuticals Janice Lansita, ToxStrategies, Inc., Baltimore, MD 1:45 PM–2:30 PM Bioanalytical Characterization of Anti-Drug Antibodies (ADA) Ian Skitt, Covance Laboratories Inc., North Yorkshire, United Kingdom 2:30 PM–3:00 PM Break 3:00 PM–3:45 PM Toxicological Consequences of Anti-Drug Antibodies (ADA), Their Correlation to Presence of ADA and Impact on Risk Assessment T. Scott Manetz, MedImmune, Gaithersburg, MD 3:45 PM–4:30 PM Strategies to Overcome /Minimize the Formation of Anti-Drug Antibodies (ADA) in Nonclinical Studies Simone M. Nicholson, MedImmune, Gaithersburg, MD 24 th 35 American College of Toxicology Annual Meeting 2014 CONTINUING EDUCATION COURSE—CE 8 Metabolites: Guidance and Considerations in Drug Development 1:00 PM–4:30 PM Regency Hall SESSION CHAIRS: Holly Dursema, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT and Kate E. Lane, Cubist Pharmaceuticals Inc., Lexington, MA Supported by an educational donation provided by: Cubist Pharmaceuticals Inc. 1:00 PM–1:45 PM Metabolites in Drug Development: Current Guidance Clay B. Frederick, CBF Safety Assessment Consulting LLC, Dresher, PA 1:45 PM–2:30 PM Metabolites in Drug Development: Assessment of Metabolites Present in Animals and Humans John-Michael Sauer, Critical Path Institute, Tuscon, AZ 2:30 PM–3:00 PM Break 3:00 PM–3:45 PM Disproportionate/Unique Metabolite: Case Study Douglas A. Keller, Sanofi US, Bridgewater, NJ 3:45 PM–4:30 PM Metabolites in Drug Development: An FDA Perspective Carol M. Galvis, US Food and Drug Administration, Silver Spring, MD 6:30 PM–8:30 PM Welcome Reception (Ticketed Event) See page 10 for more information 25 The Wilderness In the Event of Rain: Portico West SUNDAY This course will review guidance that currently exists for nonclinical assessment of metabolites discovered during drug development. This topic has matured over the last five years, therefore a review of the existing guidance and their application is timely. The objective of this course is to provide participants with an understanding of guidance and tools they may use to investigate human metabolites encountered during development. The first speaker will review current guidance documents and present ways to incorporate those assessments into drug development. The second speaker will discuss use of this guidance in assessment of a metabolite that is present in both animals and humans. The third speaker will discuss assessment of a disproportionate human metabolite and/or unique human metabolite. The final speaker will provide FDA perspective on evaluation of human metabolites. Time will be allotted for discussion of each aspect. The staging of the speakers has been done to familiarize the audience with guidance and progress them through the challenges that may be encountered in metabolite assessments. This course will be of interest primarily to members in the pharmaceutical field. Experts in the Exhibit Hall Monday Morning Break: Plenary Lecturer Deborah Blum will be in the ACT Member Lounge to sign your copy of The Poisoner’s Handbook Afternoon Break: Come congratulate ACT’s Distinguished Scientist Awardee Anthony Dayan in the ACT Member Lounge Tuesday Come to the Exhibit Hall in the afternoon and meet the Editor of ACT’s International Journal of Toxicology, Mary Beth Genter at the SAGE Exhibit Booth (#304) She’ll have tips for: • publishing in the Journal • reviewing manuscripts • preparing Symposium Overview manuscripts ACT Members’ Meeting Tuesday, November 11, 5:00 PM–6:30 PM ACT Members Only Join your colleagues for the annual Members’ Meeting. Hear important reports from: • President Drew A. Badger • President-Elect Mary Ellen Cosenza • Treasurer Alan Hoberman • Membership Committee Chair Timothy McGovern • Nominating Committee Chair Robin C. Guy • Publications Committee Chair Mary Beth Genter • And new initiatives! Be an active member of the American College of Toxicology and participate! No registration required. 26 th 35 American College of Toxicology Annual Meeting 6:45 AM–8:00 AM Past Presidents' Breakfast 2014 Gardenia (By invitation only) Monday Morning Sessions PLENARY LECTURE 1 The Poisoner's Guide to Life 8:00 AM–8:55 AM Regency Hall DEBORAH BLUM PULITZER-PRIZE WINNER AND AUTHOR OF THE POISONER’S HANDBOOK: MURDER AND THE BIRTH OF FORENSIC MEDICINE IN JAZZ AGE NEW YORK Deborah Blum is the Pulitzer Prize–winning author of five books, most recently The Poisoner’s Handbook: Murder and the Birth of Forensic Medicine in Jazz Age New York, which was named as one of the top one-hundred books of 2010 by Amazon. A highly acclaimed science journalist and former president of the National Association of Science Writers, Deborah is also co-editor of A Field Guide for Science Writers. Her work has been translated into more than a dozen languages, optioned for film, and has appeared in numerous publications, including The New York Times, Slate, The Washington Post, the Los Angeles Times, The Guardian, Discover, and Health. She also writes about chemistry and culture for the Public Library of Science in her blog, Speakeasy Science. MONDAY 27 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 1 From Mice to Men, Development of Human Drugs and Biologics under the Animal Rule CHAIR: John V. Wade, Battelle, 9:00 AM–12:00 Noon Palm Columbus, OH CO-CHAIR: Owen McMaster,CDER/US Food and Drug Administration, Silver Spring, MD Supported by an educational donation provided by: Battelle Memorial Institute The animal efficacy rule was finalized in 2002 and applies to the development of drugs to reduce or prevent serious lifethreatening conditions caused by exposure to lethal or permanently disabling toxic agents; including chemical, biological, radiological, or nuclear (CBRN) substances. The studies conducted under the animal rule are used to support approval of treatments, as human clinical trials are generally not feasible or unethical. The characterization and development of animal efficacy models is critical to the success of drugs or vaccines being developed under the animal rule. This session will provide an introduction to the animal rule, the qualification requirements for animal efficacy models development, and case studies of development programs for therapeutics and vaccines. S1–1 9:00 AM–9:35 AM Overview of the Animal Rule Barbara J. Mounho-Zamora, ToxStrategies, Bend, OR S1–2 9:35 AM–10:05 AM Animal Model Qualification Process and the Medical Countermeasures Initiative Owen McMaster, US Food and Drug Administration, Silver Spring, MD S1–3 10:05 AM–10:35 AM Development of Brincidofovir for the Treatment of Smallpox under the Animal Rule MONDAY Lars C. Trost, Chimerix, Durham, NC S1–4 10:35 AM–10:55 AM Break 10:55 AM–11:30 AM Utilizing Well-Characterized Drug Development Tools for The Assessment of Medical Countermeasures: Anthrax Antitoxins Gabe Meister, Battelle, Columbus, OH S1–5 11:30 AM–12:00 Noon Development of an Anthrax Vaccine under the Animal Rule: A Case Study Barbara J. Mounho-Zamora, ToxStrategies, Bend, OR 28 th 35 American College of Toxicology Annual Meeting S1–1 The Animal Rule (21 CFR 314.600 for drugs; 21 CFR 601.90 for biological products) provides regulation and a licensure pathway that allows FDA to grant marketing approval for candidate therapeutic products where it is neither ethical nor feasible to conduct traditional human efficacy studies. In such cases, a therapeutic product may be granted marketing approval based on adequate and well-controlled animal studies for the demonstration of clinical efficacy. There are several novel biological products under clinical development that may fall under the Animal Rule. For example, vaccines against microbial pathogens that might be used in biological warfare fall under the Animal Rule, and use animal studies (vs. human) may be performed for the demonstration of clinical efficacy. Several criteria, however, such as characterization of the pathophysiological mechanism of toxicity of the pathogen and selection of endpoints of efficacy used in the animal studies, must be fulfilled in order for the FDA to consider using animal studies to provide substantial evidence of clinical effectiveness. This presentation will provide an overview of the Animal Rule and challenges in bridging animal protection data to human efficacy. S1–2 S1–3 The talk will discuss potential challenges in developing drugs under the Animal Rule by tracing the development of Brincidofovir for treatment of smallpox, including technical and project management issues. The development of relevant animal models, which is frequently in collaboration with academic or government institutions, and the subsequent transfer of models to qualified nonclinical testing facilities capable of conducting GLP-compliant studies will be discussed. Challenges in the design of animal efficacy protocols, in particular, establishing inclusion/exclusion criteria, study blinding, and objective euthanasia criteria, of particular importance to studies in which prevention of mortality is the primary endpoint. The talk will conclude by touching briefly on the unique regulatory challenges associated with the Animal Rule, lack of precedence often chief among them, and scaling to a human dose from doses proven effective in animal models. S1–4 Battelle, with funding support from NIAID DMID/OBRA and BARDA, has worked to establish well-characterized drug development tools (small and large animal models) to assess the effectiveness of anthrax adjunct therapies in support of regulatory submissions. Preliminary studies assessed the clinical and physiological parameters following exposure to Bacillus anthracis Ames strain spores in New Zealand White (NZW) rabbits and cynomolgus macaques to build a clinical profile of disease and define appropriate therapeutic markers utilized as “triggers” for treatment in subsequent efficacy studies. Pilot efficacy studies validated the use of specific diagnostic markers and pivotal efficacy studies were conducted to further confirm the use of the markers. An FDA Anti-Infective Drugs Advisory Committee was convened to discuss the Biologic License Application (BLA) for the first monoclonal antibody (antitoxin) to treat inhalational anthrax. During the discussion, and confirmed by the voting results, the Advisory Committee agreed that the effectiveness observed in the nonclinical efficacy studies would be predictive of efficacy in humans. The US FDA approved the first antitoxin for the treatment of inhalational anthrax on December 14, 2012. Conforming to the requirements set forth in 21 CFR 314.600 for drugs and 21 CFR 601.90 for biologicals, well-characterized drug development tools are needed for drugs to be approved to treat serious or life-threatening conditions caused by exposure to lethal or permanently disabling toxic biological, chemical, radiological, or nuclear substances. S1–5 Anthrax is a serious disease that is highly contagious and can affect both animals and humans. Anthrax is caused by bacteria called Bacillus anthracis (B. anthracis), which is a gram positive, facultative anaerobic, rod-shaped, highly pathogenic bacterium that has the ability to form endospores. Due to the durability of the endospores, B. anthracis can be easily aerosolized, and therefore, has the potential to be weaponized and pose a threat for use in biological warfare. This presentation will review a case study of the development of an anthrax vaccine under the Animal Rule, including the animal studies demonstrating efficacy as well as the nonclinical safety assessment studies. 29 MONDAY Acceptable animal models have not been developed for many chemical, biological, radiological, nuclear, and infectious disease threat agents. The FDA's Animal Model Qualification Program (AMQP) encourages the qualification of animal models that simulate the human disease or condition resulting from these agents. For an animal model to be qualified, there must, among other things, be a well-understood pathophysiological mechanism of the challenge agent and its attenuation by the medical countermeasure (MCM). The AMQP evaluates animal models based on data from the human disease or condition and data from animal model natural history studies. Qualification of an animal model implies that FDA has accepted that a specific animal species given a specific challenge agent by a specific route produces a disease process or condition that corresponds to the human disease or condition of interest. This qualified model may subsequently be used for efficacy testing in development programs for multiple investigational drugs for the same disease or condition. This session will discuss FDA's Animal Model Qualification Program including (1) The conditions under which an animal model may be qualified, (2) Characterization of the animals and challenge agent (3) Required procedural information (4) Triggers for intervention (5) Primary and secondary endpoints. This talk will close with the activities of the medical countermeasures initiative since its formation in 2010. 2014 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 2 What the AEL is the NOAEL? 9:00 AM–12:00 Noon Grand Cypress Ballroom A CHAIR: Joy A. Cavagnaro, Access BIO, Boyce, VA CO-CHAIR: Lisa D. Beilke,Toxicology Solutions, Inc., San Diego, CA Supported by educational donations provided by: MPI Research and the American College of Toxicology Is the NOAEL different in the eyes of a pathologist vs. a toxicologist, a clinical reviewer/FIH clinical investigator—the animal? Does clinical indication impact NOAEL and/or application of a NOAEL to a clinical indication? The pharmacological activity that is outside the projected therapeutic profile of a new drug may include a spectrum of functional effects, which are not hazardous and are reversible. Minor, albeit undesirable, effects should therefore be distinguished from unintended reactions that are pertinent to toxicity and that may lead to target organ effects. In this way, pharmacodynamic activity, which is integral to the therapeutic action of a drug, may be viewed differently from activity, which is not indispensable for efficacy. In this respect, structural (morphological) changes in organs or tissues of potentially irreversible nature are of particular concern. For that reason, histopathology has remained the most consistent criterion by which target organs are identified and NOAELS are set. This session will draw out the common points that scientists often debate on this subject, particularly as exemplified by case examples. There will also be an interactive session where data will be presented and the audience will have an opportunity to vote on whether a NOAEL has been demonstrated. S2–1 9:00 AM–9:40 AM Recommended Practices for Defining and Communicating Adversity for Nonclinical Study Data James A. Popp, Stratoxon LLC, Lancaster, PA S2–2 9:40 AM–10:20 AM Determining Adverse Histopathology using a Weight of Evidence Approach MONDAY Daniel J. Patrick, MPI Research, Mattawan, MI S2–3 10:20 AM–10:40 AM Break 10:40 AM–11:20 AM NOAEL Selection and Risk Assessment for Biologics and Other Nontraditional Small Molecules Laine Peyton Myers, OND/OAP/DAVP/CDER/US Food and Drug Administration, Silver Spring, MD S2–4 11:20 AM–12:00 Noon When Should Pharmacology/Superpharmacology Be Considered Adverse (Ever?) Christopher Horvath, bluebirdbio, Cambridge, MA 30 th 35 American College of Toxicology Annual Meeting S2–1 Identifying and communicating a NOAEL continues to be a central issue related to toxicology studies prepared for regulatory submission. As the term indicates, the NOAEL is based on the identification of an adverse effect. The lack of concurrence in what constitutes an adverse effect and the inconsistencies around the communication of adverse effects continue to be the basis of difficulties and differences of opinion amongst practicing toxicologist and toxicologic pathologists, despite many published attempts to address the topic. The Society of Toxicologic Pathology (STP) developed a set of recommended practices to serve as a guide for determining and communicating adversity in nonclinical studies more effectively with the goal of reducing confusion regarding the use of the NOAELs in the future. In addition, the recommended practices address the role of the toxicologist and toxicologic pathologist in assessing potential human risk based on adverse findings. The STP recommended practices will be presented advocating a more consistent approach to addressing adverse findings so that reliance on the NOAEL may not be needed over time. S2–3 A NOAEL is commonly used as a benchmark for the first-inhuman studies for pharmaceuticals. NOAEL determinations hinge on multiple endpoints (e.g., histopathology panel, in vitro-binding assays). Many times, the Sponsor and the Agency will agree on the endpoints and the First in Human dosing will proceed as scheduled. For small molecules, the path forward is relatively uncomplicated. On occasion, the FIH dose for these small molecules may be confounded (e.g., host resistance study outcomes, immunomodulator effects). Moreover, for biological products and other nontraditional small-molecules, there are certain challenges during drug development that may confound defining a NOAEL. Biologic products have confounding variables (small group size, immunogenicity, species specificity/relevance) that may confound data interpretation. Furthermore, some applications include data using a surrogate molecule and/or a transgenic species used in calculating a NOAEL. Lastly, other molecules (e.g., oligonucleotide-based drugs) can often present speciesspecific issues that may confound the determination and/ or interpretation of the NOAEL. This talk will discuss unique challenges to NOAEL selection and interpretation of the NOAEL into risk assessment during drug development. S2–4 For biologics, which must be tested in a pharmacologicallyrelevant species, toxicology studies generally assess safety by using pharmacologic (comparable to anticipated human doses) and superpharmacologic doses. The intent is to maximize the pharmacodynamic response and to explore whether secondary or “super” pharmacology occurs at higher doses. Thus, at every dose level some effects will be observed. Establishing a NOAEL in these settings may be challenging. At pharmacologically-relevant doses, the intended pharmacology may not be adverse, while at higher doses, the intended effects may be adverse, and at some doses it may be unclear whether the observed effects are adverse. Additionally, the same effects may be considered acceptable (nonadverse) in one patient population (e.g., cancer) and considered not acceptable (adverse) in another population (e.g., autoimmune disease). An additional complication for determination of NOAELs for biologics is the occurrence of immunogenicity and immunerelated events. Most human biologics are immunogenic in animals, including monkeys. Anti-Drug antibodies (ADA) can precipitate a wide range of effects in animals that may have little or no relevance to human safety assessment. For example, ADA may be associated with evidence of immune stimulation, formation of intravascular or tissue immune complexes, infusion or hypersensitivity reactions, development of glomerulonephritis and/or neutralization of an endogenous protein. Some of these events may be adverse in animals, and would be adverse if they were to occur in humans, yet their occurrence in animals is not predictive of the risk to humans. Due to the immune-mediated nature of such events, when they occur they are often not dosedependent (e.g., are idiosyncratic). Paradoxically, the highest dose might provide a NOAEL while the lowest dose might not. In some cases, the immune-related events can be clearly demonstrated to be unique to animals, in which case the any immune-based NOAEL might not be relevant to humans. This talk will attempt to explore such issues, and may raise more questions than it answers. 31 MONDAY S2–2 This presentation will discuss the thought process a toxicologic pathologist might employ when distinguishing histopathologic changes that could be considered adverse, nonadverse or somewhere in between. The overall determination is based on a weight of evidence approach utilizing multiple criteria, as well as the subjective scientific opinion of the pathologist. Case examples with representative photomicrographs, associated clinical pathology, and other relevant data will be presented. 2014 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 3 Systems Toxicology: The Future of Risk Assessment CHAIR: John Michael Sauer, Predictive 9:00 AM–12:00 Noon Magnolia Safety Testing Consortium (PSTC), Tucson, AZ CO-CHAIR: A. Wallace Hayes,Harvard School of Public Health, Andover, MA Supported by an educational donation provided by: Philip Morris International R&D The vision of the National Research Council report on Toxicity Testing in the 21st Century is becoming a reality as modern high-content and high-throughput technologies are applied. The approach can improve the toxicological assessment of a wide range of substances including those developed for example by the pharmaceutical, chemical, cosmetic, and tobacco industries. This symposium will share the results from leading researchers in the field to underpin the revolution that is taking place in toxicology. S3–1 9:00 AM–9:40 AM Translating Systems Toxicology-Based Assessment into Risk Management Thomas Hartung, John Hopkins University, Bloomberg School of Public Health, Baltimore, MD S3–2 9:40 AM–10:20 AM Experimental Enablers: The Pan-Omics View Marcel Leist, University of Konstanz, Germany S3–3 10:20 AM–10:40 AM Break 10:40 AM–11:20 AM Computational Enablers: From Data Integration to Dynamic Modeling Thomas B. Knudsen, US Environmental Protection Agency, Research Triangle Park, NC MONDAY S3–4 11:20 AM–12:00 Noon Implementing Systems Toxicology Approaches Julia Hoeng, Philip Morris International R&D, Neuchâtel, Switzerland 32 th 35 American College of Toxicology Annual Meeting S3–1 This presentation is intended to set the scene for the symposium by defining Systems Toxicology, spelling out its aim(s) and explaining how it can and must be part of the new paradigm for risk assessment and risk management. S3–2 This presentation is intended to define key experimental enablers of Systems Toxicology and how they can be applied in various experimental settings from in vitro to clinical studies. It will cover the main *omics technologies as well as highcontent screening using in vitro systems. 2014 S3–3 This presentation is intended to define key computational enablers of systems toxicology and how they can be applied in various settings ranging from Big Data management to the simulation of complex pathways of toxicity and adverse outcomes in a virtual organism. S3–4 This presentation is intended to present how an integrated systems toxicology program can be implemented in an industry setting and present three case studies where it was applied in vitro, in vivo and in the clinic. MONDAY 33 th 35 American College of Toxicology Annual Meeting 12:00 Noon–2:00 PM Awards Ceremony and Luncheon 2014 Regency Hall (Included in registration fee) See Page 10 for more information. Monday Afternoon Sessions SYMPOSIUM 4 Antidrug Antibody-Independent Immune Responses to Biologics in Rodent Studies— Regulatory Success in Early Drug Development CHAIR: Krishna P. Allamneni,Jazz 2:00 PM–5:00 PM Palm Pharmaceuticals, Inc., Palo Alto, CA CO-CHAIR: Joy A. Cavagnaro,Access BIO, Boyce, VA Supported by an educational donation provided by: the American College of Toxicology Due to the advances in recombinant protein technologies and subsequent development of rodent-crossreactive biologic agents for therapeutic use in humans, a rising trend is noted for rodent toxicology studies for biologics. The development of binding, sustaining/neutralizing antidrug antibodies and subsequent safety or efficacy liabilities are well recognized. This symposium highlights the emergence of immune reactions in rodents that cannot be definitively attributed to immunogenicity of the biotherapeutic agents. A high-level overview of the pathophysiology of immune system in rodents, clinical presentation of immune reactions and differential diagnosis are discussed as a framework to assess risk. Plausible mechanisms of antidrug antibody-independent immune responses in rodents are explored. Available tools to evaluate, characterize, and/or confirm immune reactions specific to mice, rats, monkeys and their limitations are presented. Case examples of clinical development in the context of rodent-specific immune responses are presented. A comprehensive view of the regulatory perspective on such rodent-specific immune responses in the context of first-in-human risk assessment is presented. S4–1 2:00 PM–2:15 PM Overview MONDAY Krishna Allamneni, Jazz Pharmaceuticals, Inc., Palo Alto, CA S4–2 2:15 PM–2:50 PM Clinical Impact of Rodent-Specific Immune-Related Reactions to Human Proteins Joy A. Cavagnaro, Access BIO, Boyce, VA S4–3 2:50 PM–3:20 PM Non-ADA Mediated Anaphylactoid Reactions in Toxicology Studies with Rodents Krishna Allamneni, Jazz Pharmaceuticals, Inc., Palo Alto, CA S4–4 3:20 PM–3:55 PM Break 3:55 PM–4:25 PM Immunotoxicoloogy Tools to Characterize Species-Specific Immune Reactions Florence G. Burleson, Burleson Research Technologies, Inc., Morrisville, NC S4–5 4:25 PM–5:00 PM Regulatory Perspectives on the Rodent-Specific Immune Reactions Jane Sohn, US Food and Drug Administration, Silver Spring MD 34 th 35 American College of Toxicology Annual Meeting S4–1 Problem statement is presented along with industry trends on the prevalence of GLP studies in rodents for biologics. A high-level overview of the physiology of immune system in rodents, types of immune reactions, species differences, in-life, clinical pathology and histopathology presentation, differential diagnosis, relevance to humans is discussed as a framework to assess risk. S4–2 It is acknowledged in ICHS6(R1) that immunogenicity assessments are [only] conducted to assist in the interpretation of toxicity study results (e.g. immune-mediated reactions such as immune complex-related, vasculitis, anaphylaxis, etc.) and are not relevant in terms of predicting potential immunogenicity of human or humanized proteins. However as rodents are increasingly being defined as relevant species in early development programs there has been a higher incidence of unexpected fatal acute infusion reactions which may or may not correlate with antibody development that have negatively impacted clinical development. Case studies will be presented highlighting specific findings, consequences and risk mitigation strategies to support clinical use. S4–4 Biotherapeutics now make up greater than 50% of the pharmaceutical pipeline. Unintended adverse immune reactions following administration of biotherapeutics have been observed in animal studies and man. These adverse drug reactions may be hypersensitivity reactions that are antidrug antibody (ADA) mediated, or idiosyncratic drug reactions (IDRs). Non-ADA-mediated reactions include pseudoallergy and cytokine release syndrome. In these type of reactions the timing of observations, sample type, and timing of acquisition are critical parameters. This symposium will review these critical parameters, as well as the methods and assays currently available to evaluate, or try to predict, unintended adverse immune reactions. Gaps will be identified and future directions discussed. S4–5 Rodent toxicology studies are increasingly required for the development of biologic products as scientific advances have led to biologics with crossreactivity to rodents. The presentation will focus on the review of biologics products on a case-bycase basis. Case studies of biologics with crossreactivity to rats, with immune reactions will be presented. Each case will highlight hypothetical analysis of data, and the review process within CDER. 35 MONDAY S4–3 Multiple case studies are presented of clinical candidates that are not immunomodulatory in action. Clinical candidates demonstrated high nanomolar affinity to targets in rodents and in vivo functional activity in rodent models. Therefore, rodents were considered biologically relevant for preclinical safety assessment to enable first-in-human clinical trials. This presentation details the transient immune reactions noted in the toxicology studies with mice and rats. Results of the subsequent investigational studies conducted to evaluate the mechanistic basis of the immune reactions are discussed with a focus on plausible hypotheses and regulatory package for the clinical candidates. 2014 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 5 Use of Humanized Mouse Models in DMPK and Safety Testing of Compounds CHAIR: Alema Galijatovic Idrizbegovic,Merck CO-CHAIR: Charles Roland Wolf,University 2:00 PM–5:00 PM Magnolia & Co, West Point, PA of Dundee, Dundee, United Kingdom Supported by an educational donation provided by: Taconic The use of traditional laboratory animals to predict human outcomes in pharmacokinetics, drug-drug interaction or toxicity of compounds is often limited by the significant differences which exist between animals and man. In order to address this issue a lot of efforts have been made by various groups over the last years to generate humanized mouse models, which are more predictive for various aspects of drug responses in humans. One approach is the generation of genetically humanized mice expressing single or multiple human proteins involved in drug metabolism and disposition, such as xenobiotic receptors, drug metabolizing enzymes and transporters. Another strategy involves the replacement of mouse with human tissue, resulting, for example, in humanized mice for the liver and/or immune system. This symposium provides the current status and discusses the future perspectives in the field of humanized mouse models from the user’s perspective. It will be of interest to scientists involved in commercial drug development, particularly working in drug metabolism and toxicology, academic researchers, as well as regulators evaluating data generated during safety testing. Furthermore, with potential applications for risk assessment of chemicals and food ingredients, it will provide relevant information to scientists in the chemical and consumer healthcare industries. S5–1 2:00 PM–2:30 PM Nearly Human—The Potential of Chimeric Mice with Livers Colonized with Human Hepatocyte Ian D. Wilson, Imperial College London, South Kensington, London, United Kingdom MONDAY S5–2 2:30 PM–3:05 PM Application of Humanized Mouse Models to Study Anticancer Drug Efficacy and Toxicity Charles Roland Wolf, University of Dundee, Medical Research Institute, Ninewells, Dundee, United Kingdom S5–3 3:05 PM–3:35 PM Utility of Humanized Mice in the Assessment of Biological Drug Products Kristina Howard, US Food and Drug Administration, Silver Spring, MD S5–4 3:35 PM–3:55 PM Break 3:55 PM–4:25 PM Humanized Mouse Models—Applications in Preclinical Testing Alema Galijatovic-Idrizbegovic, Merck & Co, West Point, PA S5–5 4:25 PM–5:00 PM Regulatory Perspective on Alternative Animal Models Mark W. Powley, US Food and Drug Administration, Silver Spring, MD 36 th 35 American College of Toxicology Annual Meeting S5–1 There are now a number of commercially available mouse models where the liver has been largely repopulated with human hepatocytes. These so called "chimeric" humanized mice have an obvious potential for the investigation of hepatic human metabolism and toxicity of drugs and other xenobiotics. The emerging data suggest that such models do indeed provide "human-like" metabolic data and as such these chimeric animal models clearly merit further investigation. Our experiences of using liver humanized mouse models, based on colonization the livers of uPA-SCID or FRG mice with human hepatocytes, will be described together with selected published investigations that have employed these models. Particular emphasis will be given to applications in toxicology, and an attempt will be made to put these models into context with alternative methods, such as in vitro tools. The state of development of these chimeric mouse models will be considered and their advantages and current limitations will be discussed. S5–2 The efficacy of currently used and emerging anticancer drugs can often be intimately linked to their rate of metabolism by enzymes such as the cytochrome P450-dependent monooxygenases. Also, the effectiveness of these drugs is often limited by the emergence of drug resistance or drug side effects. 2014 S5–4 Transgenic mice humanized for key human proteins involved in metabolism, and chimeric mice with highly humanized livers can serve as models to address and evaluate metabolites formed uniquely in humans as well as models to evaluate endogenous metabolites to improve human safety prediction from preclinical species. In addition rasH2 transgenic mice carrying human c-Ha-ras oncogene in addition to the endogenous murine Ha-ras oncogene, which are highly susceptible to tumor formation, allow for rapid carcinogenicity evaluation as an alternative to 2-year mouse carcinogenicity studies, and are enabling significant changes to rodent carcinogenicity testing strategy being discussed through ICH. This talk will focus on the utility of these models to improve and accelerate preclinical development of molecules and reduce attrition of molecules in the clinical studies due to metabolism and toxicology. S5–5 This brief presentation will review the role of standard and alternative animal models in drug development. Examples of existing and potential applications as well as regulatory expectations of alternative models will be discussed. Focus will be placed on testing to support safety assessment. S5–3 Testing of biological drug products for safety and efficacy in animal models has been difficult to assess because common models such as rodents, canines and nonhuman primates do not necessarily share common biological receptors with humans. Immunologically humanized mice offer the ability to test the efficacy and safety of biological drug products in an in vivo model of the human immune system. This talk will present results of biological drug product testing in this model. 37 MONDAY We have been using mouse models humanized for the major P450s involved in drug disposition in man as well as the transcription factors, which regulate their expression to establish the role of these enzymes in defining the outcome of the anticancer drug therapy. Our work has involved studies into a range of tyrosine kinase inhibitors which are either hepatotoxic or where drug resistance is a major limiting factor in drug efficacy. In this presentation our work in this area, including studies on the B-Raf inhibitor vemurafenib, will be described. th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 6 Targeted Cancer Therapeutics: Concepts and Strategies to Improve Oncology Drug Development CHAIR: Vijayapal Reddy, Eli 2:00 PM–5:00 PM Grand Cypress Ballroom A Lilly and Company, Indianapolis, IN CO-CHAIR: Elaine Knight,National Cancer Institute, Rockville, MD Supported by an educational donation provided by: the American College of Toxicology Over the last decade, the focus for anticancer drug discovery has shifted from traditional cytotoxic agents (with significant and well characterized toxicities) to rationally designed, molecularly-targeted pharmaceuticals. Of the new anticancer drugs approved by the US Food and Drug Administration (FDA) since 2000, a majority have been targeted therapies, compared with only few traditional chemotherapeutic agents. The safety data derived from studies of approved targeted pharmaceuticals provide a wealth of information on the nature of toxicities identified in both nonclinical species and in patients. In many cases, the toxicity profile of a targeted therapeutic is closely linked to the biological consequences of modulating the intended targets. Continued advancement in our understanding of mechanisms of toxicity should provide a better understanding of the relationships between drug efficacy and toxicity, and possibilities for better clinical management of toxicity. This symposium will provide an overview of the advances in cancer biology that have facilitated discovery of anticancer drug targets or improved candidates for drug development. The symposium also will include an overview of approaches to nonclinical safety evaluation and tailored clinical evaluation of anticancer agents. Last, the session will include a regulatory perspective and regulatory challenges for both small and biologic targeted cancer pharmaceuticals. This topic should be interested to a broad range of attendees including those from industry, academia, government and clinical, and healthcare professionals. 2:00 PM–2:15 PM Introduction: Targeted Cancer Therapies: Concepts and Strategies to Improve Oncology Drug Development MONDAY Elaine Knight, NCI/National Institutes of Health, Rockville, MD S6–1 2:15 PM–2:50 PM Cancer Biology and the Discovery of Targeted Therapies Alan C. Rigby, ImClone Systems, a Wholly-owned subsidiary of Eli Lilly and Company, New York, NY S6–2 2:50 PM–3:25 PM Targeted Anticancer Pharmaceuticals: Off-Target and On-Target Toxicities Vijayapal Reddy, Eli Lilly and Company, Indianapolis, IN S6–3 3:25 PM–3:45 PM Break 3:45 PM–4:30 PM Transforming the NCI Experimental Therapeutic Program as It Relates to Clinical Evaluation of Targeted Therapies S. Percy Ivy, NCI/National Institutes of Health, Rockville, MD S6–4 4:30 PM–5:00 PM Regulatory Perspective on the Development of Targeted Anticancer Therapeutics Todd R. Palmby, CDER/US Food and Drug Administration, Silver Spring MD 5:30 PM–7:00 PM Poster Session and Reception See Page 10 for more information. 38 Exhibit Hall th 35 American College of Toxicology Annual Meeting S6–1 An overview of the advances in cancer biology that have facilitated discovery of anticancer drug targets or improved candidates for drug development. S6–2 Continued advancement in our understanding of mechanisms of toxicity of targeted anticancer pharmaceuticals should provide a better understanding of the relationships between drug efficacy and toxicity, and possibilities for better clinical management of toxicity. S6–3 determination must use greater than cycle 1 adverse events to established non-DLT based, but rather all grades of adverse events to define a safe and tolerable dose to be used in future studies. This presentation will present the challenges and approaches that need to be developed for development of MTAs. New and novel trial designs will need to be developed as drug development proceeds in the 21st century. S6–4 The development of anticancer therapeutics that are designed to affect specific pathways known to be modulated or deregulated in tumors or necessary for maintaining the tumor phenotype is an exciting concept that has had many successes in recent years. Nevertheless, many regulatory challenges arise in these drug development programs due to the specific nature of the products or diseases that they are being used to treat. This presentation will discuss ICH regulatory guidances S9, S6(R1) and M3(R2)relevant to the development of targeted anticancer therapeutics and when consideration should be given to the scope and specific recommendations of each. Regulatory challenges that FDA faces with these development programs include the often chronic administration in patients, use in adjuvant or first-line treatment settings, timing of nonclinical studies to support use in certain patient populations, developmental and reproductive toxicology studies with monoclonal antibodies, antibody-drug conjugates, immunomodulators and products affecting epigenetic pathways. Toxicities observed in clinical trials and in the post-marketing setting for recent development programs of small molecule targeted anticancer therapeutics were not always observed in nonclinical studies conducted with current animal models, indicating a need for better predictivity. Issues involved in identifying potential clinical toxicities will be discussed including integration of pharmacology data and considerations for the design and conduct of nonclinical toxicology studies. Clinical trial design for small molecule targeted anticancer therapeutics may need to evolve due to emergence of toxicities in clinical trials following many weeks to months of administration, particularly with regard to how the Phase2/3 clinical dose is selected. FDA is evaluating current practices by stakeholders for conducting adaptive design Phase 1 clinical trials aimed at identifying a recommended Phase 2 dose, integration of nonclinical data into dose evaluation past the first-in-human dose, integration of clinical pharmacology data and exposureresponse relationships and design and conduct of Phase 2 clinical dose-fnding trails. 39 MONDAY The National Cancer Institute’s Experimental Therapeutics Program performs the early clinical evaluation of molecularly targeted agents (MTAs) that they hold under IND for cancer treatment. NCI performs most of this work using two new networks of investigators with expertise in phase 0, 1, and 2 clinical trials for drug development. The NCI’s Experimental Therapeutics Clinical Trials Network is primarily focused on clinical trials that are used to determine the dose, schedule and safety of new drugs used for the first time in human subjects. These trials will typically focus on the pharmacokinetics, pharmacodynamics and response to treatment for the new agents. The development of MTAs also involves the identification and initial evaluation of biomarkers that can be used to select patients for enrollment or to be assigned to a specific treatment. The development of MTAs has brought with them dramatic successes and new challenges for drug development. Prior to 2000, development was focused on cytotoxic chemotherapies and dose determination based on safety and tolerability using a maximum tolerated dose that hit rapidly dividing cancer cell and relatively spared normal tissue. Development of MTAs is now focused on the determination of an optimal biological dose which may not cause dose limiting toxicity. Adverse events detected and described during the conduct of early clinical trials of small molecule targeted anticancer therapeutics are often not predicted by the nonclinical studies using the current animal models. MTAs are expected to cause both on target and off target toxicity and also cause chronic low grade or DLT in cycles of treatment beyond the first cycle of therapy. Using DLT only to determine the recommended phase 2 dose, has led to doses that are licensed for specific indications, but not tolerable for extended administration. The definition of new paradigms for pre-clinical evaluation IND directed pharm/tox are needed. Likewise, clinical paradigms for recommended phase 2 dose 2014 th 35 American College of Toxicology Annual Meeting 6:30 AM–7:30 AM ACT's Inaugural ToxTrot 2014 Portico West Patio (Free event, registration by October 30 required) 8:00 AM–9:00 AM Breakfast Reception Grand Cypress Ballroom (Open event) Tuesday Morning Sessions SYMPOSIUM 7 Breathe In, Breathe Out, It's Easy: What You Need to Know about Developing Inhaled Drugs CHAIR: Jeff Tepper, Tepper 9:00 AM–12:00 Noon Regency Hall Nonclinical Consulting, San Carlos, CA CO-CHAIR: Jim Blanchard,Aradigm, Hayward, CA Supported by educational donations provided by: Lovelace Respiratory Research Institute, Huntingdon Life Sciences, and Seventh Wave Laboratories It's no surprise that toxicologists feel unprepared for their first development program using an inhaled drug. Understanding of the area requires knowledge of lung anatomy, cell biology, respiratory physiology, particle physics and plumbing. Although Paracelsus informed us that the dose makes the poison and thus is of paramount importance, in the context of an inhaled drug, the "dose" is not easily defined. This symposium will attempt to demystify and provide background information enabling you to understand the issues and challenges associated with the assessment of respiratory safety pharmacology and designing an inhalation toxicology program. First, an overview examining ventilation, how to measure it, its influence on dose and the perturbations observed with inhalation exposure will be covered. The topic of dose will be further explored to better understand the assumptions and methods associated with determining drug deposition and clearance to arrive at lung dose. Next, the techniques used to deliver aerosols to animals in a physiologically appropriate manner, as well as the devices used to conserve test article, will be examined. With dose and inhalation exposure covered, species-specific histopathologic lesions, both common background and toxicologically significant lesions, will be discussed. Finally, insight into how regulators synthesize and evaluate these complex findings to assess clinical safety risks will be presented. S7–1 9:00 AM–9:35 AM Overview: It Sucks and Blows, Lung Function Jeff Tepper, Nonclinical Consulting, San Carlos, CA S7–2 9:35 AM–10:05 AM Dose is Dose, Right? Determining Pulmonary Dose in Clinical and Nonclinical Studies Philip Kuehl, Lovelace Respiratory Research Institute, Albuquerque, NM S7–3 10:05 AM–10:35 AM You Need How Much!!! Test Article Conservation During In Vivo Inhalation Studies TUESDAY Stuart Cracknell, Huntingdon Life Sciences, Somerset, NJ S7–4 10:35 AM–10:55 AM Break 10:55 AM–11:30 AM Tour of the Respiratory Tract with Stops at Sites of Toxicological Significance Kristen Nikula, Seventh Wave Laboratories, LLC, Chesterfield, MO S7–5 11:30 AM–12:00 Noon Regulatory Considerations in Nonclinical Safety Evaluations of Inhalation Drugs Luqi Pei, US Food and Drug Administration, Silver Spring, MD 40 th 35 American College of Toxicology Annual Meeting S7–1 For inhalation studies, the respiratory tract is generally the site most likely to show evidence of toxicity, but not all toxicity manifests as morphologically observable injury. Functional changes, which can occur with inhalation exposure, can range from acute life threatening to more subtle changes in ventilation and breathing pattern that can alter the dose and distribution of an inhaled drug. The rationale, interpretation and practical application of lung-function tests that are, or can be, used in laboratory animals, is the focus of the presentation. These tests provide analogues of clinical pulmonary function tests conducted on humans. Because measurements of respiratory rate and tidal volume are typically measured as part of the safety pharmacology assessment, attention is given to the theory behind how changes in lung volume and flow occur. Additionally, methods to estimate (allometry) and measure lung function by plethysmography, pneumotachography and telemetry in animals will be explained. Functional changes that may manifest with inhalation products, including changes in ventilation, lung irritation, bronchoconstriction, restrictive, obstructive and diffusional changes will be briefly reviewed. Finally, strategies for the assessment of lung function within the context of the ICH-S7A guideline as well as techniques to enhance the detection of functional changes where concern has been raised will be discussed. S7–2 S7–3 Inhaled drug delivery to conscious animal models has always used many times more test article than is actually delivered to the respiratory tract. Half of this effect is associated with the inconvenient reality that animals breathe out as well as in. Even this “loss” as well as everything else associated with the delivery of inhaled aerosols is subject to some measure of control. The magnitude of systematic losses is typically greatest for dry powder formulations but poor efficiency can also be a feature of liquid droplet deliveries. Losses and inefficiencies occur in both the initial aerosol generation process and in the aerosol delivery systems used to bring the atmosphere product to the breathing zone. Enhancing the efficiency of test article usage can enable the completion of critical proof of concept and IND-enabling inhalation investigations with significantly smaller quantities of an active moiety than would be necessary when employing standard laboratory techniques. This is especially important early in test article development when the quantities available are commonly both limited and produced at the highest unit cost due to lab scale manufacture. Based on recent experience conducting programs in which test article costs substantially exceed the cost of running the in vivo studies themselves, this presentation will concentrate on those techniques that have been found to be effective when test article conservation in inhalation studies has been a critical component of program survival. While focusing on methods of aerosol generation, exposure system design and aerosol delivery, factors related to study and program design as well as study logistics will also be considered. S7–4 The respiratory system, which comprises the largest mucosal surface of the body, can be divided into two portions based on anatomy and physiology: the conducting airways (nose, pharynx, larynx, and tracheobronchial airways) and the respiratory portion (respiratory bronchioles, alveolar ducts, and alveolar sacs). Injury due to inhaled toxicants occurs when innate defenses are overcome. The location and type of injury depends on the physiochemical characteristics of the toxic agent, site-specific tissue dosimetry, and the metabolic capabilities and sensitivity of the cellular components of the respiratory tissue. Respiratory system macroscopic and microscopic anatomy, particularly as it relates to toxicant-induced injury and tissue responses, will be presented. The inhalation of highly water-soluble chemicals or large particles typically cause injury in the upper respiratory tract whereas poorly soluble chemicals and small particles cause injury in the centriacinar region. Highly reactive gases and certain nanoparticles cause injury in both upper and lower respiratory sites and toxicants that are metabolically activated cause injury at the site of metabolism. Species-related differences in functional anatomy of the respiratory tract that affect the response to toxicant exposure among preclinical species used in safety studies and humans will be discussed. Lastly, common background findings in preclinical species as well as examples of toxicant-induced lesions will be illustrated. S7–5 Regulatory nonclinical safety evaluations play a unique role in the development of inhalation drug products. These evaluations present challenges to regulatory and research scientists because of special characteristics associated with inhalation products. The products generally require complex delivery devices and involve aerosol sciences and engineering. Device, dosage forms, formulations, and delivery mechanics are variable. The anatomy and physiology of the respiratory system in animals and humans have their own special feature, and responses to inhaled materials in animals and humans may be species-specific. The design and conduct of inhalation toxicity studies also requires special knowledge, skills, facility, and equipment. It is often difficult to accurately assess the local and systemic exposures of animals and humans to inhaled drugs. All of these characteristics may affect the evaluation and interpretation of findings of inhalation toxicity studies in animals. Regulatory scientists should consider these features when conducting the nonclinical safety evaluation of inhalation drug products. 41 TUESDAY Pulmonary dose has been and continues to be an area of research and debate. Pulmonary dose can be calculated based on relatively standard calculations, which include multiple inherent assumptions. While there is precedence for the use of these calculations, they require assumptions to be made within them. Current trends in inhalation drug delivery have increased the needs for more accurate empirical data in quantifying pulmonary dose. In principle, there are two methods used to empirically quantify pulmonary dose: deposition imaging and pharmacokinetic analysis. Both empirical methods have strengths and weaknesses. Pharmacokinetic analysis in a nonclinical study can include lung tissue and systemic plasma measurements while in a clinical setting only includes systemic plasma. Systemic plasma, albeit a routine and established measurement, is an indirect method to quantify pulmonary dose. This inherently increases the potential for variability and can decrease utility of plasma measurements. Deposition imaging, typically conducted by SPECT/CT, can be used in both clinical and nonclinical studies with minimal differences. Deposition imaging has the advantage that it is a direct measurement of the dose at the site of deposition. The disadvantage is that imaging modalities quantify a radiotracer, not the drug. Therefore, the homogeneity of the radiotracer and the drug must be validated in vitro in order to quantify pulmonary dose in vivo. Overall, determining pulmonary dose throughout clinical and nonclinical studies is a manageable task. Risk-based choices must be made as to the accuracy of dose needed and the methods most appropriate to quantify pulmonary dose. 2014 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 8 Cardiovascular Safety Evaluation— In Vitro to Humans 9:00 AM–12:00 Noon Grand Cypress Ballroom A CHAIR: Norman Kim, Biogen Idec, Cambridge, MA CO-CHAIR: Chris Mathes,ChanTest Corporation, Cleveland, OH Supported by an educational donation provided by: ChanTest Corporation A new scientific testing paradigm for drug-induced cardiac safety was proposed by a consortium sponsored by Cardiac Safety Research Consortium (CSRC), Health and Environmental Sciences Institute (HESI), and the US Food and Drug Administration (FDA). The proposal instituted Comprehensive In vitro Proarrhythmia Assays (CiPA) that provided a framework for a shift from the current testing guidelines as outlined in ICH S7B (nonclinical) and ICH E14 (clinical) to include novel assays in human cells and in silico models of cellular electrophysiological effects to evaluate proarrhythmic drugs. Additional concepts besides CiPA have also been proposed. This symposium will review the most current cardiovascular testing paradigm of potential therapeutics in consideration of the regulatory guidelines. The current ICH S7B discusses nonclinical testing strategy for cardiovascular safety. These include consideration of chemical/pharmacological class of the molecules, outcomes of in vitro assays (notably hERG inhibition), in vivo cardiovascular study, and other related toxicology studies where cardiovascular endpoints are evaluated. The ICH E14 provide clinical assessment of potential QTc prolongation. Nonclinical and available clinical safety information provide the basis of an integrated cardiovascular risk assessment of molecules. The focus of the ICH guideline has been centered around QT/QTc intervals and potential arrhythmia. Based on the available information, it is apparent that cardiovascular testing paradigm should include more than the hERG inhibition assay and evaluation of other ion channels should be considered along with the newer novel assays such as induced pluropotent-derived human stem cells for a more comprehensive cardiovascular evaluation. The screening of molecules during early discovery and development phases could be better utilized and optimized for cardiovascular evaluation. This session will introduce and review these in vitro assays and nonclinical in vivo evaluation. Current pharmaceutical and regulatory thinking process of cardiovascular evaluation will be discussed, including a proposal of modification/elimination of ICH guidelines S7B and E14. Experiences with CiPA in the cardiovascular evaluation will be further discussed. 9:00 AM–9:10 AM Introduction Norman Kim, Biogen Idec, Cambridge, MA, and Chris Mathes, ChanTest Corporation, Cleveland, OH S8–1 9:10 AM–9:40 AM Cardiovascular Safety Assessments During Drug Development: Current Approach and the Need for a New Paradigm Philip Sager, Stanford University, San Francisco, CA S8–2 9:40 AM–10:10 AM In Vitro Cardiovascular Safety—Discovery Screen to GLP Studies Bernard Fermini, Pfizer, Groton, CT TUESDAY S8–3 10:10 AM–10:30 AM Break 10:30 AM–11:00 AM Integrated Systems for Evaluation of Cardiovascular Electrophysiology Safety: Secondary In Vitro Assays and In Vivo Evaluations Gary Gintant, AbbVie Inc., North Chicago, IL S8–4 11:00 AM–11:30 AM Regulatory Thinking of the Current Guidelines on Cardiovascular Evaluation— Beyond QTc Norman Stockbridge, US Food and Drug Administration, Silver Spring, MD S8–5 11:30 AM–12:00 Noon Experiences with Elements of Comprehensive In Vitro Proarrhythmia Assay (CiPA) Arthur “Buzz” Brown, ChanTest Corporation, Cleveland, OH 42 th 35 American College of Toxicology Annual Meeting S8–1 The current approach, outlined in ICH S7B and E14, to cardiac arrhythmia risk is focused on preclinical and clinical assessments: 1) the effect of new chemical entities on the hERG encoded IKr current and QTc prolongation in animals and 2) measuring QTc prolongation at supratherapeutic exposures in humans, typically during Phase 2 of development. While this approach has largely eliminated the unanticipated presence of new torsadogenic drugs entering the market, it has become clear that there are important limitations to this methodology. Block of hERG alone is often insufficient in predicting delayed repolarization and hERG-related prolongation of repolarization does not necessarily translate into human torsade de pointes risk. Increases in the QTc interval are highly sensitive but not very specific for the prediction of ventricular proarrhythmia risk. The use of the hERG repolarizing ionic current assay (in early nonclinical development) and QTc interval prolongation (in later nonclinical and clinical development) as gatekeepers for the continued development of a new chemical entity has likely led to the inappropriate discontinuation of development programs for drugs with potentially high public health benefits. The talk will focus on these key issues and a potential new approach to drug-induced arrhythmia development. S8–2 During the past two decades, a number of noncardiovascular drugs have had their label revised or have been withdrawn from the market because of unexpected post-marketing reports of sudden cardiac death associated with a prolongation of the QT interval on the electrocardiogram and increased propensity to develop a ventricular tachyarrhythmia called Torsades de Pointes. Compound withdrawal not only indicates that a risk has been posed to patient safety, but also results in a significant loss of invested time, money and resources. In a majority of cases, QT prolongation results from inhibition of the delayed rectifier potassium current IKr or hERG. Consequently, assessment of cardiac ion-channel activity of new chemical entities is now an integral component of drug discovery programs. However, it has recently become apparent that the current ion channel paradigm (mostly based on hERG screening) may not assess the end point of primary concern (i.e. ventricular proarrhythmia), and a shift in paradigm has been proposed. In this presentation we will review a typical ion channel cardiovascular safety strategy and examine some of the methods and assays used from the exploratory to the GLP stages. We will put the data generated from these assays in context with translation to in vivo correlates, and take a critical look at some of the newer assays and methodologies that strive to address gaps present in the existing paradigm, such as the inability to adequately assess the proarrhythmic potential of new chemical entities. S8–3 “best practice” in vivo studies (e.g., telemetry studies) coupled with the use of so-called “secondary assays” (e.g. repolarization assays and studies with human stem-cell derived cardiomyocytes) to enhance or understanding of drug effects represents a reasonable approach for understanding cardiovascular liabilities early in drug discovery, guiding compound selection and subsequent clinical studies. This presentation will discuss considerations of how to best blend secondary and in vivo assays to inform on cardiovascular risk assessments. S8–4 The current ICH S7B and E14 guidelines have been the mainstay of the preclinical and clinical cardiovascular testing paradigm. Although ICH S7B and E14 provide the basis of the cardiovascular safety testing, this evaluation scheme has limitations. The measurement of QTc prolongation is highly sensitive but not specific for predicting ventricular proarrhythmia risk of a drug, and may inappropriately assign a drug with a Torsades de Pointes (TdP) liability. In addition to the inhibitors of rapidly activating delayed rectifier potassium current (IKr), there are other factors that may lead to QTc prolongation. This presentation will provide a regulatory perspective of cardiovascular assessment during drug development, including a brief history of molecules that have been removed from the market due to TdP risk, basis of how the current ICH guidelines was instituted, limitations of the current guidelines, regulatory views of various proposals of integrative proarrhythmia evaluations, and future direction of cardiovascular testing. Examples will be provided where applicable. S8–5 The regulatory paradigm for nonclinical cardiac risk assessment has shifted from delayed repolarization, hERG block and QT prolongation to involvement of major human cardiac ion channels and proarrhythmic effects on human cardiomyocytes. To test whether multiple ion channel effects (MICE) were superior to hERG alone, automated gigaseal patch clamp was used to measure the concentration-responses of hERG, hNav1.5 and hCav1.2 to torsadogenic and non-torsadogenic drugs and compared the predictivity of a variety of logistic regression (LR) models. The LR model for hERG was significantly inferior to the best MICE LR model. Many of these drugs were tested on induced pluripotent stem cell (iPSC)-derived cardiomyocytes using manual patch clamp to record action potential duration (APD) and a multielectrode array (MEA) to record 2D propagation. With the latter, spike amplitude, beat rate and field potential duration (FPD) were measured. Drugs that are positive for Torsade de Pointe (TdP+) drugs prolonged FPD and at higher concentrations produced early after depolarization (EAD) and arrhythmias. Negative TdP drugs had no effects. When an impedance device (xCELLigence) was used to measure cytoxicity and contractile changes, a good correlation was evident with anticipated drug effects. The results of this multi-faceted approach are being integrated to provide a hypothetical mechanism of action that can be applied to predicting cardiac risk in the clinic. 43 TUESDAY There is a need to establish robust approaches to ensure the selection of drug candidates with safe cardiovascular profiles through a combination of in vitro and in vivo mechanism-based and integrated studies. Understanding the strengths and limitations of 2014 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 9 Centrally Administered Compounds in Small and Large Species—Dose Routes, Study Considerations, and Data Interpretation CHAIR: Sven Korte, Covance 9:00 AM–12:00 Noon Palm Laboratories GmbH, Münster, Germany CO-CHAIR: Teresa L. Wright,Shire, Lexington, MA Supported by educational donations provided by: Covance Laboratories Inc., Tox Path Specialists, LLC, and Shire Many diseases affecting the central nervous system (CNS) are inadequately treated by traditional systemic delivery methods, partly because of the inability to bypass the blood-brain barrier. Delivery of large molecules, cells, and other novel therapies directly to the nervous system via intrathecal injection and/or implanted catheters overcomes this obstacle and administers the treatments close to the target region. Clinical experience using direct CNS administration of compounds for pain relief and chemotherapy has grown over the past decades, suggesting the same technologies can be applied to the treatment of degenerative and inherited disorders. Administration of drugs directly into the CSF may involve some risk, including reaction of the spinal cord or brain tissue adjacent to the device. The preclinical studies used to evaluate the safety of CNS administered compounds must differentiate between the effects of the delivery method, the therapy, and the combination. Reliable, wellcharacterized animal models for CNS administration of test articles have been developed which enable nonclinical development of these potential therapeutics. This session will discuss the technical challenges of preclinical intrathecal studies, design of studies and nonclinical programs, evaluation of results, and considerations for special endpoints in the studies. S9–1 9:00 AM–9:40 AM Development of CNS Administered Biologics: Overview, Challenges, and a Case History Brian R. Vuillemenot, BioMarin Pharmaceutical Inc., Novato, CA S9–2 9:40 AM–10:20 AM Methodology of Central Drug Delivery and Cerebrospinal Fluid Collection in the Sheep, Dog and Rat Model Eric Adams, Northern Biomedical Research, Inc., Spring Lake, MI S9–3 10:20 AM–10:40 AM Break 10:40 AM–11:20 AM Procedures for Intrathecal Drug Delivery and CSF Sampling in Nonhuman Primates Studies Sven Korte, Covance Laboratories GmbH, Münster, Germany S9–4 11:20 AM–12:00 Noon Morphologic Assessment of Studies Involving Direct Delivery to the Central Nervous System TUESDAY Mark T. Butt, Tox Path Specialists (TPS), LLC, Frederick, MD 44 th 35 American College of Toxicology Annual Meeting S9–1 This presentation will begin with an overview of theoretical considerations relevant to direct CNS administration, including blood brain barrier structure, CSF dynamics, routes of administration, delivery devices, and relevant disease indications. Design of nonclinical toxicology and pharmacology studies and IND/CTA enabling programs for CNS administered therapeutics will be discussed, with a focus on relevant regulatory guidelines. Finally, a nonclinical case history of a CNS-administered enzyme replacement therapy currently in clinical trials will be presented to illustrate some of these concepts. S9–2 This presentation will introduce surgical techniques and methodology surrounding drug delivery and CSF collection in the sheep, dog and rat animal model. Emphasis will be on stereotactic surgery techniques, the importance of performing pilot studies, various drug delivery systems as well as CSF collection methods. Intraparenchymal, intracerebroventricular and intrathecal surgery models will be discussed along with existing and novel CSF collection techniques. These techniques will include cisterna magna and lumbar puncture in anesthetized animals (sheep, dog, and rat) as well as serial and continuous CSF collection in conscious rats. 2014 S9–3 The effects of CSF (cerebrospinal fluid) infused compounds in large animal models will be reviewed. The presentation will focus on animal selection, surgical techniques, and study considerations in nonhuman primate studies undergoing continuous or slow bolus injection by the lumbar, cisterna magna or ventricular route. Commonly observed spontaneous and induced changes observed in the in-life phase of regulatory studies and under European housing conditions will also be presented. Furthermore, the assessment of juvenile toxicity in cynomolgus monkeys represents an emerging field, requiring the application of established techniques in 12-month old (or even younger) monkeys. The feasibility and precaution measurements when conducting this type of studies will be presented. S9–4 This presentation will introduce the challenges and optimal techniques used to assess the structural changes in the brain and spinal cord associated with the direct delivery of various test articles to the intrathecal space or directly to the brain parenchyma. The sectioning schemes, staining methodologies, embedding techniques, and special techniques for quantitative assessments will be discussed and illustrated. Sample gross and microscopic changes associated with the various delivery techniques will be shown, along with examples of lesions associated with various classes of test articles. TUESDAY 45 th 35 American College of Toxicology Annual Meeting 12:00 Noon–1:30 PM IJT Editorial Board Meeting 12:00 Noon–1:30 PM 2015 Program Planning Meeting 2014 Magnolia Regency Hall (All ACT members invited. Sign up at the registration desk. Box lunch provided.) Tuesday Afternoon Sessions SYMPOSIUM 10 Cytokines: The Good, the Bad, and the Deadly CHAIR: Thulasi Ramani, Huntingdon 2:00 PM–5:00 PM Grand Cypress Ballroom A Life Sciences, Somerset, NJ CO-CHAIR: Carol S. Auletta,Huntingdon Life Sciences, Somerset, NJ Supported by an educational donation provided by: Huntingdon Life Sciences Over the past 30 years, the world of pharmaceutical toxicology has seen an explosion in the area of cytokines. This symposium presents an overview of the many aspects of cytokine safety evaluation currently in progress and evolving strategies for evaluating these important entities. Cytokines play a broad role to help the immune system respond to diseases, and drugs which modulate their effect have led to some amazing therapies. Cytokines may be “good” when stimulating the immune system to fight a foreign pathogen or attack tumors. Other “good” cytokine effects include reduction of an immune response, for example Interferon beta reduction of neuron inflammation in patients with multiple sclerosis. They may be “bad” when their expression causes inflammatory diseases, such as the role of TNF-alpha in rheumatoid arthritis or asthma and Crohn’s disease. Therapeutic modulation of cytokine expression can help the “good” cytokines to generate or quench the immune system, and block the “bad” cytokines to prevent damaging inflammatory events. However, care must be exercised, as some antibody therapeutics can cause “ugly” cytokine release, which can be deadly. Well-designed toxicology studies should incorporate careful assessment of cytokine modulation that will allow effective therapies to treat unmet needs. This symposium discusses lessons learned in cytokine toxicology using case studies and suggests future directions. 2:00 PM–2:10 PM Cytokine Explosion: Rocking the World of Toxicology Thulasi Ramani, Huntingdon Life Sciences, Somerset, NJ S10-1 2:10 PM–2:40 PM Therapeutic Cytokine-Blocking Antibodies Daniel Weinstock, Janssen Research and Development, Spring House, PA S10-2 2:40 PM–3:10 PM Safety Evaluation of Cytokine Therapeutics: Translation of Preclinical Data to the Clinic Barbara Mounho-Zamora, ToxStrategies Inc, Bend, OR S10-3 3:10 PM–3:40 PM Targeting Cytokine Pathways to Treat Disease TUESDAY Patricia Ryan, MedImmune, Gaithersburg, MD S10-4 3:40 PM–4:00 PM Break 4:00 PM–4:30 PM Beyond TeGenero—Development of Targeted Therapeutics without Deadly Consequences Theodora Salcedo, Bristol Myers Squibb Company, New Brunswick, NJ S10-5 4:30 PM–5:00 PM Cytokines as Biomarkers for Immunotoxicity Gregory Bannish, Huntingdon Life Sciences, Somerset, NJ 46 th 35 American College of Toxicology Annual Meeting S10–1 Antibodies and their derivatives comprise a growing segment of approved products for treatment of a wide variety of human diseases. Soluble cytokines and their receptors make excellent targets for therapeutic intervention based on extensive documentation of their roles in specific diseases. Antibodies against tumor necrosis factor (TNF), human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR) are “blockbuster drugs.” Additional validated cytokine targets for treatment of human diseases include interleukin-1 (IL-1), IL-2 receptor (IL-2R), IL-6R, IL-12, IL-23 and receptor activator of nuclear factor-κB ligand (RANKL). Development strategies for large molecule therapeutic proteins with immunomodulatory effects require understanding of the effector functions of the therapeutic molecule as well as the role of the targeted cytokine and/or it’s receptor(s) in the normal and diseased states. Preclinical and clinical development strategies strive to characterize safety and efficacy. ICH S6 (Preclinical Safety Evaluation of Biotechnology-derived Pharmaceuticals) and ICH S8 (Immunotoxicity Studies for Human Pharmaceuticals) provide guidance for development of immunodulatory biologics. Unfortunately, treatment of patients with immunomodulatory agents sometimes may result in unwanted adverse consequences. Thorough knowledge of the mechanism of action of the therapeutic molecule and the biology of the cytokine target are required for efficient development strategies and ultimate therapeutic use. S10–3 Biologics that impact cytokine pathways are promising therapies for a variety of diseases including cancer, autoimmunity and inflammation. This presentation will describe several case examples of biologics that target cytokine pathways. In each case, the special considerations required in designing and interpreting nonclinical safety studies for biologics, which either induce cytokines or block a key cytokine pathway as part of their mechanism of action will be described. In each of these case examples, consideration of the expected pharmacology was important to appropriately interpret nonclinical safety results given that the majority of the toxicities for these types of biologics arise from on-target excessive pharmacodynamics. S10–4 TGN1412 is a monoclonal antibody that binds to CD28 and has agonistic activity on cytokine release. Although it was studied preclinically, it was administered in the clinic (in 2006) with disastrous consequences. It made the term “cytokine storm” a household word in toxicology circles. This presentation will provide an update on advances in the science associated with “cytokine storm,” and describe strategies used successfully to develop targeted therapeutics that can be safely administered to humans. S10–5 Cytokine evaluation is often included in preclinical studies for the assessment of immunotoxicity. The increasing use of these evaluations reflects the increased understanding of the role of cytokines in the immune response, the modulation of cytokines following inflammatory events, and the increased ability to measure cytokine levels through new technologies. However, significant challenges exist which can include temporal expression, low systemic expression, complexity, redundancy, species, strain, and inter-animal variability, and the limited knowledge and reagent availability for immunological evaluations in common toxicity species. This presentation will discuss significant advances in primate cytokine evaluation, including intracellular evaluation of cytokine expression to delineate lymphoid subpopulations, evaluation of cytokine levels in sera at sub-pg/mL concentrations, evaluation of cytokine mRNA expression, evaluation in skin biopsies, and a validated 24-plex cytokine panel. Many of these advances will allow for establishment of biomarkers in preclinical studies that will improve the ability to monitor for immunopharmacology and toxicity in clinical studies. 47 TUESDAY S10–2 Cytokines are a unique class of regulatory proteins that play a critical role in maintaining and regulating immunologic systems. Excessive production of certain cytokines, however, can lead to pathological consequences. Advancement in cytokine research has lead to the development of cytokine therapeutics (i.e., cytokine antagonists) for the treatment of various diseases. Modulation of the immune system by cytokine therapeutics, however, can result in adverse clinical consequences, especially with chronic administration. The preclinical safety evaluation of cytokine therapeutics is a critical component in understanding the potential adverse effects associated with cytokine therapeutics. Identifying relevant assays and incorporating safety parameters in toxicology studies for cytokine therapies can be challenging, particularly in understanding the relevance of the preclinical data to the patient population. Additionally, it is critical to understand the potential toxicities and impact on the immune system associated with chronic administration of the cytokine therapeutic, which may not be possible to demonstrate in animal studies. This presentation will review the types of assays and parameters used in the preclinical safety evaluation of cytokine therapies. The challenges in understanding the relevance of the data to the patient population and how these challenges are managed with approved cytokine therapeutics will also be discussed. 2014 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 11 Differentiating Adverse from Adaptive Changes in Toxicology CHAIR: Anthony L. Kiorpes,River 2:00 PM–5:00 PM Palm Bluff Associates, LLC, Bloomington, MN CO-CHAIR: Arun R. Pandiri,Experimental Pathology Laboratories, Inc., RTP, NC Supported by educational donations provided by: Experimental Pathology Laboratories, Inc., and BASi An important question in the practice of toxicology is, “what is an adverse finding”? Central to what every toxicologist does is the interpretation of a large experimental data set to arrive at a no-observed-adverse-effect level (NOAEL). In many cases, the difference between an adverse (pathological) finding from one that is adaptive (physiological) is not obvious. Specifically, this symposium will explore what are adverse findings and what differentiates them from normal physiological or adaptive changes from whole-animal, organ, molecular, and regulatory perspectives. The topic is timely as recent meetings, working groups, and papers have attempted to address this issue. The intended audience is the general toxicologist regardless of industry segment. S11-1 2:00 PM–2:40 PM A Whole-Animal Overview of Adaptive versus Adverse Change Peter C. Mann, Experimental Pathology Laboratories, Inc., Seattle, WA S11-2 2:40 PM–3:20 PM Identifying Adverse Organ Responses: The Liver as Paradigm Robert R. Maronpot, Maronpot Consulting, LLC, Raleigh, NC S11-3 3:20 PM–3:40 PM Break 3:40 PM–4:20 PM Using Toxicogenomics in Assessing Adaptive versus Adverse Effects Russell Thomas, US Environmental Protection Agency, National Center for Computational Toxicology, Research Triangle Park, NC S11-4 4:20 PM–5:00 PM Differentiating Adaptive and Adverse Changes: A Regulatory Perspective TUESDAY Timothy J. McGovern, US Food and Drug Administration, Silver Spring, MD 48 th 35 American College of Toxicology Annual Meeting S11–1 The first talk will introduce the problem of differentiating adaptive, physiological changes in test systems from nonadaptive, adverse, or pathological changes that are toxicologically meaningful. The perspective of this talk will be at the level of the whole organism. Practical examples will be given as part of the talk. S11–2 This talk will focus on acute, intermittent, delayed, and chronic adverse responses at the organ system level using the liver as a model organ. The talk will include practical biomarkers and will demonstrate the integrating of meaningful endpoints in supporting an overall conclusion of adaptive, physiological change vs nonadaptive, pathological or adverse change. The talk will discuss predictive value (clinical relevance) of meaningful endpoints and the difference between adverse changes or findings that are reversible versus those changes or findings that are not reversible. Case studies will be used to make key points practical and meaningful. 2014 S11–3 This talk will focus on using toxicogenomic technologies to assess adaptive versus adverse effects in toxicological research. Transcriptomic alterations are usually noted before the manifestation of histological changes and at usually lower doses. The challenging aspect of using toxicogenomics for assessing adaptive versus adverse effect is that not all transcriptomic alterations result in an adverse effect. This presentation will discuss the concepts of NOTEL (no observable transcriptional effect level) and TBMDL (transcriptional benchmark dose level), and its relationship to traditional toxicology end points. Examples will be discussed to demonstrate the value of using toxicogenomics in assessing adaptive versus adverse effects. S11–4 Identification of a no-observed-adverse-effect-level (NOAEL) from a toxicity study is a key component in identifying acceptable human exposure to a particular test article. One aspect of this exercise is distinguishing between adaptive and adverse effects. This talk will focus on the question of adverse versus adaptive changes from a regulatory point of view with an emphasis on pharmaceutical drug development. The primary points of discussion will include the issues regulators need to consider in evaluating toxicity data that are generated in support of clinical development programs and the information needed to support a “nonadverse” interpretation of clinical or pathological changes. Case examples will be discussed to illustrate the key issues in distinguishing adaptive versus adverse responses from a regulatory perspective. TUESDAY 49 th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 12 In Silico Methods for Mutagenicity Prediction vis à vis ICH M7 Guidance CO-CHAIR: Joel Bercu,Amgen, CO-CHAIR: Vijay K. Gombar,Eli 2:00 PM–5:00 PM Regency Hall Inc., Thousand Oaks, CA Lilly and Co., Indianapolis, IN Supported by an educational donation provided by: the American College of Toxicology The manufacture of the active pharmaceutical ingredient (API) of a drug involves a number of starting materials, reagents, and intermediates. Many of these chemicals could be potentially genotoxic and may end up as impurities in an API. It is the responsibility of the sponsor pharmaceutical company (Pharma) to identify, control, and assess risk posed by these potential genotoxic impurities (GTIs). Given that a GTI meets regulation on its limits, the ICH M7 draft guidance (Guidance) permits use of in silico approaches in contrast to experimental assessment of mutagenicity. Now that more than two years have passed since the Guidance was issued, the present symposium seems timely and important to share and learn about (1) what are regulatory expectations around use of in silico methods, (2) which in silico methods Pharma companies are using, and why, for compliance, (3) what developments have taken place by developers of in silico tools and methods, and most importantly (4) case studies on risk assessment by Pharmas highlighting their experience in the use of their chosen in silico methods. There could not be a more suitable forum than the ACT Annual Meeting, which brings together important stakeholders, for discussion and shared learning around these salient topics. S12-1 2:00 PM–2:25 PM Risk Characterization of Mutagenic Impurities Joel Bercu, Amgen, Thousand Oaks, CA S12-2 2:25 PM–2:50 PM Regulatory Implementation of ICH M7 Mark W. Powley, US Food and Drug Administration, Silver Spring, MD S12-3 2:50 PM–3:15 PM Living with ICH M7: The Practical Application of Two In Silico Systems in Screening for GTIs Nigel Greene, Pfizer, Inc, Groton, CT S12-4 3:15 PM–3:45 PM ICH M7: Risk Assessment with In-House and Off-the-Shelf In Silico Mutagenicity Predictors Robert A. Jolly, Lilly Corporate Center, Indianapolis, IN S12-5 3:45 PM–4:05 PM Break 4:05 PM–4:35 PM Using In Silico Tools for the Assessment of Genotoxic Impurities Chris Barber, Lhasa Limited, Holbeck, Leeds, England, United Kingdom S12-6 4:35 PM–5:00 PM Physico-Chemical Modulation of DEREK Structural Alerts TUESDAY Vijay K. Gombar, Eli Lilly and Co., Indianapolis, IN 5:00 PM–6:30 PM ACT Members' Meeting (All ACT members invited.) See page 10 for more information. 50 Regency Hall th 35 American College of Toxicology Annual Meeting S12–1 Pharmaceutical companies have processes to ensure potential exposure to impurities would not be of toxicological concern. Developments in science and regulatory guidance have allowed companies to characterize the risk of DNA reactive (mutagenic) impurities. These impurities are of particular concern since they are assumed to have no threshold. In silico assessments are an important part of the risk characterization process: hazard assessment. Computational assessments facilitate screening of actual and potential impurities and identify compounds requiring additional investigation. If an impurity is determined to be mutagenic, then it must be controlled to an acceptable dose following patient exposure. The threshold of toxicological concern (TTC) is considered a dose, which is of negligible excess cancer risk to patients assuming a mutagenic impurity is also a potent carcinogen. Chemistry uses the TTC to demonstrate process removal of an impurity to a negligible level. In conclusion, in silico technologies are a part of the risk characterization process for mutagenic impurities as it triggers further follow-up if an impurity is predicted to have mutagenic potential. S12–2 The draft ICH M7 guideline provides recommendations for assessing and controlling mutagenic impurities in drugs. For many impurities lacking empirical data, mutagenic potential can be established using (Q)SAR. Per the draft guideline, a (Q)SAR analysis should include prediction of bacterial mutagenicity (i.e., Ames assay) using 2 complementary methodologies. Expert knowledge may also be used to add perspective to the (Q)SAR prediction and support the overall decision-making process. The presentation will focus on regulatory expectations of impurity evaluations. Specific points of emphasis will include components of an appropriate (Q)SAR analysis, the utility and role of expert knowledge, as well as recommendations for reporting results. The current FDA Office of New Drugs review process and regulatory decision making will also be covered. pharmaceutical intermediates we have examined the relative predictive performances of some popular commercial systems that are in common use across the pharmaceutical industry. We have explored how these systems can be combined under the draft ICH M7 guidance to enhance the detection of DNA reactive molecules. S12–4 Recent ICH M7 draft guidance states that at least two in silico models (e.g., rules-based method and a QSAR approach), with complementary approaches, be used with expert knowledge for assessing potential mutagenicity of chemicals constituting the synthetic routes of pharmaceutical APIs. We have conducted detailed evaluation of commercial off-theshelf (COTS) packages and compared them with in-house models. We also investigated whether we can improve the accuracy of in silico mutagenicity predictions by adding proprietary data for training models. An optimization benefit analysis was done to identify the best mutagenicity predictors for risk assessments. S12–5 The nascent ICH M7 guidelines allow for the submission of in silico predictions of mutagenicity from 2 orthogonal systems following expert review. In order to support this review, such tools need to provide transparent and scientifically sound predictions with sufficient supporting information to allow the expert to understand why a prediction is made, how confident the user should be, and what evidence supports that conclusion. This talk will focus on the questions an expert should ask of the models and how such information can be used to come to an overall conclusion. It will be illustrated using two predictive tools—Derek Nexus, an expert system and Sarah Nexus, a statistical approach to the prediction of mutagenicity. S12–6 The focus of this investigation is the structural alerts (SAs) of mutagenicity underlying the in silico expert system, Derek Nexus (DN). The presence of a mutagenicity SA indicates mutagenic potential of a test compound and many DN SAs have been reported to be fairly accurate. We parsed clean, uniform data of Ames assay results for 8541 diverse compounds through DN to examine the hit rate and accuracy of DN SAs. We observed that many SAs had higher prevalence in nonmutagenic compounds. We analyzed each of the 23 DN SAs that hit on more than 10 nonmutagenic compounds by Recursive Partitioning (RP) to identify physico-chemical modulators of SAs that lead to higher accuracy in predicting mutagens. The RP trees for each of these 23 DN SAs will be presented and discussed for possible modification of rules. 51 TUESDAY S12–3 The current Step 2 document outlining the ICH M7 guidelines for the assessment and control of DNA reactive impurities in pharmaceutical products includes the use of in silico prediction systems as part of the hazard identification and risk assessment strategy. This is the first internationally agreed guidance document to include the use of computational approaches. The current proposal requires the use of two complimentary approaches, an expert rule-based method and a statistical algorithm. In addition, the draft guidance calls for the output from these computer-based assessments to be reviewed using expert knowledge to provide additional support or resolve conflicting predictions. Using a data set of 800 chemicals and 2014 You’re Invited to a Members-Only Wine Tasting Wine tasting Full page ad with 2015 ACT President Mary Ellen Cosenza Wednesday, November 12 • 5:00 PM–6:30 PM Grand View Terrace Hyatt Regency Grand Cypress, Orlando, Florida Come meet the incoming 2015 ACT President Mary Ellen Cosenza and share your thoughts on the ACT Annual Meeting over a glass of wine and light fare. This is a free event for all ACT members and advance registration is required. www.actox.org 52 th 35 American College of Toxicology Annual Meeting 2014 7:00 AM–7:55 AM See page 137 for more information. Wednesday Morning Sessions PLENARY LECTURE 2 Food and Feed Safety of Genetically Modified Organisms: The Hype and the Facts 8:00 AM–8:55 AM Grand Ballroom D ALISON VAN EENENNAAM COOPERATIVE EXTENSION SPECIALIST, ANIMAL GENOMICS AND BIOTECHNOLOGY, DEPARTMENT OF ANIMAL SCIENCE, UNIVERSITY OF CALIFORNIA, DAVIS Dr. Alison Van Eenennaam is a Cooperative Extension Specialist in the field of Animal Genomics and Biotechnology in the Department of Animal Science at University of California, Davis. She received a Bachelor of Agricultural Science from the University of Melbourne in Australia, and both an MS in Animal Science, and a PhD in Genetics from UC Davis. The mission of her extension program is “to provide research and education on the use of animal genomics and biotechnology in livestock production systems.” Dr. Van Eenennaam works particularly with the beef cattle industry and has developed a variety of extension programming for producers on topics ranging from marker-assisted and whole-genome enabled selection. Her outreach program focuses on the development of sciencebased educational materials outlining the uses of animal genomics and biotechnologies in livestock production systems, including the controversial biotechnologies of genetic engineering (GE) and cloning. She has given over 250 invited presentations in 17 states and 7 countries, and has served on several national committees including the USDA National Advisory Committee on Biotechnology and 21st Century Agriculture, (2005– 2009), and as a temporary voting member of the 2012 FDA Veterinary Medicine Advisory Committee meeting on the AquAdvantage salmon, the first GE to be evaluated for entry into the food supply. Dr. Van Eenennaam received the “National Award for Excellence in Extension” from the American Association of Public and Land-Grant Universities in 2010. 53 WEDNESDAY EXHIBITOR-HOSTED PROGRAMS WEDNESDAY th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 13 Novel Therapeutics in Oncology: Recent Advances and Considerations for Nonclinical Safety Evaluation CHAIR: Hadi Falahatpisheh, Pfizer, 9:00 AM–12:00 Noon Palm Pearl River, NY CO-CHAIR: Paul Moore,MacroGenics, Rockville, MD Supported by an educational donation provided by: the American College of Toxicology In the early 1980s, monoclonal antibodies were considered to have the potential to revolutionize cancer therapy through selective and specific targeting of tumor-associated antigen-positive cells. However, this concept has met with limited clinical success in oncology at this stage. These limitations led to the development of novel therapeutics such as antibody-drug conjugates, bispecific mAbs and cancer vaccines. ADCs combine the specificity of a mAb for a tumor-associated antigen with the potency of a novel generation cytotoxic agent and ensure selective tumor cell killing while reducing toxicity. Bispecific antibodies are designed to bind simultaneously to 2 different epitopes, such as a tumor antigen and a receptor on an immune effector cell with the objective to redirect effector cells to cancer cell killing. Therapeutic cancer vaccines take advantage of the individual's immune system to target tumor cells. This symposium will review the recent advancements for these emerging modalities and will also highlight specific considerations and challenges associated with the nonclinical safety evaluation of these molecules. We believe this symposium will be of value to a broad range of participants, including academic and industry researchers working in oncology, as well as toxicologists or regulatory scientists focusing on development of anticancer drugs. S13-1 9:00 AM–9:40 AM Antibody Drug Conjugates: Recent Developments and Future Perspectives Laurie Tatalick, Seattle Genetics, Bothell, WA S13-2 9:40 AM–10:20 AM Advances in Bispecific Antibodies Facilitating Novel Therapeutic Opportunities Paul Moore, MacroGenics, Rockville, MD S13-3 10:20 AM–10:40 AM Break 10:40 AM–11:20 AM Nonclinical Safety Evaluation of Novel Ab-based Therapeutics for Oncology Magali Guffroy, Pfizer, Pearl River, NY S13-4 11:20 AM–12:00 Noon Using the Power of the Immune System to Target Cancer David W. Clarke, Pfizer, Pearl River, NY 54 th 35 American College of Toxicology Annual Meeting Years of research and continuous optimization of the different parts of ADCs have led to a robust ADC pipeline (over 30 ADCs currently in clinical development) and the recent FDA approvals of Kadcyla (Genentech/ ImmunoGen) and ADCETRIS (Seattle Genetics). In this presentation, the recent breakthroughs in research and development of ADC modality will be discussed. S13–2 Monoclonal antibodies have provided breakthrough medicines for various diseases spanning cancer, autoimmunity and infectious disease. Their ability to selectively bind and interfere with a single target provides benefits over conventional therapy in terms of selectivity and specificity. Bispecific antibody technology provides an opportunity to extend this selectivity to dual targets, heralding opportunity to intervene in disease either through co-engagement of multiple pathways or cell types. Dual Affinity ReTargeting (DART) molecules are one such technology, which have been successfully designed to overcome limitations of prior bispecific antibodies in terms of manufacturability, stability and pharmacological properties. In this presentation, the preclinical development of bispecific DART molecules for cancer and autoimmunity will be presented showcasing their ability to dual target independent pathways or cell types simultaneously for therapeutic benefit. S13–3 The increasing molecule complexity of novel biotherapeutic modalities, such as antibody-drug conjugates and bispecific T-cell engagers, and specific considerations related to the targeted cancer patient population lead to the design of tailored nonclinical safety strategies that incorporate the most current international regulatory guidelines and rely on sound scientific principles. The presentation will first review and discuss major aspects of nonclinical development including selection of appropriate nonclinical species, design/ duration of general toxicity studies and new developments in immunosafety testing, along with reproductive toxicology and carcinogenicity assessments. We will also emphasize the need for efficient clinical development and the greater risk tolerance in patients when addressing life-threatening conditions. The presentation will then highlight major safety issues with these new therapeutic modalities and the predictivity/limitations of the nonclinical studies. S13–4 For many years the prospect of using the immune system to target cancer cells has been a goal. To date the approach has primarily been through passive immunization and the administration of mAb. There have been a number of attempts looking at active immunization, but to date this approach has met with limited success. Generating anticancer immunity is a multistep challenge which needs to generate a significantly higher immune response to be able to overcome tolerance to a self antigen, maintain the immune response, and modulate the immune suppression present in the tumor microenvironment. Many different approaches including autologous tumor cell vaccines, allogenic tumor cell vaccines, dendritic cell vaccines, protein- and peptide-based vaccines, and genetic vaccines have been evaluated in nonclinical and clinical studies and will be reviewed. Each of these approaches will have unique nonclinical studies, which pose a challenge to understanding the nonclinical safety of these cancer immunotherapy regimens. 55 WEDNESDAY S13–1 Antibody-drug conjugates (ADCs) represent an innovative and promising strategy for selective cancer treatment that combines the specificity of a monoclonal antibody (mAb) for a tumor-associated antigen with the potency of novel cytotoxic agents. Through targeted delivery of cytotoxins to cancer cells, ADCs are expected to be associated with increased efficacy, reduced systemic toxicity, and therefore improved therapeutic index. 2014 WEDNESDAY th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 14 Applied Nanotoxicology 9:00 AM–12:00 Noon Grand Cypress Ballroom A CHAIR: Dave Hobson,LoneStar CO-CHAIR: Robin C. Guy,Robin PharmTox, LLC, Bergheim, TX Guy Consulting, LLC, Lake Forest, IL Supported by an educational donation provided by: the American College of Toxicology Nanomaterials, including nanoparticles and nanoobjects, are being incorporated into everyday products at an increasing rate. These products include consumer products of interest to toxicologists such as pharmaceuticals, cosmetics, food, food packaging, household products, etc. The manufacturing of products containing or utilizing nanomaterials in their composition may also present potential toxicologic concerns in the workplace. The molecular complexity and composition of these nanomaterials is ever increasing and the means and methods being applied to characterize and perform useful toxicologic assessments are rapidly advancing. This session will include presentations by experienced toxicologists in the nanotoxicology community that are focused on the applied aspect of the discipline toward supporting state of the art toxicologic assessments for food products and packaging, pharmaceuticals and medical devices, inhaled nanoparticle and gastrointestinal exposures and addressing occupational safety and health issues and concerns. S14-1 9:00 AM–9:35 AM Introduction to Applied Nanomaterial Toxicology and Applied Nanotoxicology for Pharmaceuticals and Medical Devices Dave Hobson, LoneStar PharmTox, LLC, Bergheim, TX S14-2 9:35 AM–10:05 AM Assessing the Biological Fate of Ingested Nanomaterials Steve Roberts, University of Florida, Gainesville, FL S14-3 10:05 AM–10:35 AM Nanoparticles as an Emerging Environmental and Occupational Hazard— Toxicology Prospective Anna A. Shvedova, CDC-National Institute for Occupational Safety and Health, Morgantown, WV S14-4 10:35 AM–10:55 AM Break 10:55 AM–11:30 AM Consumer Product Example: Strategies for Setting Occupational Exposure Limits for Engineered Titanium Dioxide Nanomaterials David Warheit, DuPont Haskell Laboratories, Newark, DE S14-5 11:30 AM–12:00 Noon Applied Considerations for Safety Assessment of Food Products and Food Packaging Containing Nanomaterials Robin C. Guy, Robin Guy Consulting, LLC, Lake Forest, IL 56 th 35 American College of Toxicology Annual Meeting S14–2 The biological activity of ingested nanomaterials depends to a large extent on reactions and interactions that occur both within the gastrointestinal tract and the body. This presentation provides an overview of experimental approaches for examining nano particle behavior in the gut, as well as uptake into and distribution within the body. S14–3 Advancements in nanotechnology field with broad applications and manufacturing of nanomaterials raise the issue of their potential adverse health effects particularly in occupational and environmental settings. Carbonaceous nanoparticles (CNP) have a wide scope of uses, ranging from cosmetics and toiletry products, biomedicine, polymer chemistry, composites employed in vehicles and sports equipment, to integrated circuits for electronic components. The lung is the major portal of unintended CNP entry into the human body potentially leading to pulmonary damage, inflammation, oxidative stress, fibrosis, and granuloma formation. By employing proteomics and lipidomics analyses, in vivo ESR spin-trapping technology, redox assessments of antioxidant balance, and quantitative morphometry (including collagen) in wild-type and genetically manipulated mice, we were able to reveal the major pathways through which CNP — in doses relevant to potential occupational exposures—exert their toxic effects in the lung and distant organs of exposed animals. The talk will address important issues of comparative respiratory outcomes of CNP and asbestos, particularly with regards to pulmonary injury and potential carcinogenicity. Finally, the mechanisms of toxicity will be discussed in the context of current regulations in environmental and occupational settings. Acknowledgements: supported by National Institutes of Health 008282, NORA 92700Y, 7th Framework EU: FP7-NMP-2007. S14–4 Lessons learned from practical development of methods to evaluate toxicological effects from inhaled nanomaterial exposures using a consumer product example. The presentation will describe a bridging approach to estimate OELs for three types of nano-TiO2 particulates based on comparative intratracheal instillation toxicity studies in rats. Toxicity profiles for the nanoparticulates were compared with that for control materials (pigment-grade TiO2 particles) for which an extensive database of long-term, animal inhalation data and epidemiological data exists. S14–5 More and more, companies are considering using nano-sized materials in their food products and food packaging. These new food ingredients exhibit benefits for the consumer; and some of these benefits will be discussed. However, they also bring about chemical, functional, and possibly toxicological differences as compared to their micro-sized counterparts. New applications are being developed that may take food contact items to the next level of food safety. As with any new ingredient, safety assessments need to be conducted on each of these new ingredients. This discussion will focus on practical aspects of safety assessment for the development of foods and food contact items, which contain nano-sized materials. 57 WEDNESDAY S14–1 A brief overview and background of the session topics with examples of applied nanotoxicology for pharmaceutical and medical device products will be provided. State of the art toxicology approaches being used with success for these nanomaterials applications will be presented as well as examples of past problems and current challenges with conducting safety evaluations using various models and gaining regulatory approval. 2014 WEDNESDAY th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 15 Stem Cells Research 9:00 AM–12:00 Noon Grand Cypress Ballroom B CHAIR: Kathleen A. Funk,Experimental Pathology Laboratories, Inc., Sterling, VA CO-CHAIR: Maralee McVean,Pre-Clinical Research Services, Inc., Fort Collins, CO Supported by educational donations provided by: Experimental Pathology Laboratories, Inc. and Pre-Clinical Research Services, Inc. Stem cells have great potential in basic research and are being slowly integrated into toxicological research. This symposium will provide an overview of the state of the field, stem cell models, describe allogeneic stem cell treatments and issues of immunogenicity associated with protein therapeutics, and then concentrate on stems cell uses in regenerative medicine focusing on lung and testing strategies on engineered tissues from a pathologist’s perspective. S15-1 9:00 AM–9:35 AM Where Is the Field of Regenerative Medicine Going? Alan Trounson, Monash University, Victoria, Australia S15-2 9:35 AM–10:20 AM Applications of Stem Cell Technologies in Drug Discovery Kyle Kolaja, Cellular Dynamics, Madison, WI S15-3 10:20 AM–10:40 AM Break 10:40 AM–11:20 AM Pathology Techniques to Aid the Development and Understanding of Advanced Therapy Medicinal Products (ATMPs) Klaus Weber, AnaPath GmbH, Oberbuchsiten, Switzerland S15-4 11:20 AM–12:00 Noon Advancing Engineered Lung Tissues to the Clinic: Considerations for Animal Models and Toxicology Studies Thomas Petersen, United Therapeutics Corporation, Research Triangle Park, NC 58 th 35 American College of Toxicology Annual Meeting S15–2 This talk will serve as an introduction to the applications of stem cell derived tissues toxicology applications in preclinical discovery. The talk will highlight activities related to stem cell-derived cardiomyocytes, their use to predict arrhythmia and regulatory evaluation as well as the latest efforts related to predicting teratogenicity, hepatotoxicity, and neuronal toxicity. In addition, the prospect of genetic diversity of iPScell derived tissues in applied research will be discussed. S15–3 Histopathologic analysis can help choose relevant animal models, consider species specificity on a molecular, cellular and tissue level, illuminate immunogenicity issues, detect possible adverse effects, and the safety and suitability of all structural components. S15–4 Engineered lung tissues offer a potential solution to the shortage of available organs for lung transplantation. This presentation will cover on the following topics: 1) A scientific update on the latest advancements in the generation of functional engineered lungs; 2) A discussion of ex vivo assessments of pulmonary function; 3) A discussion of the challenges of transplantation studies using engineered lungs; and 4) the impact on selection of animal models and the design of toxicology studies. 59 WEDNESDAY S15–1 Overview of stem cells and their characteristics and the ability of stem cell technologies to represent proper organ physiology and predict response to disease states and therapies. 2014 WEDNESDAY th 35 American College of Toxicology Annual Meeting 2014 SYMPOSIUM 16 Safety Assessment Updates for Preventive Vaccines and Vaccine Adjuvants CHAIR: David W. Clarke,Pfizer, CO-CHAIR: Jayanthi Wolf, Merck 9:00 AM–12:00 Noon Grand Cypress Ballroom C Pearl River, NY & Co, West Point, PA Supported by an educational donation provided by: the American College of Toxicology With the development of more refined vaccine antigens there has been a need to help boost the immune response through the addition of adjuvants into the formulation. The objective of this session is to review the rationale for the inclusion of adjuvants in a vaccine and some of the unique challenges that the inclusion of an adjuvant brings to the nonclinical evaluation of the final vaccine product. The session will include both the industry perspective on the nonclinical development of vaccines, but also the regulatory framework under which the work is conducted. S16-1 9:00 AM–9:25 AM Rationale for Use of Adjuvants in Vaccines and Overview of Adjuvant Classes and Pathways Jayanthi Wolf, Merck, West Point, PA S16-2 9:25 AM–9:50 AM Regulatory Considerations in the Safety Assessment of Vaccine Adjuvants and Adjuvanted Vaccines Carmen Custodio, US Food and Drug Administration, Silver Spring, MD S16-3 9:50 AM–10:20 AM Industry Approaches to the Evaluation of Adjuvants and Immunostimulators Ozzie Berger, GlaxoSmithKline Vaccine, King of Prussia, PA S16-4 10:20 AM–10:45 AM Safety Considerations for Adjuvants including Update from HESI Meeting on Adjuvants and Autoimmunity Sarah Gould, Sanofi-Pasteur, Marcy L’Etoile, France on behalf of The International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute (HESI) S16-5 10:45 AM–11:00 AM Break 11:00 AM–11:30 AM Maternal Immunization and Nonclinical Safety Assessment Martha Leibbrandt, Novartis Vaccines, Cambridge, MA S16–6 11:30 AM–12:00 Noon Safety Assessment of Residuals and Contaminants in Vaccines Deborah L. Novicki, Novartis Vaccines, Cambridge, MA 60 th 35 American College of Toxicology Annual Meeting S16–2 The Office of Vaccines Research and Review (OVRR) is responsible for regulatory review of new Investigational New Drug Applications (INDs) and Biologics License Applications (BLAs) for preventive vaccines and therapeutic vaccines for infectious disease indications. Through this review process, OVRR ensures that vaccines are safe, effective, pure, and potent, as specified in Title 21 of the Code of Federal Regulations, Sections 312, 600, and 610. This presentation will review the key components in nonclinical and clinical safety assessment of investigational vaccines regulated by OVRR, with a focus on product testing and characterization, and pharmacology and toxicology study design considerations, for vaccines with novel adjuvants. In addition, updates on clinical trial design considerations, including demonstration of the “added benefit” of the adjuvant, adjuvant dose selection, and safety monitoring will be discussed briefly. S16–3 This talk will provide a global overview of the regulatory framework for the nonclinical evaluation of adjuvant systems and immunostimulants; and describe study designs and species selection for local tolerance, repeated dose, mutagenicity, reproductive/developmental toxicity, and safety pharmacology. It will also address considerations outlined in the 2013 WHO Guidelines regarding the nonclinical assessment of autoimmunity and hypersensitization and the inconsistencies between EMA and WHO guidelines for nonclinical assessment of adjuvant systems and immunostimulants. S16–4 Questions are often raised about the safety of vaccine adjuvants, particularly in relation to autoimmunity or autoimmune disease(s)/disorder(s) (AID). The International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute (HESI) formed a scientific committee (which included industry, regulators and academics), which included technical experts from academia, government regulatory agencies and industry to consider adjuvant safety and conducted a wide literature review and a two-day workshop. This presentation will summarize the groups considerations relating to key identified topics, which were as follows: 1) oil-in-water emulsions and Toll-like Receptor (TLR) agonists adjuvants, 2) use of animal models and 3) biomarkers for the evaluation and prediction of AID and key issues including: the value of animal models of autoimmunity for studying novel vaccine adjuvants; whether there is scientific evidence indicating an intrinsic risk of autoimmunity with adjuvants, or a higher risk resulting from the mechanism of action; and if there is compelling clinical data linking adjuvants and AID. S16–5 Routine vaccination of a pregnant woman for the purpose of protecting her unborn child against the effects of influenza, tetanus and pertussis is now common in many parts of the world. However, regulatory expectations for the type of nonclinical data needed to support maternal immunization with novel vaccine candidates or recently approved vaccines vary across geographies. Strategies, insights and examples pertaining to the design of reproductive and developmental toxicity studies for vaccines indicated for pregnant women in a global setting will be presented. S16–6 The talk will provide background information and an overview of the existing regulatory framework for the assessment of residuals and contaminants in vaccines. It will provide a basis, which can be used to formulate a general approach for the safety assessment of residuals and contaminants in vaccines. 61 WEDNESDAY S16–1 Vaccine adjuvants are substances that are used in conjunction with a vaccine antigen to increase or modulate the specific immune response to the vaccine antigen in order to enhance the clinical effectiveness of the vaccine. This presentation will provide a rationale for the use of adjuvants in vaccines. An overview of different adjuvant classes will be presented, with a focus on the current understanding about mechanisms of action and pathways used for immune stimulation. 2014 WEDNESDAY th 35 American College of Toxicology Annual Meeting 2014 EXHIBITOR-HOSTED PROGRAMS 12:00 Noon–12:55 PM See page 137 for more information. Wednesday Afternoon Session SYMPOSIUM 17 Hot Topics 1:30 PM–4:30 PM Grand Cypress Ballroom D CHAIR: Timothy J. McGovern,US Food and Drug Administration, Silver Spring, MD CO-CHAIR: Melissa C. Rhodes,GlaxoSmithKline, Research Triangle Park, NC Supported by an educational donation provided by: SciLucent 1:30 PM–2:00 PM FDA Tobacco/eCig Initiatives Michael Orr, US Food and Drug Administration, Silver Spring, MD 2:00 PM–2:30 PM Update on Biosimiliars Barbara Mounho-Zamora, ToxStrategies Inc., Bend, OR 2:30 PM–3:00 PM Animal Rights Extremists John Sancenito, INA Security, Harrisburg, PA 3:00 PM–3:20 PM Break 3:20 PM-3:30 PM Update on FDA/CDER Guidances, Initiatives; ICH Activities Timothy J. McGovern, US Food and Drug Administration, Silver Spring, MD 3:30 PM–3:45 PM PETA Questions the Value of AAALAC Accreditation—What Is Going on Here? David G. Serota, MPI Research, Mattawan, MI 3:45 PM–4:15 PM Update on Activities around the Ebola Outbreak Beth Leffel, Leffel Consulting Group, Washington, DC 4:15 PM–4:30 PM FDA Perspectives on Ebola Issues Christopher Ellis, US FDA/CDER, Silver Spring, MD 5:00 PM–6:30 PM Wine Tasting with 2015 ACT President Mary Ellen Cosenza (Free event for ACT members only, advance registration required) See page 10 for more information. 62 Grand View Terrace In case of rain: Grand Cypress G th 35 American College of Toxicology Annual Meeting 2014 ACT Poster Presentations Posters may be in place as early as Sunday afternoon and will be displayed from Monday through Tuesday at 4:30 pm. Designated authors (indicated in the Poster description by an underline) will present their poster during the Poster Reception Monday, November 10 from 5:30 pm to 7:00 pm. Please join this session to engage with the authors and see some of the latest work in the field. (ACT is not responsible for posting, removing, or storing posters.) ABSTRACTS Posters are located in the Grand Cypress Ballroom. 100 Series—General Toxicology 200 Series—Regulatory 300 Series—Safety Evaluation Nonpharmaceuticals 400 Series—Toxicology Methods 500 Series—Safety Evaluation Pharmaceuticals 63 th 35 American College of Toxicology Annual Meeting 2014 Poster Abstracts Poster abstracts are indicated with letters preceding the poster number: Poster (P), Student Travel Award (STP), North American Graduate Fellowship (GFP), and International Travel Grant (IGP). 100 Series—General Toxicology P101 Veterinary Pharmacovigilance Survey Conducted in Tamil Nadu State, India—A Status Report. G. Sarathchandra1,2. 1Pharmacovigilance Laboratory for Animal P103 Prevalence and Incidence of Cataracts in a Population of Yucatan Miniswines after Induction of Type I Diabetes. C. Hanks1, A. Ingerson1, M. Freeman1, S. Schlink1, L. Delaney1, S. Renna1, L. Brown1, A. Stricker-Krongrad1, J. Liu1, A. Tellez Cruz2, S. Rousselle2, J. Wicks2, G. Bouchard1. 1Sinclair Research Center ABSTRACTS Feed and Food Safety, Chennai, Tamilnadu, India, 2Tamil Nadu Veterinary & Animal Sciences University, Chennai, Tamilnadu, India Veterinary pharmacovigilance monitors the safety of veterinary medicines, including vaccines (VAC) used for the prophylaxis, diagnosis or treatment of diseases in animals once they reach the market after authorization. In India, there is no present government policy to survey and evaluate adverse drug events (ADEs) / Pharmacovigilance programme for veterinary medicines. Therefore, essential information such as frequency, severity of treated animal ADEs and reliable data about frequent ADEproducing drugs remains unknown. The objective of the study was to assess and communicate risks and benefits in the market. Ultimately to educate the veterinarians and the stakeholders on the safety and efficacy of veterinary drugs and biologicals. A 12-month period pilot study was conducted to monitor the ADE for frequently used drugs (labeled/extra labeled drugs). A survey protocol consisting of a questionnaire about used drugs in livestock was developed; the questionnaire was distributed to 300 veterinarians of Tamil Nadu state. The veterinarians were instructed to voluntarily report on the various types of drugs used and the ADEs, if any observed. More than 37% ADEs were related to antimicrobials, antiparasitic and anti-inflammatory agents. A further 27% of ADEs were due to vitamins and feed additives. Two cases of ADEs observed in FMD vaccination, in cattle and canine Parvo vaccine in dogs. In poultry, tiamulin and salinomycin ADEs induced serious mortality. The present study warrants for the need of sustained veterinary pharmacovigilance programmes in livestock for timely ADEs presenting drug detections and drug safety improvement. P102 Withdrawn LLC, Auxvasse, MO, USA, 2Alizee Pathology, LLC, Thurmont, MD, USA Cataracts as a consequence of chronic diabetes is considered a leading cause of legal blindness in humans in the United States and is also observed frequently in aged diabetic populations (>65%). Objective: Assess postinduction (PI) onset of ocular cataract(s) in a colony of over 266 castrated, male, diabetic, Yucatan miniature swine. Methods: Diabetic miniature swine were routinely screened by the veterinary staff for clinical ocular abnormalities including visible ‘mature' cataracts. Results: Over the course of a 6 month period, the prevalence was 30% (80 positive animals out of 266 animals). The most recent incidence (past 2.5 months) was 20.4% (38 positive animals with 60 affected eyes from pool of 186 previously negative animals). Eighteen animals had bilateral and 20 animals had unilateral cataracts (OD: 31; OS: 29). Cataract onset ranged from 2 to 19 months PI with an average of 11 months PI. Cataracts were detected earlier in animals when euglycaemia was intentionally less controlled, which supports the current predominant theory of glycation-induced cataract development. Interestingly, swine unlike human are not capable of glycating their hemoglobin due to the lack of penetration of glucose into the red cells. Miniswine with cataracts appear to function acceptably well despite the assumed visual handicap. Conclusions: Diabetic Yucatan miniature swine commonly manifest with cataracts on average at 11 months postinduction. Insulin regimen and glucose control are strong factors in the prevalence and incidence of cataracts in diabetic miniswines. The diabetic miniswine would provide a good model for preventative or therapeutic cataract therapies. 64 th 35 American College of Toxicology Annual Meeting P104 Reproductive and Developmental Toxicology Assessment of Oral Dietary Calcium Formate in Yucatan Miniature Swine. T. Madsen1, D. Hobson2, C. Selby1, G.P. Georges4, R. Hanzlik3, D. Brocksmith1, A. Stricker-Krongrad1, J. Liu1, G. Bouchard1. 1Sinclair Research Center, LLC, Auxvasse, MO, USA, LoneStar Pharm Tox, LLC, Boern, TX, USA, 3University of Kansas, Lawrence, KS, USA, 4NephroTech 1, LLC, Shawnee, KS, USA 2 Calcium Formate (CaF) is being considered as a dietary calcium supplement. Objective: Assess the reproductive/developmental toxicity of oral dietary CaF. Results: Female fertility index/group ranged from 80-100%. Total fetuses delivered were 174 with 170 livebirths. Piglet viability was good. Length of gestation, birth weights and body weight change were comparable. Litter size was robust (range 5.4-7.1) for all groups. CaF supplementation was associated with decreased food intake in both parents in the high dose group and with decreased weight gain in both parents during the premating phase of the study. These between group differences persisted through the gestational phase for the females, but were not statistically significant. CaF supplementation was also associated with increased serum calcium levels in both parents. Although statistically significant, the magnitudes of these differences were small, and well within the normal physiologic ranges.There were no significant findings of developmental/reproductive toxicity related to dietary exposure to CaF. Conclusion(s): Decreased food intake, decreased weight gain during premating phase, and increased serum calcium were associated with oral dietary CaF administration. These findings were consistent with previous research, and/or represented expected responses to the administration of oral CaF supplementation. P105 Toxicokinetic Approaches to Improving Accuracy of Drug Withdrawal Times in Food Producing Animals to Avoid Toxic Violative Tissue Residues. J. Riviere1, Z. Lin1, M. Li1. Institute of Computational Comparative Medicine, Kansas State University, Manhattan, KS, USA1 Monitoring for potentially toxic violative chemical residues in meat, milk and eggs is the primary regulatory tool to ensure chemical food safety in edible products from food producing animals. Although analytical approaches to monitoring residues have dramatically improved (2012 multi-residue MS/MS methods), toxicokinetic models used to establish drug withdrawal times (WDT) to avoid violative residues have not changed since their inception decades ago. WDTs are determined in healthy animals with a number of simplifying assumptions related to analysis of tissue depletion data (single log-linear decay, constant parent-drug to metabolite ratios, breed or disease effects on depletion kinetics) that are violated when animals are given label doses in field conditions; further exacerbated if drugs are legally administered off-label when increased dosages are used to treat resistant pathogens. Two toxicokinetic-modeling techniques can be used to make WDT estimation more realistic relative to variability seen in field conditions; physiological-based (PBPK) and mixed-effect population pharmacokinetic (PopPK). Both allow incorporation of disease effects on distribution (protein binding), elimination or biotransformation based on published literature; approaches could be considered meta-analysis tools that allow relevant data to be incorporated in WDT estimations. Efficiency of these approaches were assessed using published and new experimental data from tissue depletion studies on penicillin-G, oxytetracycline and flunixin in cattle and swine. Results using both PBPK and PopPK modeling for all drugs suggest that disease and dosing route changes have major impact on WDT and could result in residue violations even when label dosages are employed. (Supported by FARAD USDA-NIFA 2013-41480-21001) P106 Interaction of Silver and Gold Nanoparticles with Proteins, Biocorona Formation and Its Impact on Cellular Toxicity. N. Monteiro-Riviere1, A. Sasidharan1, R. Chen1, J. Riviere1. Kansas State University, Manhattan, KS, USA1 Nanoparticles (NP) interact with proteins to form a protein corona in biological fluids that can influence their biodistribution. The dynamic interaction of the protein corona with NP may significantly affect the bio-identity of the NP, thereby leading to diverse biological outcomes at the cellular level. Since NPprotein interactions decide the fate of a NP in vivo, understanding the complexity and dynamic interaction of the NP-protein formation is imperative. Time dependent adsorption kinetics of the protein corona were conducted with lipoic acid and BPEI coatings of 40 and 80nm silver (AgNP) and gold NP (AuNP). Ag and AuNP's were exposed to human serum albumin (HSA; 40mg/ mL), fibrinogen (2mg/ml), immunoglobulin G (IgG; 12mg/ml) and transferrin (2.5mg/ml) at their physiological concentrations from 6-24h. These studies indicated that irrespective of size or surface chemistry, rapid and prominent binding of HSA corona formed over both surface coatings with AgNP causing a slight increase in size, without aggregation. In contrast, the fibrinogen corona showed a time dependent decrease in NP stability leading to aggregation at 6h at 37°C. Interestingly, the transferrin corona induced aggregation in 80nm AuNP, while 40nm AuNP remained stable at 24h at 37°C. Further, the impact of the biocorona formation with human epidermal keratinocytes (HEK) and human vascular endothelial cells (HUVEC) were affected. Our results suggest that NP-protein interaction significantly affects the agglomeration and surface charge that further modulates the cellular responses in HEK and HUVEC. 65 ABSTRACTS Experimental Procedures: Sixty (30 male and 30 female) sexually mature minipigs were gender paired for breeding and randomized into 3 groups of 10 pairs receiving untreated control, low dose (2.25% CaF), and high dose (4.5% CaF) in the daily diet. Standard reproductive and developmental variables were assessed, including clinical, gross, and microscopic pathology (parents and piglets). 2014 th 35 American College of Toxicology Annual Meeting P107 In Silico Toxicity Evaluation of Marine Molecules with Antifungal Profile. Castro HC1, Santos TAN1, Mendonça AT2, Cabral LM2, Rodrigues CR2. 1Post-graduation Programs in Science and Biotechnology and ABSTRACTS Pathology, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil. 2Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. The fungi resistance against the antimicrobials is currently the major challenge for a proper infection treatment. Literature describes several new antifungals from natural sources such as marine organisms, but the toxicological aspects are still unknown due to the difficult on isolating and testing them. Since the use of in silico toxicology tools may help in selecting new molecules for further in vitro and in vivo toxicological evaluation, our purpose was to characterize the in silico toxicological properties of 19 marine products with antifungal potential described in the last 5 years to guide the selection for further in vitro and in vivo studies. In this work we also compared these molecules with antifungals in use such as fluconazole and voriconazole. Thus we used the programs for toxicological prediction, LAZAR and OSIRIS for evaluating 19 marine products that vary on the structural complexity degree (high, medium and low). The lowest risk toxicity profile was observed for nine compounds, including those of low complex structure (e.g. halogenated furans and curcudiol) showing a promising profile for future in vivo studies. Differently two compounds, including cucurphenol presented the highest risk profile and lower priority for in vitro and in vivo evaluations. This theoretical study reinforced the use of these tools as a step that may be included as a laboratory routine for studying marine compounds and others of difficult obtention but with high biological potential for human treatment. This step may help in selecting and identifying the potential of these molecules as safer antifungals agents. P108 A Comparison of the Type of Spontaneously Occurring Microscopic Lesions in the Göttingen and Chinese Bama Minipig. T. Zhou1, S. McPherson1, X. Yang1. Wuxi Apptec, Suzhou, China1 The minipig is often used in regulatory preclinical toxicology studies as the choice of the nonrodent species. It presents a favorable profile as a nonrodent toxicology model in terms of the similarity to man and also in terms of applicability to different study types (Bode et al 2010). Studies for general toxicology programs can be performed by oral, dermal, parenteral and inhalation routes. Physiologically the minipig also has advantages for use in reproductive and safety pharmacology studies. In USA and Europe the Göttingen minipig is often selected, whilst in China the Bama pig is usually the strain of choice. Histopathological data from control animals from toxicology studies using the Bama at Wuxi Apptec Suzhou were compared to published data for the Göttingen minipig. The type of lesions and also systems affected were compared to see if there were any notable differences between the two strains of animals. Generally the findings between the two strains were similar. However, one notable difference between the two strains of minipig was that for the Bama minipig the presence of hyaline droplets accumulation 2014 (minimal to moderate) in the glomerulus of the kidney, this hasn't been reported in the Göttingen pig. P109 Evaluation of Background Tumor Incidence in rasH2 Mice and Clinical Pathology Data from rasH2 Non-Transgenic and Transgenic Mice. K. Bonnette1, M. Morse1. Charles River, Spencerville, OH, USA1 A 26-week carcinogenicity study in rasH2 (CByB6F1-Tg(HRAS)2JIC) mice is recognized by ICH S1B (Testing for Carcinogenicity of Pharmaceuticals) as an alternative to a conventional 2 year bioassay in mice. The conduct of these studies is dependent on a low number of background tumors in controls and a notable number of background tumors in concurrent positive controls. Additionally, clinical pathology measurements are often collected during the 28-day carcinogenicity range-finding studies to monitor toxicity and pharmacology endpoints. In these range-finding studies, rasH2 hybrid (or nontransgenic littermates) mice are typically used as they are genetically similar to their transgenic littermates and the absence of the transgene does not impact toxicity. Therefore, we wanted to determine if our facility's background incidence of neoplastic findings in the rasH2 mouse is within expected limits reported in the literature. Additionally, we wanted to establish clinical chemistry and hematology historical control ranges for both transgenic and nontransgenic rasH2 mice and to compare them to the CD-1 mouse historical reference ranges as this strain of mouse is typically used in 2 year bioassays. Our results indicate that our incidence of common neoplastic findings was similar to those published in the literature. Additionally, we determined that the majority of clinical pathology parameter ranges were similar between the strains of mice; however, when older rasH2 transgenic mice were compared to younger nontransgenic littermates, and age matched CD-1 mice, some parameters (e.g. aspartate aminotransferase, aspartate aminotransferase, alkaline phosphatase) demonstrated wider ranges. P110 The Postnatal Development and Growth of the CardioRespiratory System in Sprague-Dawley Rats. A. Apreutese1,2, C. Gordon1, R. Foster3, A. Graham1, B. Palate3, J. Haruna1, M.O. Benoit-Biancamano2. 1CiToxLAB North America, Laval, Quebec, Canada, 2Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada, 3CiToxLAB France, Évreux CEDEX, France The purpose of this study was to investigate the histomorphological changes of the cardio-respiratory system in rat pups over the first month of life. The heart weight and the heart weight relative to body weight ratio were calculated for 51 rats over 13 timepoints, ranging from postnatal day 1 (PND1) to PND30. For each timepoint, whenever possible, equal number of females and males were used. The aortic wall progressively increased in thickness throughout the duration of our study as a result of an accumulation of extracellular matrix (collagen and elastic fibers). Postnatally, the cardiac volume essentially increased by cardiomyocyte hypertrophy, which showed a more mature appearance around PND21. In the newborn myocardium, mitotic figures and apoptotic bodies were frequently seen, increased on 66 th 35 American College of Toxicology Annual Meeting 2014 complicated, and is affected by systems level effects such as multiple feedback processes within and between the various onand off-target pathways. These systems level processes are often impossible to reconstruct in vitro as they involve many cell types, tissues, and organs systems throughout the body. We show here that through mathematical modeling we were able to identify, in silico, molecular properties that are critical to driving functional selectivity. The models, although simple, capture the key systems pharmacology needed to understand the balance between known on- an off- target effects. Surprisingly, in this case, the key driver of functional selectivity is not the affinity of the drugs but rather the pharmacokinetics, with drugs having a short half-life predicted to be the most functionally selective. P111 Histomorphologic Features of Neonatal and Juvenile Uro-Genital Development in Sprague-Dawley Rats. A. Apreutese1,2, C. Gordon1, R. Foster3, A. Graham1, B. Palate3, J. Haruna1, M.O. Benoit-Biancamano2. 1CiToxLAB North America, P113 Prediction of Severity and Clinical Outcome of Poisoning in India by Multiple Clinical Scoring Systems. S. Churi1, M. Ramesh1. JSS University, Mysore, Karnataka, India1 Laval, Quebec, Canada, 2Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada, 3CiToxLAB France, Évreux, France This study describes the histomorphologic key postnatal developmental events occurring in the uro-genital system of rat pups from birth to postnatal day 30 (PND30). Tissues were collected from 51 rats, using equal numbers from each sex whenever possible, at PND1, 2, 4, 6, 8, 10, 14, 17, 21, 24, 26, 28, and 30. Our study revealed that nephrogenesis in rats ceased by PND14, while few mitoses were still observed at the cortico-medullary junction at PND21. Individual cell death and mitoses were abundant at birth and followed a centripetal pattern (from outer cortex to medulla), accompanying developing renal structures. At PND26, the ovary exhibited a sharply demarcated cortico-medullary junction. Between PND21-PND26 numerous apoptotic ova were present. The first uterine glands became visible around PND14, whereas scattered apoptotic epithelial cells were still present up to PND26. By PND24, the superficial layer of the vaginal epithelium was multifocally composed by large cells containing a mucinous material mixed with few apoptotic cells. At birth, the seminiferous tubular epithelium was 1 to 2 layers thickened, composed of spermatogonia and Sertoli cells. A seminiferous lumen was rarely seen and the interstitium was abundant and hypocellular. The pachytene spermatocytes were observed around second week of life, while the first round spermatids became visible on PND30. The description of these major histological features of the male and female uro-genital systems from birth to PND30 will serve as valuable histological historical database that will be useful in pediatric drug development. Objective: Descriptive and prognostic evaluation scales (scoring systems), which are widely accepted worldwide, can be used to predict the severity and mortality rate of poisoning. We sought to assess the efficacy of GCS, PSS, APACHE II and SAPS II systems in predicting the severity and clinical outcome of pesticides, medicines and household products poisoning in India. Methods: One-year study (total patients = 198) was conducted at Indian tertiary-care teaching hospitals. GCS scores were calculated according to motor response to pain, and verbal and eye responses. PSS scores were calculated according to signs and symptoms of various systems and metabolism. APACHE II and SAPS II scores were calculated using the worst physiologic values in the first intensive care unit day. Based on the scores, the severity and clinical outcome of poisoning was predicted. Pearson correlation and Chi-square test were used to assess the statistical significance. Results: A majority of patients (n = 173, 87.4%) with mild to moderate predicted severity were recovered from poisoning. Patients with severe predicted illness were either discharged with severe illness (n = 14, 7.1%) or morbidity (n = 3, 1.5%) or expired (n = 8, 4%). There was a significant (P<0.05) association between the actual clinical outcome and scores of scales. There was also a moderate correlation between scoring systems (r = 0.61, P<0.01), indicating that tested scales provided similar efficacy. Conclusion: The study demonstrated essential sensitivity and excellent efficacy of these scoring systems to predict the severity and clinical outcome of poisoning in India. P112 A Systems Pharmacology Approach to Understand Efficacy and Toxicity to Optimize Therapeutic Window. J. Apgar1, J. Burke1. Applied BioMath, Winchester, MA, USA1 Most commonly the selectivity of a compound is defined in an in vitro or cellular assay, and it is thought of as principally a function of the binding energy of the drug to its on-target and off-target proteins; however, in vivo functional selectivity is much more 67 ABSTRACTS PND4 and showed a diffuse pattern. At birth, the trachea was lined by immature columnar ciliated epithelial cells, and submucosal glands became visible around one week after birth. The newborn rat has no alveoli and breathes with smooth, large gas exchange units termed "primary saccules". An intense interstitial cellular proliferation was observed in the lungs between PND6 and PND14, when the majority of alveoli are separated by new secondary septa, formed from the primary walls. These changes were accompanied by few mitoses and/or individual apoptotic cells. The findings described herein suggest that the cardiorespiratory system of rats is immature at birth. The data generated will serve as background historical database that will be valuable when performing postnatal developmental toxicity studies. th 35 American College of Toxicology Annual Meeting STP114 Mechanisms of Acetaminophen-Induced Cell Death in Primary Human Hepatocytes. Y. Xie1, M. R. McGill1, K. Dorko1, S. C. Kumer1, T. M. Schmitt1, J. Forster1, H. Jaeschke1. University of Kansas Medical Center, Kansas ABSTRACTS City, USA1 Acetaminophen (APAP) overdose is the most prevalent cause of acute liver failure in western countries. Numerous studies have been conducted to investigate the mechanisms of injury after APAP overdose in various animal models; however, the relevance of these mechanisms for humans remains unclear. Here we investigated APAP hepatotoxicity using freshly isolated primary human hepatocytes (PHH) from either donor livers or liver resections. PHH were exposed to 5mM, 10mM or 20mM APAP over a period of 48 hours and multiple parameters were assessed. APAP dose-dependently induced significant hepatocyte necrosis starting from 24h, which correlated with the clinical onset of human liver injury after APAP overdose. Interestingly, cellular glutathione was depleted rapidly during the first 3h. APAP also resulted in early formation of APAP-protein adducts (measured in whole cell lysate and in mitochondria) and mitochondrial dysfunction, indicated by the loss of mitochondrial membrane potential after 12h. Furthermore, APAP time-dependently triggered c-Jun N-terminal kinase (JNK) activation in the cytosol and translocation of phospho-JNK to the mitochondria. Both co-treatment and post-treatment (3h) with the JNK inhibitor SP600125 reduced JNK activation and significantly attenuated cell death at 24h and 48h after APAP. The clinical antidote N-acetylcysteine offered almost complete protection even if administered 6 hours after APAP and a partial protection when given at 15h. Conclusion: These data highlight important mechanistic events in APAP toxicity in PHH and indicate a critical role of JNK in the progression of injury after APAP in humans. The JNK pathway may represent a therapeutic target in the clinic. IGP115 Comparative Study of Muscular Regeneration after Necrosis Induced by Philodryas patagoniensis and Bothrops alternatus Snake Venoms. M.E. Peichoto1, M.N. Sánchez1, G.P. Teibler2, S.L. Maruñak2, O.C. Acosta2. 1Instituto Nacional de Medicina Tropical, Puerto Iguazú, Argentina, 2Facultad de Ciencias Veterinarias, Universidad Nacional del Nordeste, Corrientes, Argentina According to studies carried out with Bothrops (Viperidae) venoms, poor muscle regeneration is evidenced when tissues are affected by hemorrhage as this affects the microvascular supply of nutrients and oxygen necessary to accomplish a successful regeneration. On the other hand, the process of muscular regeneration after injury induced by Philodryas patagoniensis (Colubridae) venom (PpV) - which exhibits higher hemorrhagic activity than Bothrops alternatus venom (BaV) - has never been studied before. Thus, the purpose of this work was to evaluate and compare the regeneration of skeletal muscle fibers after necrosis induced by these two venoms. Mice were injected into the gastrocnemius muscle with 40 µg of either PpV or BaV. 2014 After 1, 3, 7, 14 and 28 days of inoculation, muscle fragments were extracted to have a qualitative histological assessment of the regeneration by staining sections with Hematoxylin-Eosin and Gomori's One-Step Trichrome. Samples showed an earlier appearance of regenerating fibers in muscles injected with BaV than with PpV. These results were consistent with those evaluating fibrosis, which showed an earlier and a higher amount of muscle tissue substituted by collagen fibers in PpV samples. These differences could be related with the different hemorrhagic activities exhibited by both venoms, and it would be the cause of the deficient muscular regeneration observed in P. patagoniensis envenomation, both experimentally and clinically. On the whole, these observations help us to understand the local pathological effects associated with viperid and colubrid envenomings, and they should be considered in both diagnosis and prognosis of snakebite victims. IGP116 Nanotoxicity of Arsenic Trioxide in Liver and Kidney of Laboratory Rats. Y. Verma1, S.V.S. Rana1, Kavita Rana1. C.C.S. University, Meerut, UP, India1 The impact of nanoparticles on human health and the environment is largely unknown, although many studies are under way in different parts of the world. While the small size of particles is what makes nanotechnology so useful in medicine and industry it is also one of the main factors that might make them potentially dangerous to human health. Arsenic is known human carcinogen. The adverse effects of Arsenic (III)(encapsulated with PLGA) nanoparticles cannot be predicted from known toxicity of bulk materials. We hypothesized that the noval properties may allow harmful interactions with biological system with the potential to generate toxicity. Therefore toxicological evaluation of arsenic trioxide is critical to establish the full in vitro potential of nanomedicine. Present paper describes the results on protein, MDA, GSH, GSSG, GST and CYP-450 in liver and kidney of laboratory rats treated with arsenic trioxide encapsulated with PLGA nanoparticles. Results also established the liver and kidney as accumulative organs for arsenic trioxide nanoparticles. Present study on toxicity of Arsenic (III) encapsulated with PLGA nanoparticles in rat expected to be useful from health risk of point of view among occupational workers involved in the manufacture and processing of nanoparticles and in general population using products containing these nanoparticles. GFP117 The Impact of B-Cell Activation on Cb2 Expression. J. Castaneda1, A. Harui1, S. Kiertscher1, M. Roth1. University of California, Los Angeles, Los Angeles, CA, USA1 Cannabinoids, the primary bioactive constituents of marijuana, activate cannabinoid receptor CB2, which is expressed on the surface of human B cells but not T cells. The toxicity of cannabinoids on immune function remains to be determined. In preliminary studies, human tonsillar B cell subsets were analyzed for the expression of cell surface CB2 by flow cytometry. While 68 th 35 American College of Toxicology Annual Meeting GFP118 Exposure to Neurotoxicant Results in Parkin-Dependent Changes in Proteasome Activity in Brain Regions Containing Differentially Susceptible Dopaminergic Neurons. T. Lansdell1, J.L. Goudreau2, K.J. Lookingland3. 1Dept. of Neurology Michigan State University, East Lansing, USA, 2Dept. of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA, 3Center for Integrative Toxicology, Michigan State University, East Lansing, MI, USA Parkinson disease (PD) is a neurodegenerative disorder in which motor symptoms result from the loss of nigrostriatal dopamine (NSDA) neurons, while those in the mediobasal hypothalamus are spared. The pattern of dopamine (DA) neuron susceptibility to neurodegeneration can be recapitulated with neurotoxicant exposure, including acute and chronic treatment with 1-methyl4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and rotenone. Expression of parkin mRNA and protein increases in the medial basal hypothalamus following MPTP exposure, a change that is temporally linked to the phenotypic and functional recovery of tuberoinfundibular dopamine (TIDA) neurons in the mediobasal hypothalamus. In contrast, there is no increase in parkin mRNA or protein in the ventral midbrain following MPTP exposure and these neurons do not recover function or phenotype. Parkin is an E3 ubiquitin ligase with an N-terminal ubiquitin like domain that can directly modulate the activity of the proteasome. 69 ABSTRACTS naïve (IgD+/CD38-/CD27-) and quiescent memory (IgD-/CD38-/ CD27+) B cells expressed cell surface CB2, activated B cells (IgD-/ CD38+/CD27dim) expressed low to no detectable surface CB2. We initially hypothesized that B cell activation might be responsible for down-regulation of CB2. Tonsillar B cells were therefore exposed for seven days in vitro to mega-CD40L and IL-4. This treatment promoted expression of the CD27 activation marker by all B cell subsets but also led to a selective down-regulation of cell surface CB2 only on activated memory B cells - not naïve cells still expressing IgD. Our findings suggest that CB2 is differentiallyregulated in naïve versus isotype-switched B cells. In ongoing experiments, isolated naïve B cells (>98% purity) will be activated with or without triggering by anti-IgD to examine differences in CB2 expression that occur before/after Ig isotype selection. Cells will also be treated with selective CB2 agonists and antagonists to determine the impact on isotype switching and the resulting expression of Ig subtypes. These experiments will establish a model system for examining the two-way interaction between CB2 receptors and signaling events that lead to B cell activation and differentiation. 2014 th 35 American College of Toxicology Annual Meeting 2014 200 Series—Regulatory P200 Selected Excipients Tested for Their Potential to Induce Hemolysis In Vitro in Different Species. M. Festag1, A. Eichinger-Chapelon1, C. Hager1, R. Kopf2, K. Ravuri2, C. Sarron-Petit1. 1F. Hoffmann-La Roche Ltd, Roche ABSTRACTS Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, CH - 4070 Basel, Switzerland, 2 F. Hoffmann-La Roche Ltd., Pharmaceutical Technical Development, CH - 4070 Basel, Switzerland Six different surfactants were studied in minipig, dog, cynomolgus monkey or human whole blood to identify the concentration where a disintegration or permeabilization of red blood cell membranes resulting in hemolysis and the release of hemoglobin would occur. Serial dilutions of N-Dodecyl-β-D-maltoside (DDM), Polysorbate 20 (PS20), Polysorbate 80 (PS80), Poloxamer 188 (P188), Kolliphor HS15 (Solutol), and N-Octyl-β-D-glucopyranoside (OGP) in 0.9% NaCl as the vehicle were prepared. The different concentration ranges were incubated with blood from the selected species and released hemoglobin was photometrically measured at 540 nm. Results were expressed in % hemolysis extrapolated from an internal standard curve. A less than 5% hemolysis was measured at concentrations of ≤0.195 mg/mL DDM, ≤3.75 mg/mL PS20, ≤9.36 mg/mL PS80 in dog blood and at higher concentrations in the other investigated species, ≤100 mg/mL (highest test concentration) P188 (≤80 mg/ mL in monkey blood), ≤6.25 mg/mL (highest test concentration) Solutol, and ≤1.56 mg/mL OGP in the tested species. A strong hemolysis (up to 100%) was induced by DDM and OGP at ≥1.563 mg/mL and ≥6.25 mg/mL, respectively in all investigated species. There was no major species difference identified in causing hemolysis at the investigated concentrations of the different surfactants. Due to the static nature of this in vitro assay compared to the in vivo situation determined concentrations are rather seen as a hazard than true risk assessment. P201 Value Added: A Case Study Where Unconventional Endpoints (Bone Composition and Bone Turnover Biomarkers) Provided Early Signals for Safety Monitoring in a Two-Week General Toxicology Dog. R. Yeager1, Y. Luo2, Y. Tian3, B. Hooker2, T. Cole2, A. Tovcimak2, K. Barnhart1, K. Whitney1, C. Graff3, R. Stoffel4, D. Honor1, R. Garg1, S. Fossey1. 1Preclinical Safety, AbbVie, Inc., North Chicago, IL, USA, Imaging, AbbVie, Inc., North Chicago, IL, USA, 3Drug Metabolism, AbbVie, Inc., Worcester, MA, USA, 4Immunology Discovery, AbbVie, Inc., Worcester, MA, USA 2 Early preclinical toxicology studies for small molecules generally consist of 5 to 14 days of dosing in a rodent and nonrodent species and are typically designed to evaluate for test item-related effects on a standard panel of clinical pathology parameters and an abbreviated tissue list for histopathology. This case study demonstrates the value of including additional/unconventional endpoints, specifically bone marrow fat, bone mineral density (BMD) and serum P1NP (total procollagen type 1 N-terminal propeptide; a bone formation biomarker), on a two-week dosage range-finding toxicology study in Beagle dogs with a test compound that may have a bone-related effect. Pathology findings from the standard clinical pathology panel and abbreviated tissue list for histopathology were considered nonadverse and were generally consistent with expected pharmacology. At the high dose, there was a significant increase in the bone marrow fat fraction from L4 vertebrae body. A similar trend was observed in the marrow from femoral diaphysis, but was not statistically significant. There was no significant test item-related effect on BMD. At all three dose levels, serum P1NP levels were decreased by approximately 80% on Dosing Day 14 compared to baseline. There was a strong inverse correlation between increased bone marrow fat fraction and decreased serum P1NP, which provided an early signal for undesirable pharmacology-related changes in bone. For test compounds that may have a bone-related effect, these additional study endpoints can be used for future compound safety screening in preclinical species and possibly be applied in clinical trials as safety monitoring biomarkers. P202 An Improved Workflow to Perform In Silico Mutagenicity Assessment of Impurities as per ICH M7 Guideline. R. Saiakhov1, S. Chakravarti1, A. Sedykh1. MultiCASE Inc, Beachwood, OH, USA1 Purpose: Use of in silico tools is one of the central points of ICH M7 guideline. However computational assessment of DNA reactive impurities is still challenging. Purpose of this study is to develop and apply an effective workflow using a QSAR statistical system and a quantitative read across methodology to obtain reliable genotoxicity assessments adhering to the ICH M7 recommendations. The novel multi-tier methodology provides the detailed information of toxicity alerts and reduces false positives, false negatives while increasing the test coverage. This in silico approach can be successfully used for assessment of mutagenicity of new drug candidates, impurities and metabolites. Method and Data: CASE Ultra is a QSAR statistics based computer program that automatically extracts structure-activity knowledge from chemical databases and applies the knowledge for predicting activity in test chemicals. Several models, developed by MultiCASE Inc alone and within Research Cooperation Agreement with Center for Drug Evaluation and Research of FDA were utilized, as well as a read across technique for quantitative prediction of toxicity that uses the neighborhood profiles of chemicals. A multi-tier approach in combining several models and read across engine for bacterial mutagenicity evaluation resulted in significant improvements in the sensitivity, specificity and coverage. The details of the methodology and results of prediction for a large external test set are presented. 70 th 35 American College of Toxicology Annual Meeting Results of the Study: The improvements in the predictive performance as a result of the effective workflow is demonstrated for various scenarios of mutagenic impurity assessments, using an external validation set. P203 Safety Assessments of Food Ingredients Require Consumption Analysis. R. Matulka1, A. Morales1, S. Ulm1. Burdock Group, Orlando, FL, USA1 P204 Defining Components of an Expert Review necessary for ICH M7 Compliant Submissions. G. Myatt1, R. Daniel Benz2, K. Cross1. 1Leadscope, Columbus, Ohio, USA, 2OmnyCorp, Rockville, MD, USA The recently published International Conference on Harmonisation (ICH) M7 guideline includes the use of in silico analysis to qualify actual or potential impurities as nonmutagenic. According to M7, two methodologies, one rule-based (structural alerts) and one statistical-based (QSAR models), should be used and the outcome reviewed using expert knowledge. This expert opinion can "...provide additional supportive evidence on relevance of any positive, negative, conflicting or inconclusive prediction..." This poster outlines a list of items to consider as part of the expert evaluation, starting with an assessment of the quality of any available laboratory data on the impurity. In the absence of such data, the results of applying expert alerts and statistical QSAR models covering both A:T and G:C mutations should be evaluated, including: 1) applicability domain considerations, 2) definitions and relevance of alerts and structural features used in the QSAR models, 3) mechanistic rationale, 4) the quality of data supporting the prediction results, and 5) the relevance of any deactivating fragments. The expert analysis should also describe how the results from the two methodologies were combined into an overall consensus prediction. Information on chemical analogs may also be included in this expert opinion. Other considerations, including arguments based on the chemical environment of alerting substructures as well as changes to limits based on in vivo data should be discussed when appropriate. This poster presents a semi-automated framework to rapidly generate expert opinions that are transparent, consistent and traceable. P205 Ensuring Regulatory Acceptable (Q)SAR Models and Expert Alerts for ICH M7 Reflect Proprietary Chemical Space. K. Cross1, N. Kruhlak3, G. Myatt1, L. Stavitskaya3, A. White2. 1Leadscope, Columbus, OH, USA, 2GlaxoSmithKline, Ware, Hertfordshire, UK, 3US Food and Drug Adminstration, Silver Spring, MD, USA The International Conference on Harmonisation (ICH) M7 guideline permits the use of two in silico methodologies to qualify any actual or potential drug impurities as nonmutagenic. Statistical models and expert alerts built from public domain knowledge and data are used for ICH M7 assessments as they provide the necessary transparency and are sufficiently predictive for this purpose. Knowledge from proprietary data increases the specificity for selected chemical classes as well as expands the (Q) SAR models' applicability domain. This leads to less laboratory testing and synthesis of the impurity for the sponsor. We have investigated the use of proprietary data made available through confidentiality agreements directly with pharmaceutical sponsors to identify potential solutions to specific model predictivity issues, and are working towards the release of appropriate knowledge, compounds, and experimental data necessary to solve the (Q) SAR issue while preserving model transparency. The end result will be a regular transfer of knowledge for use in the development of (Q)SAR statistical models and incorporation of those data into the Leadscope alert-based expert system. This poster outlines the collaborative program for sharing data and knowledge for inclusion in regulatory acceptable public models without releasing any confidential business information. The process for knowledge sharing through the use of structural fingerprints for several compound classes will be discussed. A case study will be presented where, as a result of the incorporation of knowledge derived from proprietary data, the specificity of a model to predict primary aromatic amines was increased by 15% with no decrease in sensitivity. 71 ABSTRACTS The safety of food ingredients has recently come into question, generated by an incomplete understanding of the safety assessment process necessary to determine that a potential food ingredient meets the standard of a "reasonable certainty of no harm" under the intended conditions of use. Critical to the safety assessment is the analysis of potential exposure on a daily basis, based on the categories of foods to which it will be added. With the use of our proprietary consumption software, we are able to calculate the mean, median, 90th and 95th percentile intake of an ingredient consumed, utilizing the USDA two-day food consumption survey data set entitled "What We Eat in America" obtained through the National Health and Nutrition Examination Survey (NHANES) every two years. We calculate the potential 90th percentile intakes by gender and different age groups, as determined by the survey data to represent consumption by different subpopulations of US consumers. The 90th percentile level of food intake is a purposeful exaggeration built into the analysis. The consumption data provides an estimated daily intake (EDI) which can then be compared to the acceptable daily intake (ADI), derived from the lowest No Observed Adverse Effect Level (NOAEL) from safety studies conducted with the ingredient. A basic criterion for determining the safety of a food ingredient is that the EDI must not exceed the ADI. Because risk is based on both hazard and exposure, our analysis of the animal safety data and our consumption software support the "safety-in-use" of food ingredients. 2014 th 35 American College of Toxicology Annual Meeting P206 SEND Architecture Facilitates Harmonization and Integration of Data from Multiple LIMS and Controlled Terminology Versioning. F. Mura1, R. Buchanan2, M. Wasko2, C. Wuermlin1, L. Kaufman2. 1PDS Preclinical Data Systems, Inc, Basal, Switzerland, P207 Comparative Pharmacology of iPSC-Derived Cardiomyocytes Assessed by Conventional Current Clamp and Multi-Electrode Array (MEA). A. Bruening-Wright, C. Obejero-Paz, N. Federov, J. Kramer, A. Brown. ChanTest Corporation, Cleveland, OH, USA PDS Preclinical Data Systems, Mt Arlington, NJ, USA 2 ABSTRACTS 2014 FDA guidances for submission of clinical and nonclinical data in standard electronic format are expected to become binding towards the end of 2016. Preparing for the nonclinical standard, SEND (Standard for the Exchange of Nonclinical Data) is a process that involves participation of multiple preclinical departments in addition to IT. Producing SEND compliant datasets requires consistency of metadata across all domains regardless of data source/ LIMS used for data collection. Correlations between records from different XPT files are required for data interpretation, as is inclusion of all data collected for a toxicology study: in-life, bioanalytical, pk, clinical pathology, postmortem, and planned and actual study design. A SEND solution must also map a sponsor’s terms to CDISC controlled terminology, along with the ability for versioning based on frequent controlled terminology releases/ updates. We wish to report a web-based software architecture for SEND that accomplishes the aggregation, harmonization, and translation of data from different sources into one complete SEND dataset including all XPTs, define files, and validation report. The architecture involves input from LIMSspecific or file-based adaptors, which are then processed through an engine that maps data to appropriate variables and domains; harmonizes metadata between domains, consolidates comments, specifies relationships, and performs controlled terminology translation. Controlled terminology versioning is managed with a self-learning technology that avoids the time-consuming activity of remapping terms that have not changed. Trial information is provided to the engine through a specialized applicationprogramming interface (API), and expert tools were developed to allow study-specific customization. The goal of this research was to compare drug effects on the electrophysiological properties of iPSC-derived cardiomyocytes (Axiogenesis Cor.4u cells) using two techniques; MEA and conventional (manual) patch clamp in current clamp (CC) mode. MEA experiments were performed using a Maestro instrument from Axion BioSystems (Atlanta, GA). CC experiments were done using MultiClamp 700 A and B amplifiers in cells grown in 35 mm culture dishes covering an area of approximately 1 cm2. Both MEA and CC experiments were carried out at 35°C in the absence of serum. Field potential and action potential durations were prolonged by hERG channel blockers (terfenadine, dofetilide, cisapride) and shortened by calcium channel blockers (verapamil and nifedipine). The electrical activity in both preparations was abolished by tetrodotoxin at comparable concentrations (3–10 μM). Our results indicate that field potential changes observed in MEA studies parallel the changes observed in action potentials recorded in comparable culture conditions. As a corollary, CC experiments can be used as a follow up technique to further characterize pharmacological mechanisms observed in MEA studies. 72 th 35 American College of Toxicology Annual Meeting 2014 300 Series—Safety Evaluation Nonpharmaceuticals P300 Metabolic and Endocrine Disorders in Rats Exposed to Malathion: Risk of Diabetes Type 2 Induction. R. Rezg2, B. Mornagui3, S. El-Fazaa1, N. Gharbi1. 1University of Tunis El Manar, Faculty of Sciences of Tunis, El Manar, Tunisia, 2 University of Monastir, High Institute of Biotechnology of Monastir, Monastir, Tunisia, 3University of Gabes, Faculty of Sciences of Gabes, Gabes, Tunisia The aim of this study was to investigate the effects of subchronic exposure to malathion, on metabolic parameters and hypothalamic corticotropin-releasing hormone (CRH) mRNA expression. Malathion was administered to Wistar rats by gavage for 32 days at a dose of 100 mg/kg bw/day. Malathion intoxication decreases hepatic Glycogen Phosphorylase activity and increases Hexokinase activity with a rise in hepatic glycogen rate. Malathion decreased hepatic proteins and lipid contents that could be associated to liver gluconeogenesis. We have recorded a significant decrease in muscular glycogen rate, which indicates a stimulated glycogenolysis.We suggest that malathion can caused a turnover of glucose by its release through a succession of glycogenolysis and gluconeogenesis, which involves abnormal hyperglycaemia and its storage via glycogenesis, in subchronic exposure to an OP compound. Continued repetition of this circle exhausts the metabolic control system, resulting in insulin resistance. Also, Malathion exposure induced a significant decrease CRH mRNA. This result is in accordance with that of diabetic type 2 rat model. We conclude that OP, the most widely used classes of pesticides present an important risk factor to diabetes type 2 induction. P301 Integrated Use of Biomarkers (Oxidative Stress Biomarkers and Genotoxicity) in Mussels Lamellidens marginalis for Assessment of Monocrotophos Toxicity. A. Mundhe1, S. Pandit1. University of Pune, Pune, Maharashtra, India1 Genotoxic studies evaluate the effects of pollutants on organisms and consequently, their implications on human health. Monocrotophos (MCP), an organophosphate pesticide is widely used in India for control of various pests. The present study is undertaken to evaluate the oxidative stress and genotoxic potential of MCP on Lamellidens Marginalis and repair of the damaged DNA after exposure. The results clearly indicated that exposure to MCP caused increase in the number of damaged nuclei in gill cells. After the recovery period, a significant reduction in the number of damaged nuclei was observed indicating repair. Our findings suggest that MCPinduced oxidative stress may, partly, be contributing to genotoxic damage of gill cells. P302 Toxicology and Risk Assessment of Chemical Mixtures. M. Krishan1, A. Kretser1. International Life Sciences Institute North America, Washington, DC, USA1 Evaluating potential health risks posed by exposures to multiple chemicals is challenging for toxicology research and risk assessment (RA). Problem formulation is complex and mixturesRA frameworks vary greatly among agencies. This study 1) describes RA approaches for chemical mixtures; 2) characterizes differences in RA frameworks; and 3) assesses regulatory acceptance of these frameworks with a focus on food mixtures. Reviews were completed for mixture RA paradigms used by US federal agencies, nonprofit organizations (WHO, IPCS) and international agencies (EU). There are significant overlaps and differences between paradigms and there appears to be no single unified approach. Considering the current challenges in food mixtures, a hypothetical case study was conducted using a tiered screening approach and hazard index method to evaluate the effects of mixtures in foods. A hypothetical new bean product is being considered to replace pinto beans for a food program. Component levels of the following differ between old beans (OB) and new beans (NB) (assuming a 10% decrease): cadmium, deltamethrin, cyfluthrin; and bisphenol A levels are the same in OB and NB. Noncancer health effects following chronic oral exposure were evaluated. First tier provided a crude filter using the most recent and conservative health reference values for mixture components. Some assumptions in the first tier were refined in the second tier. Components were grouped based on health effects and biomonitoring data was used to estimate exposure in the second tier. Results indicate that tiered approach is a useful way of rapidly screening the effects of chemical mixtures. The animals were exposed to 5.25 mg/l of MCP for 7 days and after treatment, allowed to recover for 4 days in pesticide-free water. The involvement of oxidative stress was examined by estimation of Thiobarbituric acid reactive substances (TBARS); increase in the 73 ABSTRACTS Diabetes type 2 is recognized as a global major public health problem. Obesity, lifestyles and rich diets in fats are known risk factors for this disease, but recent evidence also points to environmental toxicants as an important factor to Diabetes type 2 induction. Our attention has turned to the organophosphate pesticides (OP), which represent 50% of all the insecticide use worldwide and especially to Malathion. levels of TBARS was recorded in the gill, muscle, foot and mantle tissues. Cellular antioxidant defences i.e. antioxidant enzyme activities like catalase, superoxide dismutase, glutathione-Stransferase and glutathione reductase were used as biomarkers of oxidative stress. Altered activities of antioxidant enzymes were observed in the tissues exposed to MCP. There was a significant recovery in the above parameters in the tissues after the recovery period. To monitor genotoxicity of MCP, we used micronucleus and comet assay. Significant DNA damage was measured in terms of the Olive tail moment in gill cells of treated animals and compared to control. th 35 American College of Toxicology Annual Meeting P303 High-Content Screening Analysis of the Biological Impact of Harmful/Potentially Harmful Constituents of Tobacco Smoke. I. Gonzalez Suarez1, D. Marescotti1, S. Acali1, S. Johne1, S. Frentzel1, C. Mathis1, J. Hoeng1, M. Peitsch1. Philip Morris ABSTRACTS International R&D, Neuchâtel, Switzerland1 Exposure to cigarette smoke (CS) causes lung toxicity and increases the risk of developing chronic obstructive pulmonary disease and cancer. CS is a complex aerosol with over 6000 chemicals, thus it is difficult to determine individual contributions to overall toxicity, as well as the molecular mechanisms by which smoke constituents exert their effects. Previously [1], we performed a systems toxicology evaluation of a subset of 14 CS constituents categorized as harmful/potentially harmful by the Food and Drug Administration. Here, we investigated the biological impact of additional 34 CS constituents, on normal human bronchial epithelial (NHBE) cells. Cytotoxicity was evaluated using and impedance-based, multi-electrode array system. Furthermore, a High Content Screening (HCS) analysis platform was used to measure 13 multi-parametric indicators of cellular toxicity over a wide range of concentrations and at different time points. Toxicity profiles varied dramatically among the different tested CS constituents, with EC50 values ranging between 0.01-10mM after 24h of exposure. Based on HCS results, the most important toxicity mechanisms were: DNA damage/growth arrest, oxidative stress, mitochondrial stress and apoptosis/necrosis. Some CS constituents did not induce a toxic response in NHBE cells despite the high doses tested. In summary, combination of multiple HCS endpoints allows to investigate the biological impact of CS constituents in a rapid and reproducible manner, while gaining insight on the molecular mechanisms activated upon exposure. 1. Gonzalez Suarez et al. (2014) Systems biology approach for evaluating the biological impact of environmental toxicants in vitro. Chem Res Toxicol 27: 367-376. P304 Investigating the Performance of an In Silico Salmonella Mutagenicity Model with HPHCs from Tobacco Products and Tobacco Smoke L.G. Valerio Jr.1, R.P. Yeager1. US FDA Center for Tobacco Products (CTP), Office of Science, Division of Nonclinical Science, Silver Spring, MD, USA1 In silico toxicology approaches are of interest as risk-based nontesting strategies, to reduce the necessity to conduct animal studies, and accelerate knowledge on potential toxicities of chemicals when experimental toxicology testing are absent or equivocal. This research assessed the utility of a novel in silico quantitative structure-activity relationship model to predict Salmonella mutagenicity of constituents from tobacco products and tobacco smoke. The model is trained with 8,700 chemicals in the computational platform, Symmetry. The computation uses k-nearest neighbors and Logistic regression algorithms, and the Mold2 molecular descriptor software. Harmful and potentially harmful constituents (HPHC) from tobacco products and tobacco smoke (FR 20034, 77:64, 2012) can be an integral part of the nonclinical toxicological risk assessment process at FDA/CTP. The research examined the model's predictive performance and chemical space with HPHCs. All HPHCs were within the model 2014 applicability domain, indicating sufficient chemical representation with tobacco constituents. Predictive performance characteristics showed 90% sensitivity, 0% false positive rate, 10% false negative rate, and a 93% concordance overall in predicting DNA reactive mutagens, and nonmutagens with the model. These screening results show the knowledge base in the model is capable of predicting tobacco constituents known to cause DNA damage that leads to mutations and potentially cancer, suggesting its potential usefulness for analyzing other constituents when study data are unavailable. This study is also supportive of the application of computational predictive models as tools for rapid decision-support to inform nonclinical risk assessments on potential genotoxic/carcinogenic hazard of new chemicals in tobacco products. P305 Effects of Cigarette Smoke Exposure on the Insulin Resistance-Related Metabolic Changes in the High-Fat Diet-Fed ApoE Knock-Out Mice. A. Iskandar1, D. Sharma2, S. Boue1, H. De Leon1, B. Phillips2, M. Cabanski1, E. Veljkovic1, N. Ivanov1, J. Hoeng1, M. Peitsch1. 1Philip Morris International R&D, Neuchâtel 2000, Switzerland, 2Philip Morris International Reserach Laboratories, Pte Ltd., Singapore 117400, Singapore Insulin resistance (IR) has been linked to cigarette smoking and it is known to contribute to diabetes and lipid disorders. Cigarette smoke (CS) induces inflammation and weakens immune responses, both of which can exacerbate diabetes and atherosclerosis development. We examined the effects of CS exposure on IR-related metabolic changes in the ApoE-deficient mouse, a model of dyslipidemia and atherosclerosis that has been shown to exhibit accelerated development of diabetes upon a high fat diet (HFD) feeding. Female ApoE-/- mice were fed a HFD for 11 weeks and were exposed to CS from 3R4F or filtered air (sham). Fasting glucose and insulin levels, aortic plaque area, serum levels of lipids and metabolic analytes, muscle and liver transcriptomes, as well as plasma and liver lipidomes were analyzed. CS-exposed mice had lower fasting glucose levels than sham-exposed mice, while insulin levels remained unchanged. Aortic plaque areas were increased in the CS-exposed group as compared to mice from the sham group. Lipidomics analysis showed increased levels of plasma and liver cholesterol esters, ceramides and sphingomyelins. Transcriptomics analysis suggested that CS exposure upregulated xenobiotic metabolizing enzymes and suppressed inflammatory/immune responses. Our results indicate that CS exposure interfered with the regulation of plasma glucose levels and increased plaque formation in ApoE-/- mice. The observed gene transcription changes within various metabolic pathways in the CS-exposed mice may account for the increased atherogenic lipids in the plasma and liver. The mechanistic links among xenobiotic metabolism, suppression of immune responses and diabetogenesis in the CS-exposed ApoE-/- mice remain to be elucidated. 74 th 35 American College of Toxicology Annual Meeting P306 Characterization of microRNA Expression Patterns and their Target Genes to Discover Potential Biomarkers for Hepatacaciongenesis after Exposed to Thioacetamide in Rats. Y. Wang1, C. Castro2, B. Gong1, Y. Morikawa3, J. Yan1, T. Uehara3, T. Chen1, W. Tong1. 1National center for Toxicological Research, USFDA, Jefferson, AR, USA, 2University of North Carolina, Charlotte, NC, USA, 3Shinonogi & Co., Ltd., Osaka, Japan P307 TRENDS in Human Adverse Events Associated with Pyrethroid Insecticides. F. Suarez1. Syngenta Crop Protection, Greensboro, North Carolina, USA1 Poison control centers have been shown to reduce the number of unnecessary emergency room visits due to poisoning incidents. American Poison Control Center (APCC) reports indicate that the number of reported incidents involving human exposure associated with pyrethroid insecticide exposure have been increasing annually; however, the proportion of cases treated at health care facilities (HCF) has not changed substantially. To further understand the reasons for seeking medical attention after incidental exposure to one pyrethroid (lambda-cyhalothrin L-C), data collected over a 10 year period was extracted from an adverse incidents report database (PROSAR). The information was analyzed in terms of demographics, case severity, primary route of exposure, and primary symptom or health concern. Results were compared with an evaluation of L-C human adverse events conducted by the US EPA; and with peer-reviewed published information available through PubMed. The majority of cases reported were from dermal exposures, were categorized as minor, and did not require medical attention. However, a number of cases reported inhalation exposures. Although observed less frequently than dermal exposures, most of these cases were categorized as moderate, and were more frequently treated at HCF. Content analysis of the description of the exposure event identified odor perception as a common theme in several of these cases. These findings suggest that recording patient's perception of chemical odors as evidence of exposure via inhalation could introduce bias in the interpretation of trends from adverse incidents control data P308 Comparison of the Biological Impact of Cigarette Smoke and a Prototypic Modified Risk Tobacco Product on Human versus Rat Primary Normal Bronchial Epithelial Cells. U. Kogel1, C. Poussin1, D. E. Messinis2, S. Frentzel1, C. Mathis1, J. Hoeng1, M. C. Peitsch1. 1Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland, ProtATonce Ltd, Scientific Park Lefkippos, Patriarchou Grigoriou & Neapoleos 15343 Ag. Paraskevi, Attiki, Greece 2 Smoking causes serious, fatal diseases such as lung cancer, cardiovascular and chronic obstructive lung diseases. Along with the current global paradigm change for safety assessment of consumer products, PMI R&D is working towards creating an integrative, systems biology-based approach for the evaluation and risk assessment of modified risk tobacco products (MRTPs). With the view to investigate the level of translatability between rodent and human biology, we exposed normal human bronchial epithelial (NHBE) cells and its rat counterpart normal rat bronchial epithelial (NRBE) cells to total particulate matter (TPM) generated from a prototypic modified risk tobacco product (pMRTP) and from the reference cigarette 3R4F for four hours. Exposure to the same dose of TPM from the 3R4F resulted in twice as many differential expressed genes in NHBE cells compared with NRBE cells (FDR<0.05, FC>0). The exposure to the same dose of pMRTP resulted in a weak transcriptional response in both species. A computational-modeling approach based on tissue-specific biological networks identified proliferation, apoptosis, and senescence as the most perturbed molecular processes after exposure of NHBE cells to TPM of the 3R4F. Cell stress were impacted to a lesser extent. In NRBE cells the most perturbed biological processes are the cell stress, followed by proliferation and senescence. These processes are much less perturbed after exposure to pMRTP in both species. In conclusion, after 3R4F exposure the highest species correlation is found for the cell stress sub-network NFE2L signaling. Exposure to pMRTP induces very low biological effects in both species. 75 ABSTRACTS Given the widespread roles of miRNA cross various biological processes, understanding their patterns of expression in chemical treatment across different doses and treatment durations will provide insights into the role of miRNA underlying toxicity. In this study, an integrated analysis of miRNAs expression with their target mRNAs was conducted to investigate the dynamic nature of miRNA-mediated regulating modules in rat livers exposed to a thioacetamide at multiple dose levels and treatment durations. At each dose- and time-point, both miRNAs and mRNA expression profiling of the same rat livers were generated with next generation sequencing and microarray, respectively. Comparison of differentially expressed miRNA and mRNAs revealed distinct dose- and time-dependent characteristics of two RNA features. We found that two miRNAs, miR-34a and miR-100 were consistently differentially expressed across all the doses and time-points examined. To our knowledge, miR-100, was firstly reported in rat liver in this study which is thought to be negatively regulating the mammalian target of rapamycin (mTOR), a central regulator of cell metabolism, growth, proliferation and survival. This suggests that miR-100 could potentially be used as biomarkers or therapeutic targets for liver cancer and drug-induced liver injury (DILI). Finally, the integrative analysis based on computational target prediction and miRNA/mRNA profiling defined a network of putative functional miRNA-target regulatory relations supported by expression data. These findings demonstrated the potential application of miRNAs as biomarkers to assess liver carcinogenicity and DILI at the early stage of drug discovery and development. 2014 th 35 American College of Toxicology Annual Meeting P309 A Meta-Analysis of Human Organotypic Respiratory Cultures Exposed to Whole Cigarette Smoke. P. Leroy1, C. Mathis1, M. Cabanski1, A. Iskandar1, R. Kostadinova1, S. Frentzel1, D. Kuehn1, S. Majeed1, C. Merg1, A. Elamin1, E. Guedj1, R. Dulize1, G. Vuillaume1, Y. Xiang1, F. Martin1, W. K. Schlage1, M. C. Peitch1, J. Hoeng1. 1Philip Morris ABSTRACTS International R&D, Philip Morris Product SA, Neuchâtel, Switzerland The development and the use of human three-dimensional in vitro models that closely mimic in vivo biology are of great interest to the scientific community to address some of the limitation of species translatability and further support the reduction, refinement, and replacement framework of animal use in laboratory. Human organotypic epithelial tissue cultures (e.g., bronchial, nasal), grown at the air-liquid interface, are particularly attractive to assess the impact of airborne toxicants exposure, including pollutants and cigarette smoke (CS). As organotypic tissue models are currently still expensive and not always accessible for small laboratories, having a well-defined experimental design is essential for scientifically sound outcomes. We investigated the ‘reproducibility' of different molecular endpoints collected after CS exposure of human organotypic bronchial and nasal epithelial cultures obtained from different production batches as well as from different donors. The following endpoints were assessed after different postexposure time points of 28 min exposure to air (Sham control) or to different dilutions of mainstream CS (3R4F - Health Canada regimen): cytotoxicity (AK assay), cytochrome P450 (CYP1A1/1B1) activity, pro-inflammatory markers secretion (MAP), histological assessment, and gene expression changes. The statistical analysis shows the impact and the contribution of the use of various donors, of different production batch, and of multiple exposure smoke experiments. P310 Integration of cII and Pig-a Mutation and Micronucleus Endpoints into the Big Blue® Transgenic Mouse Mutation Assay: Results for Benzo(a)pyrene (BaP) and N-Ethyl-Nnitrosourea (ENU). R. Young1, H. Dinesdurage1, D. Bruning1, L. Stankowski1, R. Kulkarni1, T. Lawlor1, M. McKeon1, Y. Xu1, S. Avlasevich2, D. Torous2, S. Dertinger2, M. Aardema3. 1BioReliance, Rockville, MD, USA, 2Litron Laboratories, Rochester, NY, USA, 3Marilyn Aardema Consulting, LLC, Fairfield, OH, USA Integration of multiple genotoxicity endpoints into repeat-dose rodent studies provides broad assessment of genotoxic potential of different modes of action while reducing the use of animals. Using male Lambda LIZ/lacI C57BL/6 homozygous (Big Blue®) transgenic mice, we examined the induction of cII mutations in liver and bone marrow, micronuclei in blood reticulocytes and mature erythrocytes (mnRET and mnNCE, respectively), and Pig-a mutant phenotype reticulocytes and erythrocytes (RETCD24- and RBCCD24-, respectively). Animals were treated with olive oil (5 mL/ kg/day for 28 days), BaP (50 mg/kg/day for 28 days), or ENU (40 mg/kg/day on Days 1, 2 and 3), with sacrifice on Day 31. Significant increases (p<0.001) in cII mutant frequencies were observed for BaP in liver (4.89-fold) and bone marrow (4.15-fold), and for ENU in liver (17.7-fold) and bone marrow (10.6-fold). Peripheral blood was collected ~3 hours before sacrifice and analyzed for 2014 Pig-a mutant phenotype and micronucleus frequencies by flow cytometry using MutaFlow® and MicroFlow® kits, respectively (Litron Laboratories). BaP induced significant increases (p<0.001) in mnRET, mnNCE, RETCD24- and RBCCD24- (1.56-, 2.11-, 616 , and 101fold, respectively). ENU induced significant increases (p<0.001) in mnNCE, RETCD24- and RBCCD24- (1.40-, 1050-, and 208-fold, respectively). As expected, mnRET were not elevated by ENU (p>0.05) due to the long interval between last administration and blood harvest. This study demonstrates the robustness of the Big Blue® Mouse Mutation Assay to detect induced mutagenesis by a direct acting mutagen and one requiring metabolic activation. This work also demonstrated the ability to integrate additional endpoints for mutation and clastogenicity. P311 Effects of Cigarette Smoke Extracts from a Prototypic Modified Risk Tobacco Product on Chemotaxis and Transendothelial Migration of Human Monocytes. M. van der Toorn1, S. Frentzel1, M. Peitsch1, J. Hoeng1, H. De Leon1. Philip Morris International, Neuchâtel, NE, Switzerland1 Introduction. Although it is well-established that cigarette smoking increases cardiovascular risk, the mechanistic links between cigarette smoke and atherogenesis are poorly understood. Aim. We determined the effects of cigarette smoke aqueous extracts (CSE) from 3R4F, a conventional cigarette, and a prototypic modified risk tobacco product (pMRTP) on monocyte chemotaxis and migration through a monolayer of human coronary artery endothelial cells (HCAEC). Methods. CSE were generated by smoke-bubbling RPMI media (sbMedia) from 3R4F and pMRTP according to Health Canada standards. Endpoints included cytotoxicity (cell death assessed by 7-AAD staining followed by FACS analysis), inflammation (cytokine measurements), THP-1 cell migration (Boyden chambers), and transendothelial migration (TEM) determined by real-time impedance. Results. Direct treatment of THP-1 cells with increasing concentrations of sbMedia from 3R4F and pMRTP induced dosedependent increases in cytotoxicity (cell death) and inflammation (IL-8, TNF-α release). Pretreatment of THP-1 cells or HCAEC with sbMedia from 3R4F and pMRTP decreased stromal cell-derived factor 1 (SDF1)-mediated migration and TEM in a dose-dependent manner. SbMedia from 3R4F was ~20 times more potent than sbMedia from pMRTP for all endpoints. Conclusion. sbMedia from 3R4F or pMRTP cigarettes induced dose-dependent responses in assays of inflammation, cytotoxicity, chemotaxis and TEM. Our findings indicate that CSE from a pMRTP are far less cytotoxic and less proinflammatory than CSE from conventional 3R4F cigarettes. 76 th 35 American College of Toxicology Annual Meeting P312 Application of Systems Pharmacology to Identify Exposure Response Markers in Peripheral Blood After Switching to a Candidate Modified Risk Tobacco Product: The Tobacco Heating System 2.1 (THS 2.1). F. Martin1, M. Talikka1, C. Haziza1, M. Peistch4, J. Hoeng5, J. Ancerewicz6. 1Philip Morris International, Neuchatel, Switzerland, Philip Morris International, Neuchatel, Switzerland 2 By analyzing the whole blood transcriptome of the study subjects and leveraging previously identified genes that exhibited a response to smoking in blood, we derived a classification tool using statistical learning methods. The classifier consisted of eight genes anddifferentiated users of THS 2.1 from smokers of CC with a specificity and sensitivity of 0.81 and 0.74, respectively. In conclusion, our systems pharmacology approach showed that the impact of switching to THS 2.1 can be detected in 5 days in whole blood transcriptome, thus providing a sensitive and noninvasive tool to assess exposure-response. These data alone do not substantiate reduced risk, and a range of further studies, including clinical studies, are being conducted on MRTPs. P313 MicroRNA Profiling of Hepatocellular Carcinomas in B6C3F1 Mice Treated with Ginkgo biloba Extract by Gavage for 2 Years. H. Yamashita1, A. Pandiri2, S. Bhusari3, K. Shockley4, S. Peddada4, K. Gerrish5, C. Rider3, M. Hoenerhoff3, R. Sills3. 1Taisho Pharmaceutical Co. Ltd., Saitama, Japan, 2Experimental Pathology Laboratories, Research Triangle Park, NC, USA, 3National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA, 4Biostatistics Branch, NIEHS, Research Triangle Park, NC, USA, 5Molecular Genomics Core Laboratory, NIEHS, Research Triangle Park, NC, USA Ginkgo biloba leaf extract (GBE) has been used for centuries in traditional Chinese medicine and today is used as an herbal supplement touted for improving brain function and for its antioxidant and anticancer effects. Exposure of B6C3F1 mice to GBE in the 2-year National Toxicology Program (NTP) bioassay resulted in a dose-dependent increase in hepatocellular carcinomas (HCC). We have previously reported increased Ctnnb1 mutations and alterations in Wnt/Ctnnb1 signaling in GBEinduced HCC compared to spontaneous HCC in vehicle controls. MicroRNAs (miRNAs) are small noncoding RNAs that are often dysregulated in various diseases including cancer. To identify key miRNAs that modulate GBE-induced hepatocarcinogenesis, we examined the global miRNA expression using Affymetrix GeneChip® miRNA 3.0 arrays and 2-pairwise analyses (n=5/group) comparing GBE-induced HCCs and spontaneous HCCs with vehicle control age-matched normal livers from B6C3F1 mice. We observed 16 and 3 unique differentially expressed miRNAs in GBE-induced HCC and spontaneous HCC, respectively. Ingenuity Pathway Analysis of the miRNA and mRNA array data from these tumors demonstrated altered molecular pathways associated with hepatocarcinogenesis, cell cycle progression, cell migration and cell proliferation. Additionally, miR-31, miR-145, miR-329 and miR-433, which were uniquely expressed in GBE-induced HCC, are known to regulate Wnt/Ctnnb1 signaling. Since the differentially altered miRNAs were unique to either GBE-induced HCC or spontaneous HCC, we plan to examine liver samples from the 90-day GBE study to see if these miRNAs could serve as potential biomarkers for GBE exposure or chemical-induced carcinogenesis. P314 28-day Toxicity Study of 3-monochloropropane-1,2-diol in CB6F1-nonTgrasH2 Mice. S.J. Park1, B.S. Lee1, E. Ju Jeong1, K.S. Moon1. Korea Institute of Toxicology, Daejeon, Republic of Korea1 3-monochloropropane-1,2-diol (3-MCPD) is a food processing contaminant that occurs in a wide range of foods. Several studies have evaluated the toxicological properties of 3-MCPD, but its carcinogenic potential has been controversial. In the present study, 28-day oral dose range finding in CB6F1-nonTgrasH2 mice was conducted to investigate a potential toxicity of 3-MCPD and also determine dose levels for following 26-week carcinogenicity study using TgrasH2 mice. The test article was administered once daily by oral gavage to male and female mice at dose levels of 0, 25, 50, and 100 mg/kg/day for 28 days. The standard toxicological evaluations were conducted during the in-life and postmortem phase. In mortality, three males and one female died during the study period, and toxic symptoms such as thin appearance and subdued behavior were observed with corresponding significant decreases of mean body weight and food consumption in the 100 mg/kg/day groups. Furthermore, relative and absolute weights of spleen, thymus, seminal vesicle and prostate in the 100 mg/kg/day groups were significantly lower than those of the control groups. Treatment-related microscopic findings were germ cell degeneration in testis (≥25 mg/kg/day), vacuolation in brain and sciatic nerve (≥50 mg/kg/day), basophilic tubule in kidney (100 mg/kg/day), and cardiomyopathy (100 mg/kg/day). These findings are in good agreement with previous studies. In conclusion, the target organ was determined to be brain, nerve, kidney, testis and heart, and the no observed adverse effect level (NOAEL) of 3-MCPD was considered to be 25 mg/kg/day or less in males, and 25 mg/kg/day in females. 77 ABSTRACTS Establishing exposure-response markers is imperative for the assessment of candidate modified risk tobacco products (MRTPs) against conventional combustible cigarettes (CC). Biomarkers derived from the primary site, such as the airway, require invasive sampling, whereas blood offers a noninvasive alternative for the general population. Various diseases and exposures, including cigarette smoke, have been shown to alter the molecular profile of the blood. To identify exposure-response relationships, we have conducted a whole genome Affymetrix microarray analyses from blood derived from a clinical study comparing smokers switching from CC to candidate MRTP THS 2.1, and from smokers who continued smoking their own CC for 5 consecutive days. Theopenlabel, randomized, controlled, 2-arm parallel group recruited 42 healthy smokers of both genders, aged between 23 and 65 years andwas conducted according to Good Clinical Practices (GCP) and is registered on ClinicalTrials.gov, 2014 th 35 American College of Toxicology Annual Meeting P315 Safety Assessment of Fruit Body Extract of Clitocybe nuda: Cytotoxicity and Genotoxicity Studies. M. Qiao1, T. Ren1, T.S. Huang1, J. Weese1, A. Gardner1, J.T. Chen2, J.W. Huang3. 1Department of Poultry Science, Auburn University, has not been previously documented in alcohol intoxication. He appears to have the highest reported BAC in a surviving pediatric patient. Hemodialysis, as recommended by poison control, was impracticable due to the presence of shock. The rapid drop in BAC, magnesium, and potassium levels may be due rapid volume expansion during fluid resuscitation rather than more rapid elimination of alcohol by an alternative pathway. Clitocybe nuda (C. nuda) is an edible mushroom which is promising to be developed either as novel natural food antimicrobials or as new functional food due to various bioactive components found in its fruit body extract recently; however, little or no toxicity information about its fruit body extract is available. In this study, the cytotoxic potential of its fruit body ethanol extract (freeze dried and heated dried) was investigated with both MTT assay and Neutral Red Uptake assay in embryonic fibroblast BALB/3T3 and human hepatoma (HepG2) cell lines. IC50 value was calculated based on the concentration-response curves ranged from 0.045 -100 mg/ml after 48 h exposure, and acute oral LD50 value was estimated from IC50 value obtained in 3T3 NRU assay based on the prediction model. Moreover, potential genotoxic effect of the extract was evaluated with two standard genotoxicity assays: in Ames assay, a set of Salmonella tester strains (TA98, TA100, TA1535, TA1537 and TA1538) were treated with fruit body extract aqueous sample with 5 different concentrations ranged from 50 to 5000 µg/plate, with and without S9 mix as an external enzymatic metabolizing system; in cytokinesis-blocked micronucleus cytome (CBMN-cyt) assay, number of micronucleus (MN) in binucleated HepG2 cells was examined after exposure to the fruit body extract within 0.045 -100 mg/mL in medium for 24 hours. Results suggested that C. nuda fruit body ethanol extract is nongenotoxic in vitro and with little cytotoxic effect. STP317 Protective Role of Sildenafil against Carbon TetrachlorideInduced Nephrotoxicity by Augmenting the Availability of Nitric Oxide and Antioxidant Enzymes. S. Goyal1, N. Verma1, S. Sharma1. School of Pharmacy and Auburn, AL, USA, 2Division of Plant Pathology, Taiwan Agricultural Research Institute, Taichung, Taiwan, China, 3Department of Plant Pathology, National Chunghsing University, Taichung, Taiwan, China ABSTRACTS 2014 P316 Arrhythmias and Electrolyte Abnormalities in Binge Alcohol Consumption: Supportive Management and Survival. H. Shakir1, T. The1. 1The Children’s Hospital at Saint Peter’s University Hospital, New Brunswick, NJ, USA, 2Drexel University College of Medicine, Philadelphia, PA, USA A 17 years old college student was admitted from our emergency room after binge drinking in a frag party with a blood alcohol level (BAC) of 815 mg/dL. He was comatose (GCS=3/15), hypothermic (rectal temperature 94.9 F), cyanotic with an oxygen saturation of 69% and a shallow breathing rate of 12/min. His heart rate was 73/min with a sinus rhythm. He was emergently intubated and mechanically ventilated. Upon rewarming, his blood pressure (BP) dropped to 65/28. Despite receiving multiple fluid boluses, increasing his maintenance intravenous infusion of Ringer Lactate to 200-300ml/hr, and increasing his Dopamine and Epinephrine intravenous infusion, his blood pressure remained labile. Whenever, we were able to raise his BP to normotensive levels, he developed massive diuresis and his BP dropped again. He developed severe hypokalemia, hypocalcemia, hypoalbuminemia, hypomagnesaemia and accelerated junctional rhythm with tall peak T waves. At about 10 hours postadmission, with the last magnesium sulfate bolus and his BAC dropping below 500 mg/dL, the accelerated junctional rhythm resolved and his blood pressure stabilized. Accelerated junctional rhythm Emerging Sciences, Baddi University of Emerging Sciences and Technology, Village- Makhnumajra, City- Baddi, State- Himachal Pradesh, India1 Introduction: Nephrotoxicity in experimental animal can be induced by CCl4¬¬ (Carbon tetrachloride), which is a colorless, volatile and nonflammable liquid of industrial use. Free radicals are generated due to the metabolic transformation of CCl4¬¬ that results in prominent changes in the morphology of kidney, including tubulointerstitial fibrosis and vascular congestion. The present study was designed to investigate the possible mechanism involved in protective effect of sildenafil (PDE5 inhibitor) in CCl4¬¬ induced nephrotoxicity. Material and Methods: Wistar albino rats of either sex (180-260g), n=6 were employed in the study. Nephrotoxicity was induced by administration of carbon tetrachloride (0.5 ml/kg, s.c.,) for 28 days. Serum creatinine, BUN, urinary microproteins, TBARS, nitrite/nitrate and reduced glutathione estimations were done as hallmarks of renal functioning. Results: Administration of CCl4 induced prominent changes in morphology of kidney, including tubulointerstitial fibrosis and vascular congestion, increases in serum creatinine, BUN, urinary microproteins, and renal tissue TBARS levels in comparison to normal control. It also decreased reduced glutathione and tissue nitrite/nitrate levels. Sildenafil treatment (0.4 and 0.8 mg/kg) antagonized the effect of CCl4 induced renal intoxication dose dependently. L-NAME (Nitric oxide synthase inhibitor) treatment significantly reversed the effect of Sildenafil treatment. Conclusion: Therefore, it may be concluded from the above findings that Sildenafil has a protective effect in prevention of renal injury by increasing the availability of nitric oxide and antioxidant enzymes. Impact statement: Based upon our evaluation, Sildenafil shows a promising result for the treatment of CCl4 induced Nephrotoxicity. STP318 Investigating Cellular and Molecular Pathogenesis of Hydrocarbon Oil-Induced Lung Hemorrhage and Inflammation. P. Prasad1, I. Valera1, M. Fishbein1, R. Raj Singh1. University of California (UCLA), Los Angeles, CA, USA1 Exposure to toxic substances (isocyanates/pesticides), recreational drugs (crack cocaine), pharmaceuticals, organ transplantation, and inflammatory diseases (vasculitis, lupus) may result in inflammation/hemorrhage in lungs, called diffuse alveolar hemorrhage (DAH). With no effective treatment, it is invariably fatal. Limited access to human tissue and/or 78 th 35 American College of Toxicology Annual Meeting IGP319 Occupational Exposure to FA: Genotoxicity, Immunotoxicity, and Susceptibility. S. Costa1, J. García-Léston3, S. Silva1, B. Laffon3, B. Porto4, J.P.s Teixeira1. 1National Institute of Health, Environmental Health Department, Porto, Portugal, 2ISPUP, Institute of Public Health, Porto, Portugal, 3University of A Coruña,Toxicology Unit, Department of Psychobiology, A Coruña, Spain, 4University of Porto, ICBAS, Institute of Biomedical Sciences Abel Salazar, Porto, Portugal Formaldehyde (FA) is a high-volume production chemical produced worldwide with a large range of industrial and medical uses. The highest level of human exposure to FA occurs in occupational settings. Eighty-five workers from anatomy pathology laboratories exposed to FA and 87 nonexposed controls took part in the study. Air monitoring was performed in worker’s breathing zone and the level of FA-exposure in air was estimated. Genotoxicity was evaluated in peripheral blood lymphocytes (PBLs) by means of cytogenetic alterations, DNA damage (comet assay) and T-cell receptor mutation assay. Percentages of different lymphocyte subpopulations were selected as immunotoxicity biomarkers. In addition, the effect of polymorphic genes of xenobiotic metabolising enzymes and DNA repair enzymes on the endpoints studied were determined. The mean level of FAexposure was 0.38±0.03 ppm. All cytogenetic endpoints and DNA damage evaluated by comet assay were significantly increased in PBLs of FA-exposed workers compared to controls. The exposed group also presented significant alterations in %cytotoxic T lymphocytes, NK cells and B lymphocytes in comparison with control group. Results from susceptibility biomarkers suggest that polymorphisms in CYP2E1, GSTP1 and GSTM1 are associated with increased genetic damage in FA-exposed subjects. Also FANCA significantly altered the level of genetic damage induced by FA exposure, revealing a potential novel repair pathway involved in the repair of genetic lesions caused by FA-occupational exposure. This work is supported by FCT under the grants SFRH/ BD/46929/2008 and PTDC/SAU-ESA/102367/2008. IGP320 Pesticide Residues in Strawberries. D. Dhakal1, R. R. Regmi Dhakal2, S. K. Raut2. 1Patan Multiple Campus, Institute of Science and Technology, Tribhuvan University, Lalitpur, Nepal, 2National Society of Toxicology Nepal, Lalitpur, Nepal The research work was carried out in Okharpauwa and Kakani VDCs of Nuwakot district, Nepal during 2013. Strawberries are becoming the single most important commodity in improving the living standards of local farmers. It instigates hybrids which demands relatively large amount of chemical fertilizers, and are susceptible to diseases & pests in this fruit. For the better control of those attacks, farmers were indiscriminately applied the pesticides on the area. Moreover, farmers has been using pesticides in their own way by means of bare hands to their farms, rather than adopting advised application procedures & safe handling methods. Six representative samples were taken from 20 different points. The samples were extracted in ethyl acetate and cleaned up by DSPE with PSA and pesticide residues analyzed by a gas chromatograph coupled with GC-EI-MS/MS. Final confirmation of the positive findings using two MRMtransitions and accurate quantification was performed by LC-MS/MS using a hybrid triple quadrupole linear ion trap mass spectrometer. Pesticideresidue has detected in 83.33% sample like Chlorothalonil, Dicofol, Endosulfan - α, Endosulfan - β, Parathionand 100% sample identified with Fenpropathrin. The T-Testsrevealed that not significant in the case of Dicofol and Fenpropathrin since p > 0.05. Moreover, 83.33% sample has observed with residue greater than the tolerance level in Chlorothalonil & Pyrathion, whereas only 33.33% of the samples were noticed with residue greater than the tolerance level in Dicofol but others residues were identified lower than the tolerance level of EU. 79 ABSTRACTS lack of animal models have prevented investigations into its pathogenesis, until recently, when investigators detected DAH in two otherwise normal mouse strains, C57BL/6 (B6) and C57BL/10, exposed to hydrocarbon oil [2,6,10,14-tetramethylpentadecane (TMPD)]. TMPD is a component of crude/mineral oils, laxatives, diesel exhaust, and cosmetics. The broad goal in this project is to investigate the role of immune cells and immune-response genes in the pathogenesis of DAH using the TMPD model. We have found that intraperitoneal administration of TMPD (0.5 ml) led to pneumonitis/vasculitis/hemorrhage in lungs of B6 mice within 2-4-weeks in 72% of 29 TMPD-exposed versus none of 23 controls (saline or control oil n-hexadecane). 50% of animals died within 3-4 weeks post-TMPD exposure. Flow cytometry data showed significant increase in plasmacytoid dendritic cells (pDC) in bone marrow (BM), and myeloid DCs (mDC) in lungs of TMPD exposed mice versus controls. To investigate whether TMPD triggers epigenetic changes in immune response genes, lungs from 3 groups of mice (saline, nhexadecane, TMPD) are being analyzed via microRNA and mRNA microarrays for differential gene expression. Future studies, will analyze whether elevated mDCs in lungs secrete pro-inflammatory cytokines locally, whereas pDCs in BM stimulate B1and marginal zone B cells, the first cell populations to encounter antigens acquired through peritoneum and blood. 2014 th 35 American College of Toxicology Annual Meeting 2014 400 Series—Toxicology Methods P400 Geometric Determinants of Full Thickness Wound Healing in Adult Male Yucatan Miniature Swine. D. White, C. Brandt, T. Madsen, C. Hanks, L. Brown, A. StrickerKrongrad, J. Liu, G. Bouchard. Sinclair Research Center, LLC, observations up to 1 hour post administration without the need for additional or continuous sedation. Auxvasse, MO, USA ABSTRACTS Objective: We assessed the relationships between wound healing rates vs. initial size and shape of full thickness wounds. Methods: Each of 3 minipigs had 9 full thickness paraspinous wounds created. The wounds were comprised of all combinations of 3 sizes (10, 20 and 30 cm2) with 3 shapes (square, circle and equilateral triangle - apex pointing to spine). During dressing changes, all wounds were photographed and planimetric measurements of total wound areas and areas of epithelialization were obtained. Time to complete healing was determined by clinical observation and photographic documentation. Linear healing rate and distance were determined from changes in planimetric measurements of area and perimeter using the following calculations: Results: Although larger wounds took longer to heal than smaller ones, there was no appreciable association with wound shape, e.g. average healing times for 10 cm2 wounds were 33, 35 and 33 days for circles, squares and triangles respectively. Absolute wound healing rates were calculated using planimetric data. There was a strong correlation between healing rate and initial wound area. Mean healing rates were 2.1, 2.6 and 3.3 cm2 per week for 10, 20 and 30 cm2 wounds, respectively. These differences did not appear to be affected by wound shape. Conclusions: Linear healing rates in adult Yucatan MS were largely unaffected by initial wound size or wound shape. This confirms the appropriateness of this test system for further preclinical wound healing studies. P401 Sublingual Dose Administration In Rabbits. K. Polhamus1, S. Blechinger1. MPI Research, Mattawan, MI, USA1 The rabbit is an increasingly in demand model for the assessment of mucosal irritation due to sublingually administered substances. The purpose of this study was to develop an effective technique for sublingual dose administration in rabbits, which minimized leakage, mimicked swallowing, and allowed for subsequent tissue assessments with limited repeat sedation. Animals were sedated with 0.5 mg/mL intramuscular injection of Dexdomitor® at a dosage of 0.25 mg/kg and placed into a mechanical head restraint device, which supported the upper jaw and head. The lower jaw and tongue were retracted and tinted tap water administered via syringe into the sublingual cavity at volumes up to 100 µL. Assessments of leakage were performed visually and followed by a saline rinse to mimic saliva and swallowing. Tissue examination was conducted at timed intervals from 0.5 to 6 hours post dose, to determine the need for additional sedation for appropriate assessment of mucosal irritation. The use of the head restraint and sedation regime allowed for the administration of up to 50 µL of liquid with minimal leakage and the assessment of timed P402 Characterization of T-dependent Antibody Response (TDAR) to Keyhole Limpet Hemocyanin (KLH) in the Göttingen Minipig®. V. Peachee1, M. Smith3, M. Beck2, D. Stump2, K. White Jr.1. 1ImmunoTox, A Division of AIBioTech, LLC, Richmond, VA, USA, 2 WIL Research, Ashland, OH, USA, 3ImmunoTox, Inc., Richmond, VA, USA Recently, there has been a renewed interest in the use of the minipig as an alternative to dogs and nonhuman primates for conducting toxicological assessments in nonrodent species. Since the T-dependent antibody response (TDAR) is one of the most widely-accepted assays used in the assessment of immunocompetence, the present study was undertaken to characterize the primary and secondary TDAR to keyhole limpet hemocyanin (KLH) in the Göttingen Minipig®. Following primary immunization with either 2 or 10 mg KLH, anti-swine IgM and IgG ELISAs were optimized and individual animal responses were evaluated over time. Immunization with 10 mg KLH on Day 0 promoted primary IgM responses that peaked 6 to 9 days after antigen administration, while primary IgG levels peaked on Days 13 or 14. Secondary IgG antibody levels (following secondary injection with 2 mg KLH on Day 14) plateaued on Days 20-22. Anti-KLH antibody levels were decreased in minipigs treated with cyclophosphamide (CPS), a known immunosuppressant, at doses ranging from 12.5 - 50 mg/kg/day, while antibody levels in animals treated with 2.5 mg CPS/kg/day were similar to levels in saline-treated swine. These results demonstrate that the Göttingen Minipig® can be a useful alternative nonrodent species to the dog and the nonhuman primate for evaluating the TDAR to KLH in regulatory assessments of immunotoxicity. P403 Spermatogenesis in the Göttingen Minipig: Assessing the Onset of Puberty. N. Navratil1, E. Taberner2, B. Jasmin1, M. Salerno1, B. Grambo1, G. Althouse2. 1Marshall BioResources, North Rose, NY, USA, University of Pennsylvania, Kennett Square, PA, USA 2 As the use of the Göttingen Minipig for toxicological studies increases, there is a need to define age of puberty. The purpose of this study was to evaluate the testis tissue from young Göttingen Minipigs to determine age at puberty, which was defined in this study as the majority presence of elongated spermatids in the seminiferous tubules/tubule lumen. Paired tissue samples from both testes were collected from 24 male minipigs ranging in age from 5-8 weeks (e.g., 6 boars assessed/ week of age). All animals were weaned at 4 weeks of age, and subsequently housed with age-matched males on a 12-hour light/dark cycle prior to tissue collection. These young males had no contact with mature breeding boars so as to avoid influence on their reproductive development. Tissues were harvested and immersed in Bouin's fixative, followed by histological sectioning 80 th 35 American College of Toxicology Annual Meeting and H&E staining. Microscopic examination was performed to determine number of each cell type (e.g., primary spermatocytes, round and elongated spermatids, sperm in lumen) in 100 seminiferous tubules. Elongated spermatids were predominantly observed in the tubules of 12 of the 24 males; this included 3 males at 6 weeks of age, 3 males at 7 weeks of age, and in all samples (N=6) collected from males at 8 weeks of age. A highly significant (p<0.01) correlation between boar age and presence of elongated spermatids was observed (rs = 0.78). Based on these results, puberty in male Göttingen Minipigs appears to begin at a younger age than previously cited in the literature. Chemotherapy-induced peripheral neuropathy (CIPN) is a progressive condition featuring pain, numbness, tingling, and/or muscle weakness often felt in the feet/hands. Despite an average incidence rate of 30-40% in the clinic, nonclinical toxicity studies often fail to detect CIPN. Thus, the aim of the present study was to adapt an established rodent CIPN model, vincristine-induced PN, into an assay that could be used to predict the occurrence of PN and applied to standard toxicity studies. Von Frey filaments were used to assess changes in mechanical sensitivity and testing was adapted according to the literature in order to make the assessment more amenable to incorporation into a toxicity study. Three baseline measurements were performed using filaments with bending forces of 4 g, 8 g, and 15 g applied to the plantar hind paw. Withdrawal responses were counted and expressed as an overall percent response. Following baseline, animals were administered vehicle or vincristine (50 or 100 µg/kg, i.p.) once daily for 10 days. Mechanical sensitivity was assessed again on days 7, 14, and 22. A time-dependent increase in mechanical sensitivity to all 3 filaments was observed in both vincristine treatment groups when compared to vehicle. These findings suggest that this assessment may be useful for the nonclinical detection of CIPN and could be amenable to use on toxicity studies. P405 Co-Culture Assay for the Identification and Investigation of Dermal Sensitizers (EPI-DC). G. DeGeorge1, M. Troese1, L. Pratt1, M. Piehl1, P. Hayden2, S. Ayehunie2. 1MB Research Laboratories, Spinnerstown, PA, USA, activation surface marker CD86+ >1.5-fold over vehicle controls, as measured by flow cytometry. Results: for IL-18, basal secretion in vehicle control co-cultures was 28.8 pg/ml. Treatment of Epi-DC with NBB yielded IL-18 concentrations that peaked at 0.2% NBB, resulting in a release of 166.4 pg/ml (12.9-fold increase). A dosedependent increase of CD86+ pDC was observed with treatment of NBB ranging from 0.05% to 0.2% NBB, peaking at a 2.9-fold increase in CD86 detection. At 1.0% Eugenol, increases of 1.7fold over control was observed in CD86 expression. Thus, in this initial evaluation, the Epi-DC co-culture model has been shown to be a useful and predictive tool for identifying dermal sensitizers without the use of animals. P406 A Highly Differentiated 3D Epidermal Skin Model to Characterize Skin Sensitizers in Mixtures. L. Pratt1, M. Troese1, G. DeGeorge1. MB Research Laboratories, Spinnerstown, PA, USA1 Strong support exists in the consumer products, pharmaceutical and cosmetics industries to develop in vitro assays to identify skin sensitizers. We have adapted the keratinocyte interleukin-18 (IL18) response assay developed by Corsini and colleagues (2009) for use with epiCS®, a highly-differentiated 3D epidermal model. The IL-18 assay measures release of this cytokine into the culture medium of treated tissues over 24 hours, by ELISA. Results are expressed as a Stimulation Index (SI) when compared to vehicle control treatment; an SI value of >1.5 was considered indicative of a positive sensitizer response. Four vehicles - Ethanol, AOO 4:1, Ethanol:DMSO 4:1, and Petrolatum - were compared for effects on three sensitizers (dinitrochlorobenzene, eugenol and citral). Ethanol:DMSO 4:1 yielded the highest SI values for the three sensitizers. The basal amount of IL-18 release for all vehicles ranged from 1.1 pg/ml to 17 pg/ml. DNCB consistently produced the highest SI values with the vehicles. In Ethanol, doseresponses were observed for DNCB, nitrobenzylbromide and p-phenylenediamine, while cinnamic alcohol and resorcinol were negative. Resorcinol in AOO 4:1 was clearly positive (SI = 2.7). The irritant/nonsensitizers phenol and glycerol were both negative (SIs < 1.5). Hair dyes ranging from light brown, dark brown and black colors were tested neat on the epiCS® tissues. An increase in IL-18 (> 71 pg/ml) over sham-treated controls (13 pg/ml) was observed in all dyes (SI values 5.7, 6.0 and 6.8). Further work is necessary to investigate different categories of chemicals and mixtures to improve the prediction model. MatTek Corporation, Ashland, MA, USA 2 To investigate the mechanism of dermal sensitization, a co-culture model of human keratinocytes and dendritic cells was developed and tested. This model (Epi-DC) consists of a 3D epidermal organotypic tissue, EpiDerm™, grown atop a suspension culture of plasmacytoid dendritic cells (pDC) in medium that allows for normal viability, expression of IL-18, and expression of activation marker CD86 on the surface of pDC. Test chemicals were dosed topically on the EpiDerm™ tissue for 24 hours. Criteria for change in biomarkers indicating a positive sensitizer included: (1) an increase of IL-18 release into the culture medium of >1.6-fold over vehicle, or (2) an increase in the percentage of pDC-bearing 81 ABSTRACTS P404 Development of a Nonclinical Assessment of ChemotherapyInduced Peripheral Neuropathy That Can Be Applied to Standard Rat Toxicity Studies. C. Tyszkiewicz1, A. Foote1, K. Cannon1. Pfizer Inc., Groton, CT, USA1 2014 th 35 American College of Toxicology Annual Meeting P407 Circulating miRNA-133a, miR-133b, and miR-1 as Potential Sensitive Biomarkers of Myotoxicity. J. Calvano1, A. Lo1, J. DiPiero2, B. Murphy1, C. Hixon1, R. Mangipudy1, M. Tirmenstein1. 1Bristol-Myers Squibb, New of thrombocytopenia for surviving animals ranged from 12-25 d (cyno) and 10-24 d (rhesus). ANC and PLT reached nadir for surviving animals 14-19 d and 14-16 d, respectively, for cynos and on 13-15 d and 14 d, respectively, for rhesus. Brunswick, NJ, USA, 2Bristol-Myers Squibb, Lawrenceville, NJ, USA ABSTRACTS 2014 Drug-induced myotoxicity is a significant concern in drug development. Plasma myotoxicity biomarkers including fatty acid binding protein-3 (FABP3), myosin light chain-3 (Myl3), and aspartate aminotransferase (AST), lack sensitivity and specificity, which can limit their utility as biomarkers of skeletal muscle toxicity. Published literature evaluating circulating miRNAs as biomarkers of skeletal muscle injury is limited. Male SpragueDawley rats received a single intraperitoneal injection of mitoxantrone (10 mg/kg), an antineoplastic anthracenedione, or vehicle. FABP3, Myl3, skeletal muscle troponin (sTNI), and AST were compared to miRNAs miR-133a, miR-133b, and miR-1 in serum. These miRNAs were identified as muscle-enriched by screening a tissue panel from untreated rats. Although no treatment-related histopathologic findings were observed in the soleus or quadricep muscle, heart, liver or kidney samples examined microscopically at 4, 24 or 48 hours after mitoxantrone administration, there were significant increases in mean sTNI in mitoxanthrone treated rats at all timepoints (4, 24, and 48 hours) examined. Other indices of skeletal muscle injury such as mean FABP3, MYL3, and AST were not significantly elevated in mitoxantrone treated rats. Serum levels of miRNA-133a, miR-133b, and miR-1 were increased 4 and 24 but not 48 hours postdose following mitoxantroneinduced skeletal-muscle injury. The amplitude of these increases was similar to that observed for the traditional sTNI biomarker of muscle injury at the 4 and 24 hour timepoints. The highest increases were noted with miR-133a and miR-133b. Our results suggest that muscle-enriched miRNAs are as sensitive as sTNI and may serve as an additional biomarker of myotoxicity. P408 Hematopoietic Characteristics of the Macaque (Rhesus vs Cynomolgus) following a Single Dose of Irradiation. R. Love1, N. Makori1, R. Manning1, S. Glaza1, T. Beck1, K. Fukuzaki1, R. Nagata2. 1SNBL USA, Ltd., Everett, WA, USA, 2Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Japan In continuing efforts to develop well defined animal models for the acute radiation syndrome under the FDA Animal Rule (21CR314, subpart 1), total body irradiation was applied to six cynomolgus (cyno) macaques given full supportive care at a dose of 7.55 Gy and sixteen rhesus macaques at a dose of 7.25 or 7.75 Gy using a bilateral 6 MV linear accelerator photon radiation source at 0.80 Gy min-1. Results: 50% of cynos (7.55 Gy) were euthanized by d 37 and 38% (7.25 Gy) to 50% (7.75 Gy) of rhesus were euthanized by d 19 post-irradiation. Study endpoints, in addition to survival, included cell nadirs and day/duration of neutropenia (ANC <500µL, ANC <100µL) and thrombocytopenia (PLT <20,000µL). ANC was <500 μL-1 8 d (cynos) or 6-7 d (rhesus) and <100 μL-1 10-12 d (cyno) or 9-12 d (rhesus) after irradiation. ANC of survivors recovered to ≥100 μL-1 by 22 d (cyno) or 19-21 d (rhesus) and to ANC ≥500 μL-1 by 22-25 d (cyno) or 21-24 d (rhesus). PLT count was <20,000 μL-1 12-25 d (cyno) or 10-11 d (rhesus) after irradiation. Duration Overall the response of the cynomolgus and rhesus macaques to a single application of irradiation were similar, suggesting the cynomolgus macaques may be a species utilized in future acute radiation syndrome studies. P409 Isolation of Granulocyte Colony Stimulating Factor (G-Csf) Mobilized Peripheral Blood Mononuclear Cells By Apheresis of Blood from the Rhesus Macaque. N. Lalayeva1, R. Love1, R. Rose1, N. Makori1, R. Manning1, S Glaza1, T. Beck1, K. Fukuzaki1, R. Nagata2. 1SNBL USA, Ltd, Everett, WA, USA, 2Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Japan Apheresis facilitates collection of stem cells or other blood components (such as platelets) in the peripheral blood that are subsequently used in the research of blood production and repopulation of blood producing cells for therapies such as for acute radiation syndrome and certain types of bone marrow cancers. Mobilization and apheresis of rhesus macaque blood, however, has proven to be technically challenging due to the inefficient mobilization response after in vivo G-CSF treatment and the high extracorporeal blood volumes required for harvest of these cells. In this study, one animal was administered Neupogen® at a dose of 50 µg/kg once daily for 5 days. Apheresis was performed on the 6th day using the AmicusTM Separator with MNC Collection (Fenwal Inc). Primary assessments of the collected blood included PBMC isolation, hematology and flow cytometry. White blood cells in the fresh apheresis product were higher than in the preapheresis product. The increase in lymphocytes, monocytes and basophils in the apheresis product was greater than the neutrophils. Flow cytometry data showed an upward trend in the absolute counts of CD45+ CD34+, CD45+ CD34high and CD45+ CD34high CD117+ hematopoietic stem cell populations following Neupogen® administration. Using the ACK lysis buffer procedure to remove red blood cells, 1.325 x 109 PBMCs were isolated from 8 mL of apheresis product vs 2 to 2.5x107 PBMCs recovered via a standard isolation procedure. These results confirmed appropriate apheresis and product collection methods were selected, thus establishing a reliable apheresis method in the rhesus macaque. P410 Evaluating Immunogenic Responses in Cynomolgus Monkeys with a Serum Biochemistry Analyzer. R. Early1, M. Chen1, W. Lv1, H. Pei1, S. McPherson1. Wuxi Apptec, Suzhou, China1 Immunogenic responses to immunoactive agents have historically been analyzed using ELISA methods and, whilst sensitive and precise, are often time-consuming and expensive. In the present study, responses in IgM, IgG, IgA, C3 and C4 to IM injections of keyhole limpet hemocyanin (KLH) in female cynomolgus monkeys (2 per treatment) were analyzed using a Hitachi 7180 Biochemistry Analyzer. The data was normalized to pretest values 82 th 35 American College of Toxicology Annual Meeting and compared to the control animals. Treatments consisted of a single IM injection of saline (controls); 5 mg/kg KLH (KLH Low); or 15 mg/kg KLH (KLH High). Serum was collected on Days -6, -2, 6, 8, 10, 12, 14, 16, 18, 20, 23 and 28 following the KLH injection. P411 Renal Safety Pharmacology: Characterization of a Non-Rodent Model with Water Loading Using a Positive Control Agent. S. Authier1, S. Abtout1, E. Troncy2. 1CiToxLAB, Laval, Quebec, Canada, 2University of Montreal, St-Hyacinthe, Quebec, Canada Introduction: Renal safety pharmacology is commonly performed in rodents but limited data is available for nonrodents. A canine model of renal safety pharmacology with water loading was characterized. Methods: Male beagle dogs (n=6) received furosemide (0.3, 3 and 10 mg/kg) or control in a crossover design with a 7 day wash-out. Water loading (30 ml/kg) and urinary bladder emptying were completed prior to each treatment. Each treatment session was divided into baseline (0-6 h and 6-24 h) and postdose collections (0-2 h, 2-4h, 4-6h and 6-24h). Biochemistry was evaluated at 6h and 24 hours post dose. Urine volume (ml/kg), specific gravity, plasma and urine biochemical parameters, water consumption and food consumption were assessed. Glomerular filtration rate, water clearance and excretion fraction were calculated to evaluate renal function. Results: Water loading increased urine output and glomerular filtration rate (0-6 h). Furosemide (0.5, 3 and 10 mg/kg) induced a dose dependent increase in urine output at 0-6 h. The 24h urine volume was moderately increased (+60.7%) with furosemide at 10 mg/kg compared to control. from 0-6 h, furosemide increased free water clearance, urinary excretion of sodium, calcium and chloride, decreased urine osmolarity and specific gravity. Water consumption increased and was proportional to urine output while food consumption decreased slightly at high dose (10 mg/ kg) only. Conclusion: Renal safety pharmacology in a canine model with water loading enabled identification of pharmacodynamic effects following administration of a diuretic. The canine model represents a valid alternative when this species is justified. P412 Practicalities of Dermal Dosing: The Practical Aspects of Dose Administration in Minipigs and Rats. T. Ramani1, L. Jun-Hsiang1, C. Savidge1, T. Jones1, C. Auletta1. Huntingdon Life Sciences, Somerset, NJ, USA1 Dermal administration to animal models presents multiple challenges. Ingenuity is needed to achieve dermal exposure that closely mimics the human exposure and provides an adequate margin of safety. Because of the limited area for exposure to test articles, it is often necessary to administer dermal formulations at the maximum feasible dose volume, which may differ for different materials and for different species. Procedures and calculations for determination of body surface area and the area to be covered by dermal formulations methods (10%) may vary from laboratory to laboratory. Comparisons and significance of the differences are discussed in this presentation. Procedures for open, occluded and semi-occluded administration of dermal formulations also vary from laboratory to laboratory. Advantages and disadvantages of collars, jackets and various wrapping materials/procedures are discussed and procedures currently in use at HLS are described. P413 Jacketed External Electroencephalographic (EEG) Telemetry Monitoring in Conscious Beagle Dogs and Cynomolgus Monkeys: Qualification of a Central Nervous System Safety Testing Model. S. Authier1, M. Pouliot1, L. Bassett1, R. Forster1, E. Troncy2. 1CiToxLAB, Laval, Quebec, Canada, 2University of Montreal, St-Hyacinthe, Canada Introduction: Implanted EEG monitoring by telemetry was reported in nonclinical studies but noninvasive with freely moving EEG recordings by telemetry as used in humans was not previously reported in nonrodents. Methods: A system for noninvasive continuous EEG-video monitoring by telemetry was qualified in dogs and nonhuman primates. EEG traces were obtained in Beagle dogs and cynomolgus monkeys using short electrophysiology needles in a Cz-Oz configuration from the 1020 system connected to a telemetry transmitter. An intravenous infusion (1.7 mg/kg/min) of pentylenetetrazol (PTZ) was used to characterize the model for seizure detection. Results: The incidence of signal artifacts was low (<5% of total EEG traces) and adequate for EEG interpretation. Non-invasive EEG monitoring was associated with a higher incidence of signal artifacts than surgically implanted EEG telemetry. The power of the EEG spectral bands was lower than values obtained from surgically implanted EEG leads but with comparable ratios between each band (delta, theta, alpha, sigma and beta). EEG traces showed normal profiles for the species evaluated with circadian changes. Upon PTZ infusion onset, paroxysmal EEG activity was observed with the expected trace morphologies (e.g. isolated sharp waves, repeated sharp waves, increased synchrony). PTZ induced ictal activity showed expected characteristics with a dominant frequency at Fast Fourier Transform (FFT) ranging from 3-6 Hz in dogs and monkeys. Conclusion: Noninvasive EEG monitoring by telemetry in freely moving animals was confirmed to be adequate to monitor normal, preictal and ictal CNS activity in conscious Beagle dogs and nonhuman primates. 83 ABSTRACTS A dose-dependent response in IgM was observed beginning on Day 6and was approximately 1.6-fold greater (zenith on Day 10) in the KLH Low animals and 2.6-fold greater (zenith on Day 8) in KLH High animals, compared to the normalized control data. Generally, there was no clear relationship to KLH injection in the IgG, IgA, C3 or C4 analytes. However, one KLH Low animal was noted for greater increases in IgA (1.3 fold greater on the Day 10 zenith), C3 (1.8 fold greater on the Day 14 zenith) and C4 (3.1 fold greater on the Day 16 zenith) relative to the controls and other KLH-treated animals. In conclusion, the samples were rapidly analyzed using the Biochemistry Analyzer (less than one day) and was considered advantageous for rapidly screening samples for immunogenic responses when compared to ELISA methods. 2014 th 35 American College of Toxicology Annual Meeting P414 Left Ventricular Pressure (LVP) Assessment Screening Models: Comparison of High Definition Telemetry in Free-Moving with Anesthetized Rats. S. Authier1, A. Ascah1, R. Forster1, L. Bassett1, E. Troncy2. 1CiToxLAB, Laval, Quebec, Canada, 2University of Montreal, ABSTRACTS St-Hyacinthe, Quebec, Canada Introduction: Nonclinical models to screen for cardiac contractility changes are used in drug development. Methodology: Left ventricular pressure (LVP) was measured in the conscious and anesthetized rats by direct catheterization of the LV apex. Results from conscious and anesthetized animals with the same positive control agents were compared. Results: Average dP/dt max observed during day time (7949 ± 890 mmHg/sec) and night time (9906 ± 1689 mmHg/sec) confirmed the circadian changes associated with normal activities. Pharmacological agents known for their contractility enhancer (Dopamine and Sotalol) or lessener (KCl) properties produced the expected effect on dP/dt max in both models. In the anesthetized model Sotalol (6 mg/kg, IV) and KCl (0.34 mEq/kg, IV) induced a reduction in dP/dt max of -23% and -21%, respectively compared to control. Dopamine induced a dose dependent increase in dP/dt max (+11% at 0.05 mg/kg and +34% at 0.1 mg/kg) in anesthetized rats. In the conscious rats, Sotalol (6 mg/kg, IV) and KCl (0.34 mEq/kg, IV) were also associated with a decrease in dP/dT max. Dopamine (IV) induced a dose dependent increase in dP/dT max (at 0.05 mg/kg and at 0.1 mg/mg) in conscious rats. Discussion: Our results suggest that the anesthetized rat model may be more sensitive than the conscious model to detect drug induced contractility changes. The conscious model enables repeated administration which is often required to increase sensitivity of the assay. Left ventricular pressure (LVP) monitoring in free-moving or anesthetized rats represent applicable methodologies for nonclinical safety evaluations in drug development. P415 Electroencephalography (EEG) in Sprague-Dawley Rats and Cynomolgus Monkeys: Superintervals to Increase Model Sensitivity. S. Authier1, L. Bassett1, M. Pouliot1, E. Troncy2, R. Forster1. 1CiToxLAB, Laval, Quebec, Canada, 2University of Montreal, St-Hyacinthe, Canada Introduction: Application of super-intervals to EEG data from rats and nonhuman primates was evaluated to enhance sensitivity in safety pharmacology. Methods: Cynomolgus monkeys and Sprague Dawley rats with EEG and EMG telemetry transmitters were used for EEG spectral analysis. Data variability was calculated for data bins of increasing durations from 1 to 480 min over 24-hour periods. The impact of data bin durations on intrinsic variability was assessed and potential differences for daytime and night-time data. Intrinsic EEG data variability was compared to cardiovascular and respiratory data. Results: In cynomolgus monkeys, increasing EEG data bins showed optimal variability reduction with a 60 min bin duration, with lower variability during night-time compared to daytime. In comparison, respiratory rate, tidal volume and heart rate in cynomolgus monkeys showed progressive improvements up to an interval duration of 480 min.. In Sprague Dawley rats, EEG data bins also showed optimal variability reduction at 60 min 2014 without significant differences between day and night time. In comparison, optimal data bin durations for respiratory rate and tidal volume in rats was observed with bin duration of 5 min, with progressive improvements up to an interval duration of 480 min for heart rate.. Discussion: Super-intervals decrease variability for respiratory, cardiovascular and EEG data. The benefit of increasing in bin duration should be weighed against the potential dilution of drug effects with unaltered data. The current analysis supports a bin duration of 60 min for EEG data in rats and nonhuman primates when pharmacologically relevant. P416 Melanin Location in the Skin and Hair Appendages in the Long Evans Rats: Relevance to Photosafety Evaluations. M. Dougherty1, J. Baker2, M. Elliott2, T. Albers3, D. Learn1. 1Charles River Laboratories Preclinical Services, Horsham, PA, USA, 2Pathology Associates, Inc, Frederick, MD, USA, 3Charles River Laboratories, Wilmington, MA, USA The role of melanin in xenobiotic-induced ultraviolet radiation (UVR)-induced phototoxicity is not clear. Melanin produced in response to UVR exposure is classically considered a protective adaptation to this exposure. There are references to melaninrelated amplification of phototoxicity, primarily using in vitro models that has generated regulatory concern, but evidence of this effect in vivo has not been reported. The ICH S10 Photosafety Evaluation of Pharmaceuticals also recognizes that melanin binding can increase local tissue concentration of xenobiotics but that binding alone does not present a photosafety concern. The Long Evans (LE) rat is the primary in vivo phototoxicity Test System used in this Testing Facility to evaluate possible melaninbinding enhancement of phototoxicity, as both lightly and darkly pigmented skin sites can be evaluated. Questions have been raised regarding the anatomic location of the melanin visibly observed in the skin of the LE rat. Should melanin not be present in the skin but only in the hair appendages, and presuming that binding could exacerbate phototoxicity, the relevance of the model to humans could be questioned. Histopathological evaluation of lightly and darkly pigmented skin revealed melanocytes located at the dermal/epidermal junction in the darkly pigmented skin, but absent in the lightly pigmented skin. The darkly pigmented skin was less sensitive to UVR-induced erythema and edema, demonstrating that the constitutive pigmentation, regardless of location, was, as expected, protective. These results indicate that the LE rat is a relevant model for potential melaninenhanced phototoxicity. 84 th 35 American College of Toxicology Annual Meeting P417 Modeling of Flow and Evolving Aerosol Particles Dynamics for Computing Local Deposition in Air-Liquid Interface Experiments. A. Kuczaj1, M. Nordlund1, S. Majeed1, S. Frentzel1, J. Hoeng1, M. Peitsch1. 1Philip Morris International R&D, Philip Morris Products SA, Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland, 2Multiscale Modeling and Simulation, Faculty EEMCS, University of Twente, AE Enschede, The Netherlands Deposition of aerosol is governed by the various physical mechanisms (e.g., diffusion, impaction, sedimentation), and implicitly is also dependent on the flow and surrounding aerosol particles' thermal conditions. This is in particular important when considering that aerosols consisting of liquid droplets are being diluted with air as they may still evolve. Using Computational Fluid Dynamics, a computational model of the well was constructed, in which we considered flow with embedded droplets characterized by its mean size droplet diameter. The simulations took in account fully coupled equations for mass, momentum and energy in the Euler-Euler framework as well as one-way coupling of gas and liquid phase. For the account of the sedimentation the driftflux approach was used. The numerical investigations indicated that the flow rate, droplet diameter, diffusion and gravitational settling play significant roles in the process of aerosol deposition. Simultaneously the influence of impaction was found to be negligible at considered droplet diameters. P418 Qualification and Comparison of Big Blue® Mouse and Rat Transgenic Rodent (TGR) Mutation Assays with N-Ethyl-NNitrosourea (ENU) and Benzo(a)pyrene (BaP). R. Young1, H. Dinesdurage1, M. McKeon1, R. Elbekai1, D. Bruning1, T. Lawlor1, M. Aardema2. 1BioReliance, Rockville, MD, USA, 2Marilyn Aardema Consulting, LLC, Fairfield, OH, USA Background mutant frequency in each tissue was similar in mice and rats. Liver background mutant frequency was 43.6±6.2x10-6 in mice and 43.4±8.8x10-6 in rats. Bone marrow background mutant frequency was 38.1±14.2x10-6 in mice and 29.7±14.3x10-6 in rats. In mice, significant (p<0.001) increases in mutant frequencies were observed with BaP in liver (4.9-fold) and bone marrow (17.7fold) and ENU in liver (4.2-fold) and bone marrow (10.6-fold). In rats, significant (p<0.001) increases in mutant frequencies were observed with BaP in liver (5.8-fold) and bone marrow (9.6-fold) and ENU in liver (3.6-fold) and bone marrow (6.8-fold). Both Big Blue® mouse and rat systems are robust with the ability to detect significant increases in mutagenicity in slow dividing (liver) and fast dividing (bone marrow) tissues by a direct acting mutagen and one that requires metabolic activation. P419 Methods for Successful Intra-Vaginal Dosing in Rats RM. R.M. Gendron1, P. Mansell1. Charles River Laboratories Preclinical Services Montreal, Montreal, Quebec, Canada1 With an increase in the development of new drugs specifically targeting female patients, including hormone replacement therapies, contraceptives, anti-infectives and infertility therapies, various forms of intra-vaginal administration (creams, gels, suppositories and rings) have been developed for local and/or systemic use. The intra-vaginal route presents advantages for women, including a reduced need for daily administration (e.g. the vaginal ring), user controlled administration, discreteness, single treatments lasting for weeks or months, and treatment regimens tailored to the individual. These advantages result in increased compliance and a decrease in potential side-effects. Additionally, the use of the vaginal route for drug delivery avoids first-pass effects and provides optimum pharmacokinetic profiles. With this focus on intra-vaginal delivery, there is also a need to successfully and efficiently perform intra-vaginal dose administration in nonclinical studies. Our laboratory has established procedures required for intra-vaginal dosing of different formulations of controlled-released drugs in Sprague Dawley rats. Due to the limited surface area within the female reproductive tract of rats, it was determined that the procedures and/or frequency of application may need to be varied depending on the formulation, dose level of the material to be delivered, and body weight of the rat. It was also determined that for a successful study, procedures were required to minimize loss of test material. In conclusion, our laboratory has established procedures for intra-vaginal dosing in Sprague Dawley rats that allows for accurate assessment of local and systemic responses to drug exposure and allows for standard observations to be evaluated in toxicology studies. In vivo transgenic rodent (TGR) mutations assays are used to follow up genotoxicity results with a suspected mutagenic mode of action. One advantage of the Big Blue® model compared to other TGR mutation models is that both mouse (C57BL/6 or B6C3F1) and rat (Fisher 344) models are available. We qualified both species to new OECD TG 488 standards and to permit comparisons between the species. Homozygous male Big Blue® C57BL/6 mice and F344 rats received olive oil vehicle (5 mL/kg/ day) or BaP (50 mg/kg/day) for 28 days or ENU (40 mg/kg/day for mice or 20 mg/kg/day for rats on Days 1-3) with necropsy on Day 31. Mutations in liver and bone marrow were evaluated. 85 ABSTRACTS Understanding physical conditions that govern deposition of aerosol droplets and its influence on the cell functioning is a key step towards the ultimate goal to relate the exposure of inhaled and deposited aerosols to health outcomes. This is important twofold, i.e., the same physical mechanisms are acting in much more complex geometries (e.g., human airways), and simultaneously acquired knowledge allows for improved in vitro inhalation toxicology experiments. We present here our approach to model the flow and evolving aerosol dynamics together with aerosol deposition in a simple trumpet-like geometry frequently used in in vitro exposure systems (e.g., Vitrocell®). 2014 th 35 American College of Toxicology Annual Meeting P420 Intrathecal Administration and Cerebrospinal Fluid Collection in the Juvenile Beagle Dog. J. Douville1, F. Emond1, R. St-Jacques1, F. Claudia1, B. Lise1. Charles River Laboratories, Preclinical Services Montreal, ABSTRACTS Senneville, Quebec, Canada1 Delivery of drugs directly into the cerebrospinal space is an effective means to treat neurological diseases when systemic delivery fails to reach the target or alleviate the symptoms. Neurological disorders often appear or develop during childhood and there is an increased interest in conducting nonclinical studies in juvenile animals to mimic the age/life phase of the target population. Additionally, these studies oftentimes require collection of cerebrospinal fluid (CSF) to evaluate central nervous system (CNS) drug concentrations or disease-specific biomarkers. At this early growth stage, morphology of the weanling dog presents particular challenges requiring refinement of our surgical techniques. Also, our established approach for CSF collection via the lateral ventricle in adult dogs necessitates modification, including determination of stereotaxic coordinates and cranial landmarks in juvenile dogs. Therefore, we investigated suitable methods for accurate delivery of potential therapeutics to the CNS and CSF collection in 9 to 11 week old dogs. An intrathecal catheter was inserted via a microlaminectomy of the L5 vertebra and a durotomy. Once the catheter was secured, a subcutaneous pocket was made in the lumbar region, and the access port was fixed to the muscle; the catheter was then fixed to the access port. Different approaches were evaluated for CSF collection, including direct puncture or catheter implantation into the cisterna magna and implantation of a guide port for repeated sampling from the lateral ventricle of the brain. Herein we describe the success rate of our optimized approaches for both intrathecal delivery and CSF collection in juvenile dogs. P421 Comparative Evaluation of Background Data for the Incidence of Foot Lesions In Sprague-Dawley Rats Housed in Different Types of Caging. S. Kwok1, P. Mansell1. Charles River Laboratories Preclinical Services, Montreal, Senneville, Quebec, Canada1 Use of polycarbonate (solid-bottom) cages containing bedding for rodents has been previously shown to provide improved housing conditions with particular emphasis on a reduction in the incidence of foot lesions known to occur in rats during the conduct of chronic toxicology and carcinogenicity studies. The background incidence of foot lesions was therefore compared between metal wire and solid-bottom housed animals for carcinogenicity studies conducted in our facilities with ad lib fed Sprague Dawley Crl:CD(SD) rats. Data from 104 week carcinogenicity studies performed in solid-bottom cages and conventional metal (stainless-steel wire mesh or perforated floor) cages was evaluated to investigate the relationship between housing regimens, body weight as well as to compare the incidence and onset of foot lesions. Characteristic clinical signs associated with foot lesions considered in this comparative assessment included limited hind limb usage, skin lesions, skin papules, skin redness, skin scabs, and firm/soft swelling of the hind limbs and/or hind paws. As anticipated, there was general 2014 reduction (0.02X to 0.6X) in the incidence and a delayed onset of clinical signs associated with foot lesions in rats housed in solidbottom cages, when compared to rats housed in metal cages. Incidence was shown to be positively correlated to bodyweight, with higher occurrence and earlier onset of lesions being noted in the heavier male population when compared to females. Data from this laboratory was generally comparable to existing literature and further supports use of solid bottomed cages for chronic rodent studies. P422 Long-Term Intrathecal Infusion/Sampling Via a Surgically Implanted Access Port with Functional Observation Battery Observation in Cynomolgus Monkeys. R. Tavcar1, S. Authier1, M. Said Meghazzi1, S. Groom1, R. Forster2. 1CiToxLAB North America, Laval, Quebec, Canada, 2CiToxLAB France, Évreux, France Intrathecal administration and sampling continues to be critical in nonclinical pharmaceutical development for drugs that must be delivered across the blood-brain barrier or to allow sampling of cerebral spinal fluid (CSF). In alignment with the 3Rs we have refined a method to allow long term administration and/or sampling. In addition, a modified function observation battery (FOB) often conducted as part of the ICH S7A safety pharmacology core battery, was developed for the evaluation of neurological changes in nonhuman primates. A cerebrospinal fluid catheter was inserted into the intrathecal space in the vertebral lamina (L3) and connected to a vascular access port. Implanted animals were first used for the evaluation of the cerebrospinal pharmacokinetics of a pharmaceutical, with CSF sampling over a period of 36 hours followed by a washout period and ending with the 28 day continuous infusion feasibility Phase. Following the 28 day period, the animals were euthanized for histopathological evaluation. No neurological abnormalities were observed and minimum to mild catheter-associated chronic granulomatous inflammation was associated with the catheter in the subarachnoid space at the injection/sampling site, next to the spinal cord and spinal nerve roots. Similar moderate catheterassociated chronic granulomatous inflammation was also noted along the catheter track beside the vertebral body and dorsal spinal process below the lumbar muscles. In conclusion, the port/ catheters were patent for the duration of the experiments (over 100 days in total). Therefore the improved method was deemed to be appropriate for long term toxicological, neurological and pharmacokinetic evaluations in Cynomolgus monkeys. P423 Repeated Intratracheal Powder Aerosol Delivery In SpragueDawley Rats. M. Stoute1, A. Dumais1, S. Groom1, R. Tavcar1, R. Forster2. 1CiToxLAB North America, Laval, Quebec, Canada, 2CiToxLAB France, Évreux, France Inhalation administration is a highly effective delivery route allowing the bypass of first-pass metabolism and permitting local delivery to the lungs but is expensive and requires large quantities of material. We evaluated an alternative approach using a novel device for repeated intratracheal administration 86 th 35 American College of Toxicology Annual Meeting P424 Human Whole Blood In Vitro Cytokine Release—How Does Variability Impact Study Design? C. Dumont1, R. Bourgeois1, K. Bryant1, E. Marcotte1, S. Lavallée1, M.S. Christin-Piché1. Charles River, Senneville, Quebec, Canada1 Interpretation of in vitro cytokine release assays (CRA) can be challenging. Appropriate study design and knowing the expected variability are critical in a successful outcome. In this study, we focused on the comparability of the response of 9 cytokines following in vitro stimulation of human whole blood with an anti-human CD3 (24 hours). Blood from up to 35 donors was stimulated by soluble and dry-coated methods and data were evaluated for inter-donor variability. The data collected over different occasions were used to assess donor reproducibility. Globally, the inter-donor variability was high and inter-day variability was observed. However, the donors with the higher or lower levels remained generally consistent between occasions. Both stimulation formats showed similar inter-donor and interday variability. Detectable levels of cytokines were measured in both assay format but differed significantly for some cytokines. IL-1β was released at least 1.5X more using the soluble format while IL-6, IL-8, IL-10 and IFNg were released at least 1.9X more using the dry-coated format. The levels of IL-2, IL-12, TNF-a and G-CSF were not significantly altered by the stimulation format. In conclusion, based on the variability observed in CRA data, it is not recommended to use less than 8 donors. The interpretation must be conducted with caution as a positive or negative response in this type of assay might not be representative of the response obtained with all donors. Lastly, the choice of the cytokines analyzed and the mode of action of the drug must be taken into consideration when choosing the assay format. P425 Comparative Photomicrographic Examination of Integument from Eight Species of Mammals Including Two Lineages of Research Miniswine. L. Brown1, D. Kim3, C. Hanks1, S. Schnapp1, D. Brocksmith2, D. White1, A. Stricker-Krongrad1, J. Liu1, G. Bouchard1. 1Sinclair Research Center LLC, Auxvasse, MO, USA, 2Sinclair BioResources LLC, Auxvasse, MO, USA, 3Veterinary Medical Diagnostic Laboratory, Columbia, MO, USA Introduction: Skin is the largest organ in the body. Animals have skin which is generally similar to human skin, however, species specific anatomical and biochemical differences exist. The integument of animal models may vary in skin surface topography, overall thickness and thickness of specific layers, stratum corneum, epidermis, dermis density and collagen content, regional blood flow, pelage (hair count), hair follicle size or density, and sub-dermal characteristics. Determination of which animal model most closely matches the skin of humans is important for translational dermal research. Objective/Rationale: Prepare magnified images of comparative skin histology and perform simple image analysis for differences or similarities. Methods: Animal skin samples collected included Yucatan miniswine, Hanford miniswine, Cynomolgus monkey, Beagle dog, NZW rabbit, Hartley guinea pig, Sprague-Dawley rat, and CD-1 mouse. The human skin image was acquired from a medical school. Samples were fixed in neutral buffered formalin, processed, sectioned, stained with H&E, examined by microscope and photographed by veterinary dermatopathologist (DYK). The resulting skin images generated by Olympus MicroSuiteTM were compared side-by-side at equivalent magnification. Results: Visual comparison of images suggest the skin of swine and human look the most similar while the skin of rodents (rat, mouse) has a much thinner epidermis. The skin of rabbit and guinea pig appear to consist predominantly of hair shafts/hair follicles combined with thin epidermis. Monkey and dog skin were next most similar to the human skin. Conclusion(s): The swine (Yucatan & Hanford) skin images are visually most like those of the human images. 87 ABSTRACTS (IT Rx). We performed single dose and 7 day repeat dose studies with administration of air, PLGA (50/50 DL-lactide/glycolide copolymer) (10 mg/day) or a pharmaceutical test item (10 mg/ day) to groups of 6 female Sprague Dawley rats. Only the results of the air and PLGA control groups are presented. Repeated IT Rx resulted in procedure-related lung changes in both control groups, including perivascular/peribronchiolar infiltrate, increased alveolar macrophages, and alveolar hemorrhage. These changes were seen with higher incidence and/or severity in rats receiving PLGA. Lung changes attributable to PLGA, but not seen in the air control, included bronchioloalveolar inflammation, bronchiolar epithelium hypertrophy/hyperplasia and/or erosion, exudate/cellular debris in bronchioloalveoar lumens, foreign material, foreign body granuloma, and necrosis with or without trachea epithelium hypertrophy/hyperplasia and inflammation. Although repeated IT Rx of dry powder aerosols was feasible with this novel device, the lung mechanical damage and dose delivery variation represented significant limitations. This approach was not considered ideal for a 28-day regulatory study with the pharmaceutical test item because of these limitations. Repeated IT Rx of a dry powder aerosol in this way can be feasible and cost effective alternative approach to inhalation exposure during early discovery allowing for preliminary toxicological and pharmacokinetic evaluation before committing to longer term inhalation studies. 2014 th 35 American College of Toxicology Annual Meeting P426 Application of a DNA Adductomics Software Platform to the Investigation of DNA Damage in a Bacterium. R. Kanaly1, A. Maeda1, R. Micheletto1. Yokohama City University, P428 Comparison of Selected Clinical Pathology Parameters from Sprague-Dawley Rats Sampled via Abdominal Aorta versus Jugular Vein. B. Attalla1, P. Mansell1. Charles River Laboratories, PreClinical Yokohama, Japan1 ABSTRACTS 2014 DNA adductomics is an emerging field of toxicology that was conceived of as a comprehensive top down approach to detect DNA modifications in human tissues by utilizing liquid chromatography coupled to electrospray ionization tandem mass spectrometry (LC/ESI-MS/MS). A triple quadrupole MS is used to detect the neutral loss of 2'-deoxyribose from positively ionized modified 2'-deoxynucleosides by utilizing single reaction monitoring mode transmitting the [M + H]+ > [M + H - 116]+ transition over a range of multiple predefined transitions. A software platform to aid in the processing of DNA adductomics data was written in Python and utilizes a UNIX/Linux-compatible environment via a graphical user interface. This software was applied in an investigation of DNA damage in the bacterium Serratia marcescens when it was grown under different conditions. A total of 250 SRM transitions were monitored and all data were merged and converted into bubble-type chart adductome maps that allowed for direct visual sample comparisons. The software platform also enabled the creation of transition-specific data plots that may be extracted from the large data sets. The utility of this software as it is applied to DNA adductome analyses in the context of bacterial DNA shall be presented. P427 QT Interval Prolongation and ECG Restitution Assessment Using Beat-to-Beat Method in the Minipig. R. Brandon Borders1, K. Kearney1, B. Roche1, A. Fossa2. 1WIL Research, Ashland, OH, USA, 2iCardiac, Brighton, NY, USA The FDA has rejected many potential pharmaceutical agents due to QTc prolongation as a surrogate marker for potential risk of fatal arrhythmias but assessing arrhythmogenic risk based solely on QTc prolongation is an inherently flawed decision in the drug safety and development paradigm (Hondeghem et al 2008, Ranjan et al 2013). Though certain types of fatal arrhythmias share a positive correlation to the delayed repolarization period, QTc cannot discriminate the importance of changes in autonomic state or define reference bounds for normal physiological states. Fossa (et al 2006, et al 2010) has shown that the QT instability of the QT-RR and QT-TQ interval (restitution) relationship assessed on a beat-to-beat method is an important and often misunderstood factor in arrhythmogenic risk analysis. Using beat-to-beat analysis derived from ECG data is proving to be a more pragmatic screen because: 1.) the technique discriminates proarrhythmic vs. safe drugs despite QT prolongation because: it considers the changes in QT interval in relationship to normal physiological reference bounds, 2) it can examine beat-to-beat restitution (hearts ability to recover from one beat to the next) quantifying the risk of the QT-TQ heterogeneity leading to re-entrant arrhythmias. In order to use restitution data as bridge between nonclinical and clinical risk analysis, it is important to define the QT-TQ variability in each of the common species used in drug development. The purpose of this study was to observe changes in QT/TQ and RR restitution relationship of mini-pigs during control and after exposure to a QT prolonging drug. Services, Montreal, Quebec, Canada1 Blood sampling for routine clinical pathology assessments in rats can be performed using different sites and approaches, many of which require sampling at termination. Refinement of blood sample collection via direct jugular puncture has presented an opportunity for nonterminal sample collection for these assessments. This can allow for monitoring of clinical pathology parameters over the course of the study as well as at termination. A comparison of standard clinical chemistry, hematology and coagulation parameters for samples from Sprague Dawley rats where blood was drawn via jugular or abdominal aorta was done to confirm that values obtained by either collection methods are comparable. The rats used for the comparison were between 10 and 20 weeks old. Males and females were compared separately. The data was gathered from studies where rats were group housed, up to three animals per cage and fasted prior to the blood collection procedure. There were no meaningful differences on the hematology, clinical chemistry, or coagulation parameters from fasted Sprague Dawley rats between 10 and 20 weeks old, when sampled from the jugular vein or abdominal aorta. In conclusion, both methods are considered suitable and the resultant data can be appropriately compared within the same study or program. P429 Optimization of T-Cell Dependant Antibody Response (TDAR) Conditions for Juvenile Rat Toxicity Studies. R. T. Laureano1, N. Collins1, J. Piccotti2, L. Morford3, S. Rochelle Mikkelsen4. 1Celgene Corporation, Summit, NJ, USA, 2Celgene Corporation, San Diego, CA, USA, 3WIL Research, Ashland, OH, USA, Burlenson Research Technologies, Morrisville, NC, USA 4 For pediatric therapeutics where there is cause for concern for the developing immune system, it is recommended that immunotoxicity endpoints such as TDAR be added to the juvenile study. However, there is little information on the appropriate KLH dose for juvenile animals or the ability of immunosuppressive therapeutics to quantifiably suppress the TDAR. To determine the optimal KLH concentration for eliciting a TDAR in juvenile rats and compare the response to adult rats, rats (n=8/sex/group) were administered 0.1 or 0.3 mg KLH (IV) on PND 42 (juveniles) or PND 70 (adults) and blood samples collected 6 and 21 days postsensitization for anti-KLH IgM and IgG titers, respectively. To investigate whether a immunosuppressive agent would suppress a TDAR in juvenile animals, cyclophosphamide (8mg/kg/day) was administered (IP) on PND 35-49 (juveniles) or PND 63-77 (adults). There were no remarkable differences in juvenile TDAR between the 0.1 and 0.3 mg KLH doses. A statistically significant increase in anti-KLH IgM production in adults compared to juveniles was observed. Cyclophosphamide suppressed antiKLH antibody responses at both KLH dosages, except for the IgG response in juvenile males, and adult females immunized with 0.3 mg of KLH. These results indicate that a KLH dose of 0.1 mg could elicit a robust TDAR response in juvenile rats and the response can be quantifiably suppressed with an appropriate 88 th 35 American College of Toxicology Annual Meeting dose of an immunosuppressive therapeutic. Anti-KLH IgM were higher in adults. There was no difference in anti-KLH IgG response between juveniles and adults; however, female titers were generally higher than males. P430 Comparative Specular Microscopy in Laboratory Animals (Rabbits, Dogs, and Nonhuman Primates). S. Wise1, Mark Vézina1. Charles River Laboratories Preclinical Services Montreal, Senneville, Quebec, Canada1 P431 Assessment of Maximum Volume Injected Intramuscularly to Rats. M. Felx1, A. Varela1, S. Haile1, S. Y. Smith1. Charles River Laboratories, Senneville, Quebec, Canada1 injecting ≤0.10 mL of Indian ink, the ink remained within rectus femoris muscle with minimal leakage into the subcutaneous tissue and some spread to surrounding muscles. Injection of ≥0.20 mL indicated that ink spread to the surrounding muscles with increased leakage into the subcutaneous tissue. Based on Faxitron radiographs of contrasting agent, gross examination of the spread of injected ink, 0.10 mL was determined to be the optimal volume that could be injected into the muscle with minimal spread to the neighboring muscles and limited leakage into the subcutaneous tissue. P432 Flow Cytometry in Combination with Benchmark Dose Software is a Powerful Tool for Evaluating In Vitro and In Vivo Genotoxic Dose-Responses. S. Bryce1, J. Bemis1, D. Torous1, S. Avlasevich1, S. Phonetapswath1, J. MacGregor2, S. Dertinger1. 1Litron Laboratories, Rochester, NY, USA, 2Toxicology Consulting Services, Bonita Springs, FL, USA Flow cytometry-based micronucleus (MN) scoring (In Vitro and In Vivo MicroFlow®) is an efficient means of generating large datasets that allow the study of dose-response relationships. We propose that the utility of such data can be maximized by applying Point of Departure-type analyses as provided by Benchmark Dose (BMD) software (e.g., PROAST). For proof-of-concept, we compiled high quality flow cytometric in vivo MN data for 7 diverse clastogens that were evaluated in male rats exposed for 3 days with up to 9 dose levels of melphalan, chlorambucil, thiotepa, 1,3-propane sultone, hydroxyurea, azathioprine, or methyl methanesulfonate. These same chemicals were studied for in vitro MN responses by treating TK6 cells with up to 20 concentrations in quadruplicate. In vitro MN frequencies were determined via flow cytometric analysis using a 96 well plate autosampler. In vivo and in vitro MN frequencies were evaluated with PROAST, and the dose corresponding to a doubling of baseline MN frequency (BMD100) was calculated. The resulting analyses rank-ordered the in vivo and in vitro potency of these chemicals, with melphalan demonstrating the lowest BMD100 values and hydroxyurea and 1,3-propane sultone the highest. For these systemically available chemicals, a direct relationship between in vitro and in vivo BMD100 values was observed (R2=0.839). These results suggest that flow cytometric scoring in combination with quantitative approaches for dose-response modeling can be useful for risk assessment and prioritization. The objective of this experiment was to determine the maximum volume that could be injected intramuscularly into the rectus femoris of albino rats. To female albino rats, different volumes of Indian ink and a contrasting agent (OmnipaqueTM) were injected into the rectus femoris. The rats were 8 to 9 weeks at the time of injection and weighed between 200 to 250g. Faxitron radiographs were performed immediately post, 15 minutes post and 5 hours post injection. At the location of the Indian ink injection, the skin, subcutis and muscle were opened and the spread of the ink within the muscle and the extent of leakage were evaluated. Faxitron radiographs indicated that a ≤0.10 mL injection of OmnipaqueTM was localized at the muscle area with minimal or no leakage. Injections of ≥0.20 mL resulted in leakage outside of the muscle and showed distal expansion cranially along the patella and patellar ligament. Gross observations showed that when 89 ABSTRACTS Specular microscopy is a noninvasive imaging technique that allows visualization and morphological analysis of the corneal endothelium (EC) layer, a monolayer of specialized cells covering the posterior surface of the cornea that maintains the health and transparency of the corneal stroma. Clinical instruments can be used to monitor longitudinal in-life changes during nonclinical toxicology studies following administration of drugs to the eye. Our laboratory has used the noncontact Nidek CEM-530 specular microscope to compare baseline cell density, pleomorphism (cell shape), and central corneal thickness values in New Zealand White rabbits, beagle dogs, and cynomolgus monkeys (NHP). Rabbits, having the thinnest corneas, had the greatest EC density (2968 ± 235 cells/mm2, n=20 eyes) and highest percentage of hexagonal cells (74% ± 2) of the three species. Conversely dogs, with the thickest corneas, had the lowest EC density (2540 ± 105 cells/ mm2, n=20 eyes) and lowest percentage of hexagonal cells (66% ± 3). EC density in NHP was 2900 ± 351 cells/mm2 (n=10 eyes), with 71% ± 7 hexagonal cells. In comparison, histomorphometric evaluation of dual-stained (0.25% trypan blue and 0.2% alizarin red S) fresh rabbit corneas produced density values 18% higher than specular microscopy (3510 ± 351 cells/mm2, n=6 eyes). Due to species-specific eye position and corneal curvature, the angle of image capture is important for accuracy of specular microscopy imaging and likely contributes to differences when compared to histomorphometry. However the advantage of longitudinal data obtained by specular microscopy outweighs this discrepancy and potentially reduces animal use. 2014 th 35 American College of Toxicology Annual Meeting P433 Evaluation of the Use of Transponders in the Tg.rasH2 Mouse Model. M. Paranjpe1, J. Belich1, R. Elbekai1. BioReliance by SAFC, Rockville, ABSTRACTS MD, USA1 Subcutaneous implantation of transponders is a commonly used method for animal identification in preclinical studies involving conventional rodents. However, no robust data is available to support the use of subcutaneous transponders in the transgenic Tg.rasH2 mice. The Tg.rasH2 mouse model is commonly used in 26-week carcinogenicity assessment studies, and the objective of this study was to determine if implantation of glass encapsulated microchip transponders increases the incidence of tumors in this model over a 26-week period. Male and female Tg.rasH2 mice were assigned to three groups. Group 1 consisted of mice in which the transponders were implanted subcutaneously. Group 2 consisted of mice in which transponders were not implanted but were subjected to subcutaneous injection procedure. The third group consisted of mice that were implanted with transponders but were also inoculated intra-peritoneally with positive control material, urethane. In none of the mice was there any tumor formation at the site of injection. The most common finding was a cavity formation at the transponder site with surrounding compressed connective tissue. In a few mice, there was infiltration of inflammatory cells around these cavities. The incidence of spontaneous tumors in these mice remained within the historical control range. As expected, the positive control material, urethane, induced pulmonary tumors and splenic hemangiosarcomas. It is therefore concluded that the use of transponders Tg.rasH2 mice would not negatively impact the outcome of a 26-week carcinogenicity study. P434 Suitable Identification Method for Transgenic Hemophilia Type A Mice (HA). J. Forget1, L. Bernier1, E. Arlund2. 1Charles River Laboratories, Montreal, Quebec, Canada, 2Somark Innovations, San Diego, CA, USA Reliable identification of laboratory animals is critical to safety assessment study conduct to generate quality data. Various identification methods are available, including ear tags, manual tattoos, and radio frequency identification devices (RFID); however, most of these are not suitable for transgenic HA mice as this model has less than 1% of normal factor VIII activity and exhibit prolonged clotting times. Minor lesions induced by ear tags, RFIDs or manual tattoos can result in excessive blood loss leading to death within few hours. The LabStamp tail tattoo system developed by Somark Innovations was evaluated for its suitability. The system allows a controlled depth of needle penetration and delivery of the ink at the desired level; the mid dermal layer of the skin. Injury to blood vessels is avoided therefore alleviating risks of hemorrhage in these hemophilic mice. This method was evaluated on 210 transgenic HA mice that were 6 to 7 weeks of age at time of identification. Animals were observed for up to 7 weeks during which time there were no clinical observations associated with the identification procedure, confirming absence of substantial damage to blood vessels at the set depth of penetration. Integrity of legibility of the tattoo was 2014 also noted to be maintained throughout this 7 week period. The automated characters of the LabStamp system also eliminated risks of ambiguity in characters which may exist with manual tattoo inscriptions. The LabStamp system was therefore considered to provide an appropriate identification method for use with transgenic Hemophilia type A mice. P435 Assessing Mitochondrial Liabilities Using Multiple Assays and Specific Cell Types. K. Gareau1, S. Qin1, J. Bradley1, C. Strock1. Cyprotex US, LLC, Watertown, MA, USA1 Mitochondrial toxicity is one of the most important and widely recognized liabilities associated with drug development. Although this toxicity is well recognized, the proper selection of assays and cell types is not well understood to assess the different types of mitochondrial toxicity. Here we show that with the proper assays and cell types, one can screen multiple mitochondrial toxicities. The first assay which analyzes the maintenance of mitochondrial potential utilizes primary rat hepatocytes and the mitochondrial potential sensing dye, TMRE, which is a highly cell permeable live cell dye. This assay will detect any compound that interferes with the maintenance of potential which is necessary for the production of ATP through the electron transport chain. Another assay utilizes the HepG2 cell line and its dependence on glycolysis for growth which is known as the Warburg effect. Here we show that there is an increased sensitivity for mitochondrial toxins when the HepG2 cells are grown in the presence of galactose. HepG2 cells grown in glucose can also be stained with the mitochondrial dye, Mitotracker red, which can identify compounds which stimulate an increase in the overall numbers of mitochondria due to an interference in the production of energy through glycolysis. A fourth assay analyzes cells for their expression of mitochondrial coded proteins. This assay is effective for analyzing compounds which interfere with the expression of mitochondrial coded proteins such as antivirals and antibiotics. Overall, the selection of cell type and assay are important for screening compounds and identifying their liabilities. STP436 High-Throughput Screening of Toxicity of 24 Metal Oxide Nanoparticles in Escherichia Coli. C. Kaweeteerawat1, A. Ivask2, R. Liu2, H. Godwin2. 1Molecular Toxicology Interdepartmental Program, UCLA, Los Angeles, CA, USA, 2UC Center for Environmental Implications of Nanotechnology (UC-CEIN), Los Angeles, CA, USA Nanotechnology has grown rapidly over the past decade, promising benefits in diverse areas of society. However, the rate of toxicological analysis of nanoparticles (NPs) has failed to keep pace with new NP development, leading to concerns over the potential biological toxicity and environmental contamination of NPs. Here, we developed high-throughput screening assays as a robust platform to rapidly determine magnitude and mechanisms of toxicity of 24 metal oxide nanoparticles (MOx) in the bacteria E. coli. A suite of sublethal assays, consisting of PI/SYTO, XTT, DiBAC, and H2DCFDA, were used to measure viability, respiration rate, 90 th 35 American College of Toxicology Annual Meeting membrane potential, and ROS production, respectively. Growth inhibition analysis revealed that 7 out of the 24 MOx NPs (ZnO, CuO, CoO, Mn2O3, Co3O4, Ni2O3 and Cr2O3) exerted significant toxicity, causing membrane damage and accumulation of ROS. Moreover, in vitro ROS generation assays revealed that 5 out of the 7 toxic particles (Mn2O3, CoO, Co3O4, Ni2O3 and CuO) were strong oxidizing agents. Interestingly, both ZnO and CuO dissolved at high levels after 24 hours in bacterial media (5.27% and 7.89%, respectively), and their toxicity is likely augmented by dissolved metal ions. Structure-Activity relationships (nanoSARs) indicated that hydration and conduction band energies are strongly correlated with toxicity. Our data suggests that the probability of toxicity for a given MOx NP increases when its conduction band energy is closer to that of the redox potential for biological molecules and as its hydration energy decreases. Environmental Health, Cincinnati, OH, USA1 Zinc is both an essential and potentially toxic metal. It is widely recognized that oral zinc supplementation can reduce the effects of the common cold; however, there is strong clinical evidence that intranasal zinc gluconate treatment for this purpose causes anosmia, or the loss of the sense of smell, in humans. Using the rat olfactory neuron cell line, Odora, we have investigated the means by which zinc is toxic to olfactory neurons. Using RNAseq and in silico analyses, we identified pathways associated with oxidative stress, calcium regulation, and ATP generation as being up-regulated upon zinc exposure. Many genes in these pathways are polymorphic in humans, suggesting that individuals may be differentially susceptible to zinc toxicity. We are currently utilizing shRNA to reduce expression of polymorphic genes that are involved in zinc metal response (Mt1/2), oxidative stress (Gclm), and ATP generation (Akr1b) to demonstrate their critical role in mediating zinc toxicity and to identify adaptive pathways in response to reduction of expression of the respective genes. We have also established the time course of recovery in wild-type mice exposed to intranasal zinc gluconate. Future studies are planned using metallothionein knockout mice and glutamatecysteine ligase knockout mice to test the hypothesis that these knockout mice will show increased olfactory mucosal damage and delayed histological and behavioral recovery in olfactory neurons in response to intranasal zinc gluconate administration. STP438 Cigarette Smoke-Induced BBB Toxicity under Normal and altered Glycemia: Proteomic Assessment of BBB Endothelial Dysfunction. S. Prasad1, P. Naik1, M. A. Kaisar1, R. Sajja1, L. Cucullo1. Texas Tech University Health Sciences Center, Amarillo, USA1 Cigarette smoking (CS) is a major risk factor for diabetes mellitus (DM). Both DM and CS have independently been reported to enhance the chances of cerebrovascular and neurological disorders such as stroke and Alzheimer’s disease. However, their combinatorial pathogenic potential through involvement of common pathways lies unexplored. Previous studies from our group show that BBB impairment by CS extracts (CSE) is mediated through induction of vascular adhesion molecules, pro-inflammatory cytokines and matrix metalloproteinases from activated leukocyte and endothelial cells (EC). Similar levels of BBB impairment was observed in BBB EC cultures following exposure to hyperglycemia (HG). Thus, we hypothesized that concomitant exposure to HG and CSE will impair the physiological and functional (tight junctions) properties of BBB ECs in an additive manner through common molecular mechanisms. For this purpose, we assessed the impact of CSE on protein expression levels in BBB EC. Herein we report the specific impact of CSE on BBB EC dysfunction under normal and HG conditions. Upregulation of junction proteins such as ZO-1, claudin-5 and VE-cadherin was observed following the independent exposure to HG and CSE. The resulting effect was further increased by co-exposure to both conditions. Both Glut-1 and SGLT-1 was upregulated by CSE and HG-CSE co-exposure (although to a lesser extent). We further observed an additive increase of IL-6 and VEGF release following HG-CSE co-exposure when compared to controls. In summary, our results show an additive effect of CSE over HG exposure on BBB ECs which suggests the involvement and potentiation of common molecular mechanisms. STP439 Protecting the BBB from Tobacco Smoke Damage Using Popular Antioxidants Supplements: Is It Really Working? M. A. Kaisar1, S. Prasad1, P. Naik1, L. Cucullo1. Texas Tech University Health Sciences Center, Amarillo, Texas, USA1 We have recently shown that BBB exposure to soluble cigarette smoke extract (CSE) at physiological concentrations triggers a strong endothelial inflammatory response through induction of vascular adhesion molecules, pro-inflammatory cytokines and matrix metalloproteinases. Nrf-2- a transcription factor involved in anti-oxidant response signaling was also increased in CSE-exposed endothelial cells substantiating the role of the ROS generation in BBB toxicity. We have also shown that loss of BBB function and integrity following an ischemic/reperfusion injury is significantly worsened by CSE and pretreatment with α-tocopherol and/or ascorbic acid proved highly protective for the BBB. Recent studies suggest that High mobility group box 1 (HMGB1) can be actively secreted in response to exogenous and endogenous inflammatory stimuli including ROS. Whereas, HMGB1 elicits inflammatory responses at the BBB by increasing the expression of pro-inflammatory cytokines and MMP activation. Based on these premises, our underlying hypothesis is that prophylactic administration of antioxidants can reduce CSE and/or inflammatory-dependent BBB damage. Thus the goal of this study is to evaluate and rank the prophylactic BBB protective effectiveness of 5 popular and readily available antioxidants (Coenzyme Q10, Lipoic acid, Glutathione, Resveratrol and Melatonin) against exposure to CSE using human BBB endothelial cells. Our preliminary data analyzing the inflammatory response (Il-6, VCAM-1 and E-selectin) of BBB endothelial cells to CSE following antioxidant treatment suggest Resveratrol to be the most effective. Interestingly no significant protective effect was observed in response to GSH administration. 91 ABSTRACTS STP437 Investigation of Zinc Toxicity in Olfactory Neurons Using In Silico and Molecular Techniques. H. Hsieh1, M.B. Genter1. University of Cincinnati, Department of 2014 th 35 American College of Toxicology Annual Meeting STP440 Comparative Analysis of Endogenous Gene Expression Levels Across Human and Rat Primary Endothelial Cells. C. Snyder1, A. Kauss1, E. Doudement1, V. Musinipally1, T. Ngo1, T. Zabka1, H. Uppal1, D. Misner1, D. Dambach1, R. Pai1. Genentech, ABSTRACTS South San Francisco, CA, USA1 Endothelial cells (ECs) play important roles in regulation of blood flow, nutrient exchange, vascular structure, and pathological response. Different tissues and vascular networks possess unique EC populations, each with distinct gene expression profiles and functional capabilities. EC activation is a major mechanism in the development of drug-induced vascular injury (DIVI), a preclinical phenomenon that often delays or prevents drug candidates from progressing through development. Translation of preclinical DIVI to the clinic is poorly understood and no legitimate biomarkers have been identified so far for its early detection. In an attempt to select an appropriate EC type for developing an in vitro screening tool, we first established the similarities and differences across human and rat species using primary human and rat ECs from more than 20 different organs and tissue types. Over 100 EC structure- and function-related genes were quantified in isolated RNA using Taqman® probes. Expression levels relative to endogenous controls were compared by hierarchical clustering. We further validated some of these distinct profiles for tissuedependent protein expression using immunocytochemistry. Our results show that about 40% of the genes analyzed were consistently expressed across in both rat and human ECs. In addition, protein expression data correlated well with individual gene expression profile. Based on these comparative data, changes in genes consistently expressed across rat and human are being used to evaluate the effects of drugs known to induce DIVI in a rat EC culture system. IGP441 Evaluation of Functional Impairment of Neonatal Rat Ventricular Cardiomyocytes In Vitro: An Atomic Force Microscopy Study. G. Del Favero1, V. Martinelli2, T. Lanzicher1, L. Mestroni3, O. Sbaizero1. 1Department of Engineering and Architecture University of Trieste, Trieste, Italy, 2International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy, 3University of Colorado Cardiovascular Institute, Aurora, CO, USA 2014 nuclear cell elasticity (Controls 1.12x103 ±7.7x102 Pa vs 1 μM Cyt-D 1.09x103 ± 5.6x102 Pa) but a decrease in both cell viscoelasticity behavior and adhesion strength of the cardiac cell to the AFM probe. Moreover, contractile activity of cardiac cells in response to mechanical stimulation was impaired at all concentrations tested. We propose a panel of endpoints that may suggest early cytotoxic events: namely morphology, elasticity, viscoelasticity, adhesion and contractile activity that are directly related to the ability of the cell to respond to the extracellular environment and to mechanical stimulation. GFP442 Detection of Algal Toxins in Water and Rumen Contents Utilizing the Protein Phosphatase 1 Inhibition Assay. C. Moore1, B. Puschner1. UC Davis School of Veterinary Medicine, Davis, CA, USA1 Microcystins (MCs) are acute hepatotoxins of increasing concern in drinking and recreational waters worldwide. Produced by bluegreen algae, MCs inhibit serine/threonine protein phosphatases (PP) 1 and 2A, and are a major public health risk. A cost-effective PP1 enzymatic assay using p-nitrophenyl phosphate was developed to quickly assess water and rumen samples for PP1 inhibiting toxins. Significant inhibition was determined via a linear model comparing increasing concentrations of sample extract and the log-transformed ratio of the exposed rate of PP1 activity to control rate of PP1 activity. In August 2013 a dog became ill and died after swimming in Clear Lake, California. Water samples were collected one to five weeks after clinical signs developed. PP1 inhibition correlated with detected levels of MC congeners MC-LR, -LA, YR and -RR via LC-MS/MS: week one samples had the highest levels of all four MCs and had significant potent PP1 inhibitor activity (p < 0.001), while week five samples had trace amounts of MC-LR and no significant PP1 inhibition (p < 0.1), with the slope of the linear models becoming less steep with decreasing MC congener concentrations. In August 2013 six cattle died in and around two ponds in Kentucky, with pond B identified to contain potential toxin-producing algae. MC-LA, -LR, -YR and –RR were not detected in Pond A (Pond B was not tested). Pond B and all three tested rumen contents, but not Pond A, had significant PP1 inhibition (p <0.001), showcasing the diagnostic strength of using functional assays in conjunction with LC-MS/MS. Compression, shear, and flexural stresses: cells physiological environment is all but static and cellular ability to respond to mechanical stimulation is crucial for its proper function. In this light, there are increasing evidences that suggest how variation of cell mechanical properties can be associated to cytotoxicity [Zou et al., 2013 PlosONE 8(8):e73499; Del Favero et al., 2014 Toxicology Letters 225: 285-293; Dulińska-Molak et al., 2014 J Biomed Nanotechnol. 10(4):651-9 ]. In the present work, we propose the use of Atomic Force Microscopy (AFM) as a tool for assessing morphological and mechanical changes of primary cultures of neonatal rat ventricular cardiomyocytes in response to exposure to Cytochalasin D (Cyt-D). Cyt-D is a mycotoxin and a well know cytoskeleton-disrupting compound, which blocks actin polymerization. AFM was able to confirm cell morphology alteration due to Cyt-D, with the appearance of granulation on the cell surface; at the same time no apparent variation of the 92 th 35 American College of Toxicology Annual Meeting 2014 500 Series—Safety Evaluation Pharmaceuticals P500 Acute and Repeated Dose Toxicity Studies of Novel Pyridazine Substituted S-Triazin-2-Thione Derivatives as New Class of Antihypertensive Agent. R. Mishra1, A. A. Siddiqui2, A. Husain2, M. Rashid2. 1School of Pharmacy and Emerging Sciences, Baddi University of Emerging Sciences and Technology, Makhnumajra, Baddi, Distt. Solan, Himachal Pradesh 173205, India, 2Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India P502 A Case of Acute Hepatic Failure due to Acetaminophen verdose Treated with Molecular Absorbents Recirculating System®. B. K. Yang1, J. S. You1, Y. S. Joo1, S. P. Chung1, H. Schick Lee1. Severance Hospital, Seoul, Republic of Korea1 We report on a patient who developed acute hepatic failure despite intravenous N-acetyl cysteine therapy who was treated with the Molecular Adsorbents Recirculating System (MARS). She presented 20 hours after the ingestion of 13 g of acetaminophen. The MARS is based on albumin dialysis principle which can be applied for patients with acute poisoning from drugs that have high protein-binding capacity because of its ability to selectively remove from circulation protein-bound toxins. The clinical toxicologist should be consider this technology when treating patients with hepatic failure following acetaminophen poisoning. P501 Surveillance of Chloramphenicol Residues in Milk, Eggs, and Chicken Meat By LCMSMS. G. Sarathchandra1. 1Pharmacovigilance Laboratory for Animal Feed and Food Safety, Chennai, India, 2Tamil Nadu Veterinary & Animal Sciences University, Chennai, India Chloramphenicol has been banned for use in all foodproducing animals by the European Union (EU), and Most of the developed countries.. The EU recently set a minimum required performance limit (mrpl) for chloramphenicol determination at 0.3 μg/kg (ppb) in all foods of animal origin. The growing food safety concerns call for intensive surveillance of chloramphenicol in food products. The objective of the study was to assess whether milk, eggs and chicken meat produced by the livestock farmers in TamilNadu state of India were contaminated with chloramphenicol residues. Liquid chromatography/mass spectrometry (LC/MSMS) method was employed for the determination of chloramphenicol (CAP) 93 ABSTRACTS Pyridazines are important biologically active scaffolds, possessing antihypertensive, cardiotonic, analgesic, anti-inflammatory, antidepressant, antibacterial, anticancer, nephrotopic drugs, antithrombic, diuretic and anti-HIV activities. Some new 7-substituted-phenyl-3,4,8,9tetrahydro-2H-pyridazino[1,6-a] [1,3,5]triazin-2-thione derivatives were synthesized. Pyridazines and its derivatives are known for their cardiovascular effects. To support the safety of synthetic compound 7-substituted-phenyl3,4,8,9-tetrahydro-2H-pyridazino[1,6-a][1,3,5]triazin-2-thione derivatives, it has been examined in an acute and in a 4-week repeated dose toxicity study in rats. Animals were divided into groups of 5 animals each. The compound (20mg/kg body weight and 40 mg/kg body weight) was injected ip after suspending in 1% carboxymethylcellulose solution in single dose resulted in no adverse events or mortality. Also, the compound administered as a daily dose of 40 mg/kg for 4 weeks by gavage resulted in no adverse events or mortality. No evidence or treatmentrelated toxicity was detected during both studies. Data analysis of body weight gain, food consumption, clinical observations, blood biochemical, haematology, organ weight ratios and histopathological findings did not show significant differences between control and treated groups. It is concluded that the synthetic compound orally administered to rats was safe and that no treatment-related toxicity was detected in both acute ip route of exposure and repeated dose (4 weeks) oral route of exposure (40 mg/kg of body weight) toxicity studies. residues in milk, eggs, chicken muscle and liver, and kidney. CAP was extracted from the samples with acetonitrile and defatted with hexane. The acetonitrile extracts were then evaporated, and residues reconstituted in 10mM ammonium acetate--acetonitrile mobile phase and injected into the LC system, and detection was by a triple quadrupole mass spectrometer operated in selected reaction monitoring (SRM) mode. The method studied was sensitive enough to detect and quantify 0.050 ug/kg (ppb) chloramphenicol for screening purposes, much lower than the Minimum Required Performance Limit (MRPL) of 0.3 μg/kg imposed by European Commission's regulation. The study revealed that most of the samples were in compliance with MRL and growing awareness amongst farmers to avoid banned antibiotic CAP. th 35 American College of Toxicology Annual Meeting P503 AMG 595: A Novel ADC with Therapeutic Potential in Glioblastoma. M. Santostefano1, M. Engwall2, N. Everds1, R. Guzman2, V. Chow3, V. Upreti3, K. Hamblett4, J. Hill5, H. Vargas2. 1Comparative ABSTRACTS Biology and Safety Sciences, Amgen Inc., Seattle, WA, USA, 2 Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA, 3Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, WA, USA, 4Therapeutic Innovation Unit, Amgen Inc., Seattle, WA, USA, 5Early Development, Amgen Inc., Thousand Oaks, CA, USA AMG 595 is an antibody drug conjugate (ADC) composed of a fully human anti-EGFRvIII specific IgG1 antibody conjugated to the semi-synthetic derivative of maytansine (DM1), via a noncleavable linker, MCC. AMG 595 is directed against the unique splice junction epitope, only present in EGFRvIII, which is expressed in a significant proportion of glioblastomas (GBM). The antibody "backbone" does not cross react with wild-type EGFR. AMG 595 induces tumor growth regression in EGFRvIII-positive subcutaneous and orthotopicallyimplanted xenograft models. AMG 595 is internalized into cells and catabolized into lysine-MCC-DM1 (the intracellular active moiety), which binds tubulin and inhibits microtubule polymerization. Nonclinical toxicology studies consisted of a single-dose or 1-month rat IV study with AMG 595 or DM1, respectively, and a 1-month cynomolgus monkey IV study with AMG 595. The toxicity was consistent with the cytotoxic nature of the maytansine (typical DM1 effects on highly proliferative cell populations) and related to total DM1 dose delivered. The highest nonseverely toxic dose in the monkey (10 mg/kg) was used to set the clinical starting dose of 0.5 mg/kg. In a Phase 1 clinical trial in patients with EGFRvIII positive GBM, tumor response (partial response) has been observed in 2 patients with evidence of sustained stable disease in 3 patients. Adverse events were consistent with data from other DM1 containing ADCs. These data demonstrate that AMG 595 has biological impact in Phase 1 and the safety profile is consistent with the cytotoxic nature of DM1 and not related to the EGFRvIII. P504 Safety Evaluation of the Anti-Ang2-TNFalpha Bispecific Antibody in Cynomolgus Monkeys. S. Nicholson1, W. Iverson1, S. Karanth2, J. Connor1, L. Yan1, X. Chen1, N. Dimasi1, R. Faggioni1, P. Ryan1. 1MedImmune LLC, Gaithersburg, MD, USA, 2Charles River Laboratories Preclinical Services, Reno, NV, USA 2014 signs, or changes in food consumption, body weights, injection site observations, physical examination parameters, blood pressure, electrocardiology, ophthalmology or clinical pathology (hematology, coagulation, clinical chemistry, and urinalysis) parameters. However, histopathological evaluation at the end of the dosing phase revealed axonal degeneration and astrocyte hypertrophy of the dorsal funiculi at all levels of the spinal cord at all dose groups. Incidence was 2/3 and 1/3 in the high and mid dose females respectively and 1/3 in the low dose males. Lesions were characterized by multiple dilated axons and associated enlarged astrocytes. There appeared to be no damage to the blood brain barrier or the vasculature in the spinal cord or in any other tissues examined histopathologically. P505 Chronic Repeat Dose Administration of Monoclonal Antibody MEDI-557 in Cynomolgus Monkeys Results in Immunogenicity-mediated Findings. P. Ryan1, M. Rebelatto1, G. Robbie1, S. Ren1, R. Criste1, J. Rojko2, R. Dixit1. 1MedImmune, Gaithersburg, MD, USA, 2CRL Pathology Associates, Frederick, MD, USA MEDI-557 is a Mab directed against the F protein of RSV with three amino acid changes in the Fc region engineered to prolong serum half-life. Toxicity of MEDI-557 was assessed in a repeat dose study in cynomolgus monkeys with monthly intramuscular or intravenous injection of 30 or 108 mg/kg to cynomolgus monkeys for 6-months with a 20-week recovery period. There were no adverse findings in any parameter assessed. Histopathologic findings included hypercellularity of the red pulp of the spleen which is a common finding following administration of foreign proteins to monkeys and may relate to clearance of antibodies or immune complexes (IC) from circulation. A few animals had findings of inflammation in the choroid plexus and the perivascular/ vascular region of the sciatic nerve. Selected tissues were evaluated by immunohistochemistry to investigate the possible immunologic/IC-basis for these findings. Granular deposits of MEDI-557, monkey IgG or IgM were judged as evidence of IC deposition and inflammatory changes in sciatic nerve and choroid plexus were directly associated with the sites of IC deposition. As outlined in ICH S6 guidance, these types of immunogenicity assessments assist in the interpretation of nonclinical results but are not relevant for predicting potential immunogenicity in humans. The toxicokinetics, pharmacodynamics, and potential toxicity of MEDI3549, a novel human anti-Ang2-TNFa bispecific antibody were evaluated in a GLP cynomolgus monkey study. Animals were administered a single loading dose intravenous infusion of MEDI3549 to avoid immunogenicity, followed by subcutaneous injections, twice per week, at 18, 30, or 75 mg/kg for 13-weeks with a 12-week recovery period. All but 3 animals in the low dose group maintained exposure to MEDI3549 following 13 weeks of dosing. AntiDrug antibodies led to loss of exposure in these animals. Administration of MEDI3549 did not result in any clinical 94 th 35 American College of Toxicology Annual Meeting P506 Leveraging Early Pharmacology Studies to Learn About Relevant Safety Characteristics During Early Drug Discovery —A Case Study. S. Mohr1, A. Herrmann1, S. Kustermann1, R. Jagasia1, J. Wichmann1, T. Singer1. Roche Innovation Center Basel, Basel, Switzerland1 P507 Repeat Dose Toxicity Studies of 3-Bromopyruvate in Dogs Following Intrahepatic Artery and Intraperitoneal Infusion. C.S. Godin1, K.E. Engelke1, J.F. Geschwind2. 1Smithers Avanza, Gaithersburg, MD, USA, 2Johns Hopkins University School of Medicine, Baltimore, MD, USA The anticancer efficacy of the pyruvate analog 3-bromopyruvate (3-BP) has been demonstrated in tumor models, and has been shown to inhibit glyceraldehyde3-phosphate dehydrogenase resulting in a depletion of intracellular ATP and cellular death. Thus, 3-BP may be a potent and promising anticancer agent. Groups of dogs were dosed during two 5-day intervals over 14 days. Each dose was administered to the hepatic artery via a 2-hour infusion at 0, 0.25, 2.5 and 7.5 mg/kg/dose. Mortality occurred at doses of 7.5 mg/kg. Other test article-related effects included emesis, decreased body weight and food consumption, elevated liver enzymes and leukocytosis, macroscopic findings in the liver and lymph nodes, and microscopic changes in the hepatic artery, bile duct, peritoneum, lymph nodes, liver, gallbladder and mesentery. The no-observed-adverse-effect level (NOAEL) is less than 0.25 mg/kg/dose and the highest nonseverely toxic dose (HNSTD) is <2.5 mg/kg/dose. In a separate study, groups of dogs were dosed for 21 consecutive days via a 2-hour intraperitoneal infusion at doses of 0, 0.25, 2.5 and 5 mg/kg. No mortality occurred, and test article-related emesis, decreased body weight and food consumption, decreased RBC count, hematocrit, and hemoglobin, increased serum chloride, and macroscopic and microscopic findings at the infusion site, lymph nodes and peritoneum were noted. The NOAEL is < 0.25 mg/kg/dose based on the microscopic changes in the peritoneum (deposition of fibrin and fibrosis along with inflammatory cells and hemorrhage) of all treated groups, and the HNSTD is 2.5 mg/kg/dose. P508 Two Tales: ADA-Mediated Toxicities in Mice and Monkeys Treated with a Therapeutic Monoclonal Antibody. J. Wheeler1, C. Thompson1, S. Eble1, D. Devona1, B. Wang1, R. Li1, G. Rao1, S. Clark2, B. Philip2, T. Bunch2, H. Haggerty1, F. Burleson3, W. Freebern1. 1Bristol-Myers Squibb, New Brunswick, NJ, USA, 2Bristol-Myers Squibb, Mt. Vernon, IN, USA, 3Burleson Research Technologies, Inc., Morrisville, NC, USA During preclinical safety evaluation, apparent anti-drug antibody (ADA)-mediated toxicities were observed in mice and monkeys intravenously administered mAbX at 0, 10 or150 mg/kg weekly. Mice were found dead or moribund within 3 hours after repeated doses of 10 mg/kg mAbX, with the timing of mortality indicative of ADA-mediated hypersensitivity reactions. An investigative study was performed, with continual observations following dosing, cohorts for scheduled necropsies following the 3rd weekly dose, and analyses including ADA, complement, mast cell protease-1, histamine, bradykinin, tryptase, serotonin, and circulating immune complexes. Mortality was reproduced in the 10 mg/kg group, with morbidity occurring within 30 minutes after repeated doses. A striking correlation between ADA responses and incidence of clinical observations and mortality was observed, although no correlations with tested chemical mediators were found. These findings suggest that responses did not involve IgE-mediated mast cell degranulation, further supporting an IgG1 ADA-mediated hypersensitivity. ADA-mediated toxicities were also observed in cynomolgus monkeys administered mAbX. Two of 6 monkeys dosed at150 mg/kg had decreased platelets following the second dose, with 1 euthanized on Day 25 to further investigate concomitant decreases in food consumption and body weight. These monkeys had the highest ADA levels, and flow cytometric and immunohistochemical analyses demonstrated mAbX/ADA complexes on platelets and in spleen (germinal center lymphocytes and macrophages) supporting that platelet decreases were likely due to mAbX/ ADA complex-platelet bound mediated splenic clearance. In conclusion, these two ‘tales' highlight investigations used to confirm toxicities in preclinical studies were ADA-mediated and not direct effects of the mAb. 95 ABSTRACTS Roche was able to significantly reduce the safety-related attrition of selected clinical leads over the last couple of years through safety representation at earlier phases of drug discovery. Compound failure could be significantly reduced by identifying potential target-related safety liabilities early on via a thorough safety target assessment and the generation of more predictive safety data. The importance of an early safety assessment will be discussed using an innovative project as case example. There is accumulating evidence that neurogenesis in the adult hippocampus contributes to brain physiology and disease. We developed a human embryonic stem cell-derived neural precursor cell model for phenotypic screening of compounds with neurogenic properties to then translate into in vivo models of disease. from the start, a tailor-made safety strategy was developed to optimize safety and efficacy in tandem. In early pharmacology studies the brain was used to evaluate efficacy based on hippocampal neurogenesis while the peripheral organs were collected to assess safety. The team identified molecules with excellent properties in efficacy at the cellular and behavioral level. But this came with the price of safety liabilities in form of multi-organ tissue proliferation. from the start, the project team realized safety would play a vital role and the project was intensively supported by early and mechanistic safety, ADME and discovery pathology. Moreover, the early project toxicology leader was not only team member, but also sharing project leadership equally with the chemistry and biologist leader. Ultimately, this approach allowed implementing safety readouts very early during drug discovery. 2014 th 35 American College of Toxicology Annual Meeting P509 An Approach to Assess the Potential Reproductive and Developmental Toxicity of Anti-rHuPH20 Antibodies that are Cross-Reactive to Species-Specific PH20 in the Mouse and Rabbit. R. Veneziale1, M. A. Printz1, F. Araiza1, J. Bahn1, B. J. Sugarman1, B. Dietrich2, A. M. Hoberman3, J. A. Cavagnaro4, G. I. Frost1, F. Drake1, D. C. Maneval1, P. J. Lapinskas1. 1Halozyme Therapeutics, Inc., San Diego, CA, USA, Baxter Innovations GmbH, Vienna, Austria, 3 Charles River Laboratories Preclinical Services, Horsham, PA, USA, 4 Access BIO, Boyce, VA, USA, 5Intrexon Corporation, San Diego, CA, USA ABSTRACTS 2 Regulatory guidances do not currently provide specific advice on assessing the potential safety risk of antibodies to therapeutic proteins that have an endogenous counterpart. A recombinant human PH20, rHuPH20, is used clinically to facilitate the subcutaneous absorption of fluids and drugs by rapid and transient degradation of hyaluronan in the skin. PH20 is known to be expressed only in the adult male reproductive tract on sperm, where it plays a nonessential role in fertilization by aiding in the penetration of the sperm through the hyaluronan-rich cumulus layer of the oocyte. Recently, a single reference reported PH20 expression in the central nervous system (CNS). Therefore anti-rHuPH20 antibodies may theoretically impact offspring fertility and neurological development, if PH20 expression is confirmed in CNS. In a pre/postnatal developmental toxicity study of rHuPH20, conducted in mice, anti-rHuPH20 antibodies were characterized and the potential impact on offspring neurobehavioral development and fertility was evaluated. Dedicated studies with anti-rHuPH20 antibodies were also conducted in the rabbit to evaluate adult fertility, embryo-fetal development, and postnatal development through adulthood and mating. Anti-rHuPH20 antibodies generated in these nonclinical species were cross-reactive to the species-specific PH20 and could also cross-neutralize the PH20 hyaluronidase activity in the rabbit. These studies did not detect any impact of anti-PH20 antibodies on reproduction or development including: adult fertility, embryo-fetal development or offspring growth, neurological development or fertility. Life cycle evaluations in both rodent and nonrodent species that generate cross-react antibodies, provides a comprehensive assessment of the potential effects of anti-therapeutic antibodies on an endogenous protein. 2014 P510 Antibodies Generated to Recombinant Human PH20 Hyaluronidase, rHuPH20, Do Not Impact Fertility in Multiple Species. R. Veneziale1, M. A. Printz1, F. Araiza1, J. Bahn1, B. J. Sugarman1, B. Dietrich2, A. M. Hoberman3, J. A. Cavagnaro4, G. I. Frost5, F. Drake1, D. C. Maneval1, P. J. Lapinskas1. 1Halozyme Therapeutics, Inc., San Diego, CA, USA, 2Baxter Innovations GmbH, Vienna, Austria, 3 Charles River Laboratories Preclinical Services, Horsham, PA, USA, 4 Access BIO, Boyce, VA, USA, 5Intrexon Corporation, San Diego, CA, USA The recombinant human PH20 hyaluronidase, rHuPH20, is used clinically to facilitate the subcutaneous absorption of fluids and drugs by rapid and transient degradation of hyaluronan in the extracellular matrix of the skin. Endogenous PH20, expressed in the adult male reproductive tract and localized to the sperm membrane, plays a nonessential role in fertilization by aiding in the penetration of the sperm through the hyaluronan-rich cumulus layer of the oocyte. As a result of PH20's role in fertilization, it was proposed as a target for development of an immunocontraceptive vaccine; however, this vaccine approach failed to impact fertility in mouse, rabbit, monkey, and sheep, though reversible infertility was observed in the guinea pig. Thus, a theoretical risk exists that anti-rHuPH20 antibodies could bind sperm and impact fertility. Nonclinical safety assessments of anti-rHuPH20 antibodies on fertility were performed in a chronic monkey toxicity study, fertility studies in male and female rabbits and in the postnatal phases of reproductive toxicity studies in mouse and rabbit offspring. Anti-rHuPH20 antibodies generated in nonclinical species were cross-reactive to the species-specific PH20. In monkeys and rabbits, these antibodies could cross-neutralize the species-specific PH20 hyaluronidase activity. Anti-rHuPH20 antibodies had no effect on adult male or female fertility or on the fertility of offspring exposed throughout development. Our approach provides a comprehensive assessment of the potential for effects of antibodies to a sperm antigen on fertility and is consistent with the prevailing concept that antibody responses to multiple sperm antigens are required to impact fertilization. P511 Pharmacokinetics and Metabolite Profile of Diclofenac and Lumiracoxib in Mice. C. E. Wilson1, K. Schreiter2, R. Wehr2, R. J. Riley1, A. Lewis1, A. P. Dickie1, I. D. Wilson3. 1Evotec (UK) Ltd, Abingdon, UK, 2Evotec AG, Göttingen, Germany, 3Imperial College, London, UK Diclofenac and lumiracoxib are nonsteroidal anti-inflammatory drugs (NSAIDs) taken or applied to reduce inflammation and as an analgesic reducing pain in certain conditions. Both have been associated with adverse drug reactions involving drug induced liver injury (DILI). Diclofenac was first introduced in the UK in 1979 and despite the potential for DILI is available as a generic drug in a number of formulations. Lumiracoxib is manufactured by Novartis, and whilst it was withdrawn from many markets as a direct result of DILI, it is still available 96 th 35 American College of Toxicology Annual Meeting P512 Preclinical Safety Assessment of a Bile Acid Receptor Agonist Intended for the Treatment of Type 2 Diabetes. J. Horvath1, T. Dorr1, C. Hixson1, R. Mangipudy1, E. Dierks2, K. Foster2, M. Orsini2, J. Smalley2, Q. Sun2, L. Buchanan2, J. Li2, J. Panzica2, J. MacGuire3. 1Bristol-Myers Squibb Drug Safety Evaluation, New Brunswick, NJ, USA, 2Bristol-Myers Squibb R&D Discovery, Princeton and Hopewell, NJ, USA, 3Bristol-Myers Squibb Veterinary Sciences, Hopewell, NJ, USA Agonism of the G-protein-coupled bile acid receptor 1 (GPBAR1/TGR5) promotes GLP-1 secretion and was proposed as a mechanism for the treatment of type 2 diabetes. BMSTGR5-A, a TGR5 agonist, was evaluated for nonclinical safety. Mice given BMS-TGR5-A orally (2 weeks) had gallbladder distension (a pharmacologically-mediated effect) and minimal vacuolation of gallbladder epithelium (nonadverse) whereas dogs developed marked hyperplasia/hypertrophy of the walls of the extrahepatic bile ducts and gallbladder (no NOAEL established) and at high doses, periductular edema/ fibrosis in the larger bile ducts in the liver and pancreatic ducts. Additionally, cardiovascular (CV) safety studies in these species showed significant increases in heart rate and decreases in blood pressure (BP). To determine if these effects were target-based, an investigative approach was developed to evaluate BMS-TGR5-A with other TGR5 agonists (> potency to BMS-TGR5-A but of different chemotypes). Daily doses of BMS-TGR5-A and BMS-TGR5-B given to dogs resulted in gallbladder concentrations of BMS-TGR5-B > BMSTGR5-A and no gallbladder pathology was observed with BMS-TGR5-B. Following high single doses of BMS-TGR5-C (chemotype with increased systemic bioavailability), TGR5 knock-out mice maintained normal sinus rhythm and BP whereas wildtype controls had similar CV effects as noted previously. TGR5 expression (qPCR) in gallbladder and heart was also established. These results demonstrate that BMSTGR5-A-associated gallbladder toxicity was likely independent of TGR5 whereas CV changes were likely a pharmacologic TGR5-based class effect. Overall, the preclinical studies on TGR5 agonists demonstrated the potential of TGR5 agonism to treat type 2 diabetes, but also identified a specific risk of pharmacologically-mediated hemodynamic changes. P513 Measuring T-Cell Dependent and Independent Antibody Responses (TDAR/TIAR) in the Cynomolgus Monkey: Abatacept (CTLA4-Ig) and Methotrexate Lead to Immunosuppression in a TDAR but not TIAR Response. G. Bannish1, M. Perpetua1, T. Ziegelhofer1, A. Curran1, L. Coney2. 1Huntingdon Life Sciences, East Millstone, NJ, USA, Huntingdon Life Sciences, Huntingdon, Cambridgeshire, UK 2 The TDAR response is used to evaluate drug effects on the immune system, assessing antigen presentation and T and B lymphocyte function. Cynomolgus monkeys were injected intradermally with 100 ug Keyhole Limpet Hemocyanin (KLH) in the absence of adjuvant, injected subcutaneously with 6 limit of flocculation (LFU) of tetanus toxoid (TT), or injected intravenously with 25 ug DNP-LPS. Animals were boosted after approximately 2 months, and sera was collected twice weekly to evaluate antibody responses. Animals developed a detectable IgM response with mean inverse titers of 60,000 ten days following primary immunization. IgG responses were detectable 14 days post primary injection with inverse titers of 66,000 and 7 days post secondary challenge with inverse titers of over 316,000. Treatment of a cohort of these animals with Abatacept led to a near elimination of antibody responses. Antibody responses to KLH were reduced but not ablated when treated with 1 mg/kg methotrexate via subcutaneous injection. Maximal mean IgM responses to KLH 10 days following primary immunization were reduced from 60,000 to 8,300. Maximal mean IgG responses to KLH were reduced from 316,000 to 191,000 with maximal responses delayed from 7 to 10 days post secondary KLH challenge. TIAR responses were observed following treatment with the T-independent antigen type 1 (Ti-1) DNP-LPS, with average inverse titers of 26,000. Neither Methotrexate nor Abatacept treatment reduced the magnitude of response, consistent with the lack of need for T cell help. 97 ABSTRACTS in a few countries, including Mexico, Ecuador and the Dominican Republic. Given the need for reduced attrition in drug development caused by DILI there is a need to develop more predictive models of metabolism and toxicity including both in vitro and in vivo model systems. One such in vivo model is represented by the recently introduced "chimeric" humanized mice, where human hepatocytes can replace up to 90+% of the murine hepatocytes. Prior to studies in this mouse model to investigate how well such mice recapitulate the human metabolism for diclofenac and lumarocoxib it was necessary to fully characterize the biotransformation of these compounds in normal mice. The pharmacokinetics (PK) and metabolite profile of diclofenac and lumiracoxib were therefore investigated over 24h following the administration of 10 mg/kg oral doses to male C57Bl/6 mice (n=3/compound). The pharmacokinetics and metabolite profiling will form the basis for further work using chimeric humanised and chimeric murinised mice (FRG KO/c57Bl6 ; Yecuris, USA). 2014 th 35 American College of Toxicology Annual Meeting P514 Two-Year Oral Carcinogenicity Study of Udenafil, a Potent Novel Phosphodiesterase-5 Inhibitor in B6C3F1 Mice. K.S. Moon1, Y.B. Kim1, K.K. Kang2, E. J. Jeong1. 1Korea Institution of ABSTRACTS Toxicology, Daejeon, Republic of Korea, 2Dong-A ST Pharmaceutical Company, Yong-In, Republic of Korea Udenafil is a new PDE5 inhibitor that has been approved in South Korea for the treatment of erectile dysfunction (ED). As a part of safety evaluation program for human use, a twoyear oral carcinogenicity study was conducted to investigate a tumoriogenic potential of Udenafil in mice. The test article was given orally to male and female B6C3F1 mice (60 animals/sex/ group) once daily for at least 104 weeks at dose levels of 0, 30, 100/150 and 300/500 mg/kg for males, and 0, 50, 150 and 500 mg/kg for females. There were no test article-related changes on survival rates, clinical signs, ophthalmology, and bone marrow smear examination in either sex of any treated groups. In addition, transient changes in body weights and food intake were observed in males at 300/500 mg/kg/day and in females at 500 mg/kg/day throughout the study. In microscopic examination, there were no test article-related non-neoplastic or neoplastic findings in all treatment groups. Therefore, it was determined that there was no evidence of carcinogenicity of the Udenafil, based on the incidence of benign and malignant tumors in the study and the no-observed-adverse-effect level (NOAEL) was considered to be 500 mg/kg/day. P515 Gamma-Secretase Inhibitor BMS-708163 (avagacestat) Decreases Beta Amyloid Levels in Brain and Cerebrospinal Fluid in the APPSWE 2576 Hemizygous and Wild Type Mice. C. Korgaonkar1, G. Pilcher1, V. Guss2, G. Cadelina2, J. Meredith2, H. Gu3, B. Wang4, R. Bunch1, T. Sanderson1. 1Bristol-Myers Squibb Company, Mt. Vernon, IN, USA, 2Bristol-Myers Squibb Company, Wallingford, CT, USA, 3Bristol-Myers Squibb Company, Lawrenceville, NJ, USA, 4Bristol-Myers Squibb Company, New Brunswick, NJ, USA BMS-708163 (avagacestat), a gamma secretase inhibitor, was evaluated in a transgenic mouse model that overexpresses human Amyloid Precursor Protein (APP) and is used to study amyloid lowering and microhemorrhage potential in the brain. Avagacestat was dosed orally to B6;SJL Tg(APPSWE)2576 hemizygous mice (Tg2576) and wild type mice (Wt2576) at 0, 100 or 300 mg/kg/day for 2 weeks to evaluate its pharmacokinetic profile; plasma and brain concentrations of avagacestat and its weakly active metabolite, BMS 737622; and Aβ peptide (Aβ40 and Aβ42) levels in brain and cerebrospinal fluid (CSF). The CSF and brain samples were collected at 5 and 10 hours post dose on Day 14. Plasma avagacestat AUC values generally increased dose proportionally in Wt2576 mice. Mean avagacestat plasma concentrations at 5 and 10 hours post dose were similar for both strains. Brain (cerebellum) and plasma avagacestat and BMS-737622 concentrations increased in a dose-related manner with generally similar mean cerebellum/ plasma concentration ratios over time, across doses, and between mouse strains. At 5 hours post dose, dose-dependent decreases in brain Aβ40 levels relative to controls were noted 2014 in Tg2576 (up to 80%) and Wt2576 (up to 50%) mice. In Tg2576 mice, dose-dependent decreases in Aβ40 and Aβ42 levels in CSF (up to 100%) relative to control were noted. Overall, brain Aβ40 and Aβ42 reductions were higher in the Tg2576 mice compared to the Wt2576 controls. Collectively, these data confirmed that avagacestat was pharmacologically active in Wt2576 and Tg2576 mice at doses that may be useful for the evaluation of brain microhemorrhage potential. P516 Biotherapeutics and Immune Reactions: What are the Incidences and the Endpoints that Support an Immune Response? A. Prefontaine1, L. Bernier1. Charles River, Senneville, Quebec, Canada1 Biotherapeutics are recognized to present a risk of eliciting immune responses in nonclinical models and responses range from the simple presence of antibodies to clinically expressed adverse events. These immune reactions can confound interpretation of test article toxicities and it is therefore important to characterize the immune reactions in these studies in order to support the occurrence of an immune reaction. A review of historical data for studies conducted with biotherapeutics in our facilities was performed with respect to the incidences of positive ADA, clinical signs and pathology findings in different species. Moreover, a summary of investigations performed to demonstrate that the adverse events were caused by an immune reaction was assessed in order to define which endpoints provided supportive evidence. Clinical signs indicative of an immune reaction were observed in approximately 28% of studies where positive ADAs were noted and ranged from nonadverse redness/ swelling of extremities (27%) to adverse respiratory effects and/or death (73%). Four out of ten studies with adverse reactions required additional investigative studies to further demonstrate that the findings were caused by an immune reaction. For these investigative studies, cytokines release, complement factor and/or immunohistochemistry/immunocomplex assays were performed. Given the incidence in our facilities of adverse immune reaction and investigative studies, we consider that the addition of cytokine, complement factor, or immunohistochemistry/immuno-complex to study designs helps distinguish between compound toxicity from speciesspecific secondary immune reaction. 98 th 35 American College of Toxicology Annual Meeting P517 Simvastatin and Dipentyl Phthalate Display Different Modes of Action but Exhibit Dose Additive Effects on Fetal Testicular Testosterone Production in Sprague-Dawley Rats. B. Beverly1, C. Lambright1, J. Furr1, H. Sampson1, G. Travlos2, P. Foster2, B. McIntyre2, V. Wilson1, L. E. Gray Jr.1. 1US Environmental Protection Agency, Research Triangle Park, NC, USA, 2National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA Abstract does not reflect the views of the EPA. P518 Stem Cells and Regeneration of Toxic Liver Fibrosis. A. F. Tohamy2, H. M.E. Imam1, A. F. Tohamy2, H. M. Rezk3. 1Anatomy and Embryology Department, Faculty of Veterinary Medicine Suez Canal University, Ismaillia, Egypt, 2Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt, 3Anatomy and Embryology Department, Faculty of Veterinary Medicine Cairo University, Giza, Egypt Aims: To investigate the potential therapeutic effect of hUCMSCs on CCl4-induced induced liver fibrosis in rats. Methods: Ninety adult female rats were used in this study, sixty of them were injected with CCl4 (0.5ml/kg) to induce liver fibrosis. hUCMSCs (CD34, CD45 and CD105) separated by MiniMACS Separator and cultured for 14 days; then infused into the rats' tail vein (1×106 cells/rat) after 72 h of the last CCl4 dose. Rats were divided into 4 groups (15 rats/each): control, CCl4, CCl4 & hUCMSCs, and hUCMSCs. After 6th and 8th weeks of treatment, blood samples were collected to assess liver functions (ALT, AST, GGT and ALB) and level of Procollagen III. Histopathological and immunohistochemical assessment of liver were conducted using anti-human monoclonal antibodies against CD34, CK19 and albumin. Results: Antifibrotic effect of hUMSCs evidenced by a significant decrease in (AST, ALT, GGT and liver Procollagen III levels) and significant increase of serum albumin level in the treated groups compared to CCl4 group at both 6th and 8th weeks of therapy. Pathological findings revealed significant increase in normal liver function in stem cell treated groups compared to CCl4 group while there was no significant difference between treated groups at the 6th and 8th weeks post-therapy. Immunohistochemistry finding indicates homing and differentiation of hUMSCs in the rats' liver and expression of albumin. Conclusion: Our findings indicate transplantation of hUCMSCs may be a novel therapeutic approach for treating liver fibrosis. Key Words: CCl4, liver fibrosis, mesenchymal stem cells and rat STP519 Effects of Inflammation on Nilutamide-Induced Liver Injury. A. Al Maruf1, P. O'Brien1. University of Toronto, Toronto, Canada1 Nilutamide (NIL) is used in the treatment of metastatic prostatic carcinoma in association with castration. Its therapeutic use is overshadowed by the rare development of hepatotoxicity that occurs in approximately 0.5 to 1 % of patients. The mechanism of NIL-induced hepatotoxicity is still unknown. Inflammation caused by infections or endotoxins could activate NADPH oxidase, which produces hydrogen peroxide (H2O2) and other reactive oxygen species (ROS) and thus causing oxidative stress. The potential molecular cytotoxic mechanisms of NIL towards isolated rat hepatocytes were investigated in this study using an in vitro oxidative stress inflammation model. A significant increase in NIL-induced (400 µM) cytotoxicity, ROS and H2O2 formation, and a decrease in mitochondrial membrane potential (% MMP) was observed when glutathione (GSH)-depleted hepatocytes were used and this toxicity was decreased by the addition of N-acetylcysteine (a GSH precursor). Catalase-inhibited hepatocytes also increased NIL-induced cytotoxicity, while the direct addition of catalase to the hepatocytes delayed cytotoxicity. When a nontoxic H2O2 generating system (glucose/glucose oxidase) with peroxidase or Fe(II) (to simulate in vivo inflammation) were added to the hepatocytes prior to the addition of NIL, an increase in NILinduced cytotoxicity, ROS formation and a decrease in % MMP were observed that were reversed by 6-N-propyl-2-thiouracil (a peroxidase inhibitor) or desferoxamine (an iron chelator), respectively. Potent antioxidants, resveratrol (a polyphenolic compound), and trolox (the water-soluble vitamin E analogue) significantly decreased NIL-induced cytotoxicity, ROS formation, and increased % MMP. These results raise the possibility that inflammation may be another susceptibility factor for drug-induced liver injury. 99 ABSTRACTS Sex differentiation of the mammalian male reproductive tract is driven, in part, by fetal androgen production. In utero, some phthalate esters (PEs) alter fetal Leydig cell differentiation, reducing the expression of genes associated with steroid synthesis/transport, and consequently, lowering fetal androgen levels. Simvastatin (SMV) is a cholesterol-lowering drug that inhibits HMG-CoA reductase in the cholesterol pathway. As cholesterol is a precursor of steroid hormone biosynthesis, we hypothesized that in utero exposure to SMV during the critical period of sex differentiation would lower fetal testosterone (T) production without affecting genes involved in cholesterol and androgen synthesis/transport. Secondly, we hypothesized that a mixture of SMV and a PE would reduce testosterone levels in an additive manner. Pregnant Sprague Dawley rats were dosed orally with 0, 15.6, 31.25, or 62.5 mg/ kg/day SMV from gestational days (GD) 14-18, and fetuses were evaluated on GD18. On GD18, fetal T and serum lipids were reduced with increasing doses of simvastatin. Further, PEs downregulate several genes in the fetal testes that were unaffected by SMV treatment. Individual administration of SMV (62.5 mg/kg/day) and DPeP (50 mg/kg/day) reduced fetal T by 44% and 37% of control, respectively (p < 0.0001 versus control). When administered as a mixture, SMV and DPeP reduced fetal T in an additive manner (19% of control; p < 0.0001 versus control, SMV, and DPeP), demonstrating that a mixture of chemicals can induce additive effects on fetal T even though they display different modes of action. 2014 th 35 American College of Toxicology Annual Meeting STP520 Cognitive Effects of Simvastatin in the Rat D. Miller1, M. B. Genter1. University of Cincinnati Department of ABSTRACTS Environmental Health, Cincinnati, OH, USA1 Statins are widely prescribed drugs primarily used to lower cholesterol and decrease the risk of cardiovascular disease. These drugs are generally well-tolerated; however there are reports of patients developing confusion and memory loss, which appear to be caused by statins. These reports were considered significant enough for the US FDA to update the warning label of all statins in 2012 to include the potential for negative cognitive side effects. Interestingly, there are also numerous reports regarding the neuroprotective effects of statins in conditions such as stroke, subarachnoid hemorrhage, multiple sclerosis, Alzheimer's and other neurodegenerative diseases. There is considerable controversy in the literature surrounding these contradictory effects. Given the growing rate of cardiovascular disease in the population, as well as the potential of statins to be used as treatment in neurological illness and injury, the need arises to better understand the mechanisms of action of statins in the brain. We have completed a pilot study investigating the effects of simvastatin demonstrating memory impairment in rats in both the Barnes maze and novel object recognition tests. These data will guide future experiments in which behavioral assessments will be conducted, and gene expression changes will be determined at the time that behavioral effects, whether adverse or beneficial, are observed. Our findings may eventually provide a further screen in drug development to avoid negative cognitive effects as the next generation of statins are developed, or identify target pathways to allow optimization of existing statins to increase their effectiveness in the treatment of neurological illness. 100 2014 AMERICAN COLLEGE OF TOXICOLOGY There Are Lots of Reasons Why You Should Be a Member of the American College of Toxicology •Be a part of a collegial organization focusing on applied toxicology •Attend ACT’s Annual Meetings at a reduced registration cost •Access the members-only online directory and other member-restricted resources •Receive your subscription to ACT’s International Journal of Toxicology (and IJT archives) •Learn College and industry news from the ACT Newsletter •Participate on committees and Council positions within the College •Invitations to members-only functions at the Annual Meeting, and much more! Visit www.actox.org to learn more and to submit your membership application today! 101 th 35 American College of Toxicology Annual Meeting 2014 Author Index Poster abstracts are indicated with letters preceding the poster number: Poster (P), Student Travel Award (STP), North American Graduate Fellowship (GFP), and International Travel Grant (IGP). ABSTRACTS A A Siddiqui, A.............................................P500 Aardema, M..................................P310, P418 Abtout, S....................................................P411 Acali, S........................................................P303 Acosta, O.C........................................... IGP115 Albers, T......................................................P416 Althouse, G...............................................P403 Ancerewicz, J............................................P312 Apgar, J.......................................................P112 Apreutese, A.....................P110, P110, P111 Araiza, F..........................................P509, P510 Arlund, E....................................................P434 Ascah, A......................................................P414 Attalla, B.....................................................P428 Auletta, C...................................................P412 Authier, S..........................P411, P413, P414, .................................................. P415, P422 Avlasevich, S.................................P310, P432 Ayehunie, S...............................................P405 B Bahn, J............................................P509, P510 Baker, J........................................................P416 Bannish, G.................................................P513 Barnhart, K................................................P201 Bassett, L............................P413, P414, P415 Beck, M.......................................................P402 Beck, T.............................................P408, P409 Belich, J.......................................................P433 Bemis, J.......................................................P432 Benoit-Biancamano, M.O.........P110, P111 Benz, R. D...................................................P204 Bernier, L........................................P434, P516 Beverly, B...................................................P517 Bhusari, S...................................................P313 Blechinger, S.............................................P401 Bonnette, K...............................................P109 Borders, R. B. R.........................................P427 Bouchard, G..........P103, P104, P400, P425 Boue, S........................................................P305 Bourgeois, R..............................................P424 Bradley, J....................................................P435 Brandt, C....................................................P400 Brocksmith, D...............................P104, P425 Brown, A.....................................................P207 Brown, L.............................P103, P400, P425 Bruening-Wright, A................................P207 Bruning, D.....................................P310, P418 Bryant, K.....................................................P424 Bryce, S.......................................................P432 Buchanan, R..............................................P206 Buchanan, L..............................................P512 Bunch, T......................................................P508 Bunch, R.....................................................P515 Burke, J.......................................................P112 Burleson, F.................................................P508 C Cabanski, M..................................P305, P309 Cabral, L.M. ..............................................P107 Cadelina, G................................................P515 Calvano, J...................................................P407 Cannon, K..................................................P404 Castaneda, J........................................ GFP117 Castro, C.....................................................P306 Castro, H. C................................................P107 Cavagnaro, J. A............................P509, P510 Chakravarti, S...........................................P202 Chen, R.......................................................P106 Chen, T........................................................P306 Chen, J.T. J.................................................P315 Chen, M......................................................P410 Chen, X.......................................................P504 Chow, V.......................................................P503 Christin-Piché, M.S.................................P424 Chung, S.P..................................................P502 Churi, S.......................................................P113 Clark, S........................................................P508 Claudia, F...................................................P420 Cole, T..........................................................P201 Collins, N....................................................P429 Coney, L......................................................P513 Connor, J....................................................P504 Costa, S.................................................. IGP319 102 Criste, R.......................................................P505 Cross, K...........................................P204, P205 Cucullo, L.............................STP438, STP439 Curran, A....................................................P513 D Dambach, D..............................................P118 De Leon, H.....................................P305, P311 DeGeorge, G.................................P405, P406 Del Favero, G....................................... IGP441 Delaney, L..................................................P103 Dertinger, S...................................P310, P432 Devona, D..................................................P508 Dhakal, D.............................................. IGP320 Dhakal, R. R.......................................... IGP320 Dickie, A.....................................................P511 Dierks, E......................................................P512 Dietrich, B......................................P509, P510 Dimasi, N....................................................P504 Dinesdurage, H............................P310, P418 DiPiero, J....................................................P407 Dixit, R.........................................................P505 Dorko, K.....................................................P428 Dorr, T..........................................................P512 Doudement, E..........................................P118 Dougherty, M...........................................P416 Douville, J..................................................P420 Drake, F...........................................P509, P510 Dulize, R.....................................................P309 Dumais, A..................................................P423 Dumont, C.................................................P424 E Early, R........................................................P410 Eble, S.........................................................P508 Eichinger-Chapelon, A..........................P200 El-Fazaa, S..................................................P300 Elamin, A....................................................P309 Elbekai, R.......................................P418, P433 Elliott, M.....................................................P416 Emond, F....................................................P420 Engelke, K.E..............................................P507 Engwall, M.................................................P503 th 35 American College of Toxicology Annual Meeting 2014 Poster abstracts are indicated with letters preceding the poster number: Poster (P), Student Travel Award (STP), North American Graduate Fellowship (GFP), and International Travel Grant (IGP). Everds, N....................................................P503 F G García-Léston, J.................................. IGP319 Gardner, A.................................................P315 Gareau, K...................................................P435 Garg, R........................................................P201 Gendron, R.M...........................................P419 Genter, M.B.........................STP437, STP520 Georges, G.P..............................................P104 Gerrish, K...................................................P313 Geschwind, J.F..........................................P507 Gharbi, N....................................................P300 Glaza, S...........................................P408, P409 Godin, C.S..................................................P507 Godwin, H............................................STP436 Gong, B.......................................................P306 Gonzalez Suarez, I..................................P303 Gordon, C......................................P110, P111 Goudreau, J.L..................................... GFP118 Goyal, S..................................................STP317 H Hager, C......................................................P200 Haggerty, H...............................................P508 Haile, S........................................................P431 Hamblett, K...............................................P503 Hanks, C.............................P103, P400, P425 Hanzlik, R...................................................P104 Harui, A................................................. GFP117 Haruna, J........................................P110, P111 Hayden, P...................................................P405 Haziza, C.....................................................P312 Herrmann, A.............................................P506 Hill, J............................................................P503 Hixon, C..............................P407, P512, P510 Hoberman, A. M......................................P510 Hobson, D..................................................P104 Hoenerhoff, M..........................................P313 Hoeng, J............................P303, P305, P308, ...........................P309, P311, P312, P417 Honor, D.....................................................P201 Hooker, B...................................................P201 Horvath, J..................................................P512 Hsieh, H.................................................STP437 Huang, T.S..................................................P315 Huang, J.W................................................P315 Husain, A....................................................P500 I Imam, H. M.E............................................P518 Ingerson, A................................................P103 Iskandar, A.....................................P305, P309 Ivanov, N....................................................P305 Ivask, A..................................................STP436 Iverson, W..................................................P504 103 J Jaeschke, H...............................................P428 Jagasia, R...................................................P506 Jasmin, B....................................................P403 Jeong, E.J.......................................P314, P514 Johne, S......................................................P303 Jones, T.......................................................P412 Joo, Y.S........................................................P502 Jun-Hsiang, L............................................P412 K Kaisar, M. A..........................STP438, STP439 Kanaly, R.....................................................P426 Kang, KK.....................................................P514 Karanth, S..................................................P504 Kaufman, L................................................P206 Kauss, A......................................................P118 Kaweeteerawat, C..............................STP436 Kearney, K..................................................P427 Kiertscher, S........................................ GFP117 Kim, D..........................................................P425 Kim, YB........................................................P514 Kogel, U......................................................P308 Kopf, R.........................................................P200 Korgaonkar, C..........................................P515 Kostadinova, R.........................................P309 Kramer, J....................................................P207 Kretser, A....................................................P302 Krishan, M..................................................P302 Kruhlak, N..................................................P205 Kuczaj, A.....................................................P417 Kuehn, D....................................................P309 Kulkarni, R.................................................P310 Kumer, SC..................................................P428 Kustermann, S..........................................P506 Kwok, S.......................................................P421 L Laffon, B................................................ IGP319 Lalayeva, N................................................P409 Lambright, C.............................................P517 Lansdell, T............................................ GFP118 Lanzicher, T.......................................... IGP441 Lapinskas, P. J...............................P509, P510 ABSTRACTS Faggioni, R................................................P504 Federov, N.................................................P207 Felx, M.........................................................P431 Festag, M...................................................P200 Fishbein, M...........................................STP318 Foote, A......................................................P404 Forget, J......................................................P434 Forster, R...........................P413, P414, P415, ...................................................P422, P423 Forster, J.....................................................P428 Fossa, A.......................................................P427 Fossey, S.....................................................P201 Foster, R..........................................P110, P111 Foster, K......................................................P512 Foster, P......................................................P517 Freebern, W...............................................P508 Freeman, M...............................................P103 Frentzel, S.........................P303, P308, P309, ...................................................P311, P417 Frost, G...........................................P509, P510 Fukuzaki, K....................................P408, P409 Furr, J...........................................................P517 Graff, C........................................................P201 Graham, A.....................................P110, P111 Grambo, B..................................................P403 Gray Jr., L. E...............................................P517 Groom, S........................................P422, P423 Gu, H............................................................P515 Guedj, E......................................................P309 Guss, V.........................................................P515 Guzman, R.................................................P503 th 35 American College of Toxicology Annual Meeting 2014 ABSTRACTS Poster abstracts are indicated with letters preceding the poster number: Poster (P), Student Travel Award (STP), North American Graduate Fellowship (GFP), and International Travel Grant (IGP). Laureano, R. T...........................................P429 Lavallée, S..................................................P424 Lawlor, T.........................................P310, P418 Learn, D......................................................P416 Lee, B.S........................................................P314 Lee, H.S.......................................................P502 Leroy, P........................................................P309 Lewis, A......................................................P511 Li, M.............................................................P105 Li, R..............................................................P508 Li, J...............................................................P512 Lin, Z............................................................P105 Lise, B..........................................................P420 Liu, J.........................P103, P104, P400, P425 Liu, R.......................................................STP436 Lo, A.............................................................P407 Lookingland, K.J................................ GFP118 Love, R............................................P408, P409 Luo, Y...........................................................P201 Lv, W.............................................................P410 M MacGregor, J.............................................P432 MacGuire, J................................................P512 Madsen, T.......................................P104, P400 Maeda, A....................................................P426 Majeed, S.......................................P309, P417 Makori, N......................................P408 , P409 Maneval, D. C................................P509, P510 Mangipudy, R...............................P407, P512 Manning, R....................................P408, P409 Mansell, P...........................P419, P421, P428 Marcotte, E................................................P424 Marescotti, D............................................P303 Martin, F........................................P309 , P312 Martinelli, V.......................................... IGP441 Maruf, A.A.............................................STP519 Maruñak, S.L........................................ IGP115 Mathis, C............................P303, P308, P309 Matulka, M. R............................................P203 McGill, M. R...............................................P428 McIntyre, B................................................P517 McKeon, M....................................P310, P418 McPherson, S................................P108, P410 Mendonça, A.T. .......................................P107 Meghazzi, M. S.........................................P422 Meredith, J................................................P515 Merg, C.......................................................P309 Messinis, D. E............................................P308 Mestroni, L........................................... IGP441 Micheletto, R............................................P426 Mikkelsen, S. R.........................................P429 Miller, D.................................................STP520 Mishra, R....................................................P500 Misner, D....................................................P118 Mohr, N. S..................................................P506 Monteiro-Riviere, N................................P106 Moon, K.S......................................P314, P514 Moore, C............................................... GFP442 Morales, A..................................................P203 Morford, L..................................................P429 Morikawa, Y...............................................P306 Mornagui, B..............................................P300 Morse, M....................................................P109 Mundhe, A.................................................P301 Mura, F........................................................P206 Murphy, B..................................................P407 Musinipally, V...........................................P118 Myatt, G..........................................P204, P205 N Nagata, R.......................................P408, P409 Naik, P....................................STP438, STP439 Navratil, N..................................................P403 Ngo, T..........................................................P118 Nicholson, S..............................................P504 Nordlund, M.............................................P417 O O'Brien, P...............................................STP519 Obejero-Paz, C.........................................P207 Orsini, M.....................................................P512 P Pai, R............................................................P118 Palate, B..........................................P110, P111 Pandiri, A...................................................P313 104 Pandit, S.....................................................P301 Panzica, J....................................................P512 Paranjpe, M...............................................P433 Park, S.J.......................................................P314 Peachee, V.................................................P402 Peddada, S................................................P313 Pei, H...........................................................P410 Peichoto, M.E....................................... IGP115 Peitsch, M.............P303, P305, P308, P309, .......................................P311, P312, P417 Perpetua, M..............................................P513 Philip, B.......................................................P508 Phillips, B....................................................P305 Phonetapswath, S..................................P432 Piccotti, J....................................................P429 Piehl, M.......................................................P405 Pilcher, G....................................................P515 Polhamus, K..............................................P401 Porto, B.................................................. IGP319 Pouliot, M. M................................P413, P415 Poussin, C..................................................P308 Prasad, P................................................STP318 Prasad, S...............................STP438, STP439 Pratt, L.............................................P405, P406 Prefontaine, A..........................................P516 Printz, M. A....................................P509, P510 Puschner, B.......................................... GFP442 Q Qiao, M.......................................................P315 Qin, S...........................................................P435 R Ramani, T...................................................P412 Ramesh, M.................................................P113 Rana, S.V.S............................................ IGP116 Rana, K................................................... IGP116 Rao, G..........................................................P508 Rashid, M...................................................P500 Raut, S. K............................................... IGP320 Ravuri, K.....................................................P200 Rebelatto, M.............................................P505 Ren, T...........................................................P315 Ren, S..........................................................P505 th 35 American College of Toxicology Annual Meeting 2014 Poster abstracts are indicated with letters preceding the poster number: Poster (P), Student Travel Award (STP), North American Graduate Fellowship (GFP), and International Travel Grant (IGP). S Saiakhov, R................................................P202 Sajja, R. .................................................STP438 Salerno, M. ...............................................P403 Sampson, H. .............................................P517 Sánchez, M.N. ..................................... IGP115 Sanderson, T.............................................P515 Santos, T.A.N.............................................P107 Santostefano, M......................................P503 Sarathchandra, G........................P101, P501 Sarron-Petit, C..........................................P200 Sasidharan, A. .........................................P106 Savidge, C..................................................P412 Sbaizero, O........................................... IGP441 Schlage, W. K............................................P309 Schlink, S....................................................P103 Schmitt, T. M.............................................P428 Schnapp, S................................................P425 Schreiter, K................................................P511 Sedykh, A...................................................P202 Selby, C.......................................................P104 Shakir, H.....................................................P316 Sharma, D..................................................P305 Sharma, S..............................................STP317 Shockley, K................................................P313 Sills, R..........................................................P313 Silva, S.................................................... IGP319 Singer, T......................................................P506 Singh, R. R.............................................STP318 Smalley, J...................................................P512 Smith, M.....................................................P402 Smith, S. Y..................................................P431 Snyder, C....................................................P118 St-Jacques, R.............................................P420 Stankowski, L...........................................P310 Stavitskaya, L............................................P205 Stoffel, R.....................................................P201 Stoute, M...................................................P423 Stricker-Krongrad, A.................P103, P104, ...................................................P400, P425 Strock, C.....................................................P435 Stump, D....................................................P402 Suarez, F.....................................................P307 Sugarman, B. J.............................P509, P510 Sun, Q..........................................................P512 T Taberner, E.................................................P403 Talikka, M...................................................P312 Tavcar, R.........................................P422, P423 Teibler, G.P............................................ IGP115 Teixeira, J.P............................................ IGP319 Tellez Cruz, A............................................P103 The, T...........................................................P316 Thompson, C............................................P508 Tian, Y..........................................................P201 Tirmenstein, M.........................................P407 Tohamy, A. F..............................................P518 Tong, W.......................................................P306 Torous, D........................................P310, P432 Tovcimak, A...............................................P201 Travlos, G...................................................P517 Troese, M........................................P405, P406 Troncy, E.................P411, P413, P414, P415 Tyszkiewicz, C..........................................P404 U Uehara, T....................................................P306 Ulm, S..........................................................P203 Uppal, H.....................................................P118 Upreti, V......................................................P503 V Valera, I..................................................STP318 105 Valerio Jr., LG............................................P304 van der Toorn, M.....................................P311 Varela, A.....................................................P431 Vargas, H....................................................P503 Veljkovic, E................................................P305 Veneziale, R...................................P509, P510 Verma, Y................................................. IGP116 Verma, N...............................................STP317 Vézina, M...................................................P430 Vuillaume, G.............................................P309 W Wang, Y.......................................................P306 Wang, B......................................... P508, P515 Wasko, M....................................................P206 Weese, J......................................................P315 Wehr, R........................................................P511 Wheeler, J..................................................P508 White, A......................................................P205 White, D..........................................P400, P425 White Jr., K.................................................P402 Whitney, K.................................................P201 Wichmann, J.............................................P506 Wicks, J.......................................................P103 Wilson, C. E................................................P511 Wilson, I. D.................................................P511 Wilson, V.....................................................P517 Wise, S.........................................................P430 Wuermlin, C..............................................P206 X Xiang, Y.......................................................P309 Xie, Y............................................................P428 Xu, Y.............................................................P310 Y Yamashita, H.............................................P313 Yan, J...........................................................P306 Yan, L...........................................................P504 Yang, X........................................................P108 Yang, B........................................................P502 Yeager, R....................................................P201 Yeager, R.P..................................................P304 You, J.S........................................................P502 Young, R.........................................P310, P418 ABSTRACTS Renna, S.....................................................P103 Rezg, R........................................................P300 Rezk, H. M. H.............................................P518 Rider, C.......................................................P313 Riley, R. J. R................................................P511 Riviere, J.........................................P105, P106 Robbie, G...................................................P505 Roche, B.....................................................P427 Rodrigues, C.R......................................... P107 Rojko, J....................................................... P505 Rose, R........................................................P409 Roth, M................................................. GFP117 Rousselle, S...............................................P103 Ryan, P.............................................P504, P505 th 35 American College of Toxicology Annual Meeting 2014 Poster abstracts are indicated with letters preceding the poster number: Poster (P), Student Travel Award (STP), North American Graduate Fellowship (GFP), and International Travel Grant (IGP). Z ABSTRACTS Zabka, T......................................................P118 Zhou, T........................................................P108 Ziegelhofer, T............................................P513 106 107 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory Exhibit Hall Hours: Monday, November 10 9:30 AM–12:00 Noon 2:00 PM–7:00 PM ACT Member Lounge 1821 Michael Faraday Dr., Suite 300 Reston, VA 20190 Contact: Nancy Rollman Tuesday, November 11 8:00 AM–4:30 PM Grand Cypress Ballroom Tel: 703.547.0875 Fax: 703.438.3113 Email: [email protected] Website: www.actox.org All participants are welcome to stop by the ACT Member Lounge to learn more about the College's educational programs and webinars, view membership benefits, update your interACT profile, view the ACT's scientific journal, the International Journal of Toxicology, and more. Advinus Therapeutics Limited EXHIBIT INFO 21-22, Phase II, Peenya Industrial Area Bangalore, Karnataka 560 058 India Contact: Ms. Y R Sapna 504 Tel: +91 80 28394959 Fax: +91 80 28394015 Email: [email protected] Website: www.advinus.com Advinus is an integrated Development Center promoted by the TATAs catering to pharma & Agro sectors. Located in India, Advinus is a 22-yr GLP-accredited & AAALAC-certified lab with integrated CMC, DMPK, Safety Pharmacology & Toxicology capabilities. We have a track record of submitting 30 INDs (USFDA -11) & >6000 standalone toxicology studies to global regulatory bodies. We have conducted >50 carci studies including Transgenic mouse models. We comply with ICH, EPA, EEC, OECD, CIPAC guidelines. Alliance Pharma, Inc. 17 Lee Blvd. Malvern, PA 19355 Contact: Bingbing Feng 218 Tel: 610.608.5917 Fax: 610.296.3153 Email: [email protected] Website: www.alliancepharmaco.com As a CRO, Alliance Pharma specializes in GLP and non-GLP studies for preclinical and clinical programs in the following areas: small molecule LC-MS/MS bioanalysis of drugs, biomarkers and agrochemicals, large molecule immunoassays to support PK, immunogenicity studies and biomarker analysis, cell based assays, drug metabolism and pharmacokinetics, and sample storage. Antech GLP 600 Airport Blvd., Suite 500 Morrisville, NC 27560 Contact: Susan St. Clair 319 Tel: 253.277.0811 Email: [email protected] Website: www.antechglp.com Antech Diagnostics GLP offers a full-service, Good Laboratory Practice compliant, clinical pathology reference laboratory performing hematology, chemistry, urinalysis, coagulation, immunoassays, hormone analysis, and esoteric tests. Antech now offers veterinary clinical trial testing and non-GLP studies. 108 109 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory BASi (Bioanalytical Systems, Inc.) 2701 Kent Avenue West Lafayette, IN 47906 Contact: Philip Downing 310 Tel: 765.463.4527 Email: [email protected] Website: www.basinc.com BASi provides world-class research to the pharmaceutical industry. We offer an extensive array of contract research services and manufacture specialty scientific instrumentation. Our non-clinical/GLP toxicology services include IND-enabling studies, non-clinical animal studies, and pathology services. Battelle401 505 King Avenue Columbus, OH 43201 Contact: Philip Downing Tel: 800.201.2011 Email: [email protected] Website: www.battelle.org Battelle provides GLP compliant, nonclinical services to the pharmaceutical industry and government agencies. Our capabilities include safety and efficacy testing; GLP toxicology, pharmacology; pathology; analytical/bioanalytical chemistry; biomarker discovery, method/assay development & validation. Battelle is the world’s largest independent nonprofit research organization, specializing in Life Sciences Research. Bio Medic Data Systems, Inc. EXHIBIT INFO 1 Silas Road Seaford, DE 19973 Contact: Geoff Hunt 317 Tel: 302.628.4100 Fax: 302.628.4110 Email: [email protected] Website: www.bmds.com Bio Medic Data Systems offers an electronic RFID solution for accurate animal identification. Our injectable transponders can display ID and body temperature. They can be paired with our advanced reader systems to display animal IDs as well as collect data from other connected devices; calipers and scales. The readers can work as independent cordless devices or connected to a computer for real time transfer. We are available in the USA, Europe and Asia to provide support and expert advice. Bioo Scientific 7050 Burleson Road Austin, TX 78744 Contact: Dawn Obermoeller 506 Tel: 512.707.8993 Email: [email protected] Website: www.biooscientific.com Bioo Scientific, an Austin, TX based biotechnology company, offers a complete line of kits for preclinical analysis including highthroughput, innovative assays for in vivo toxicity testing. These assays are ideal for assessing damage to liver, heart, kidneys and other organs. Bioo Scientific also offers preclinical services designed to meet your specific needs. BioReliance Corporation 14920 Broschart Road Rockville, MD 20850 Contact: Scott Hickman 300 Tel: 301.738.1000 Email: [email protected] Website: www.bioreliance.com BioReliance is a leading contract services company in the area of product safety. We specialize in genetic toxicology screening and GLP assays, as well as transgenic mouse carcinogenicity testing. BioReliance has all the experience and expertise needed to design and execute a toxicology testing program to meet your needs. 110 Leading Journals from SAGE JOURNAL OF VETERINARY DIAGNOSTIC INVESTIGATION vet.sagepub.com ISSN: 0300-9858 Veterinary Pathology OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF VETERINARY LABORATORY DIAGNOSTICIANS, INC. jvdi.sagepub.coMsISSN: 1040-6387 Toxicologic Pathology ISSN: IISSN ISS SSN SN: N: 0748-2337 07 0748 Mouse: Glomerular m Amyloidosis JCVP TOXICOLOGY and the British Society of Toxicological Pathology ISSN: 0192-6233 ECVP INTERNATIONAL JOURNAL OF The Official Publication of the Society of Toxicologic Pathology www.toxpath.org ACVP OfÄcial Journal of the American College of Toxicology Hepatocellular ular intra intracytoplasmic racyto oplasmic inclusions Journal of Feline Medicine and Surgery Official Journal of the International Society of Feline Medicine and the American Association of Feline Practitioners Clinical Practice http://jfms.com www.sagepub.com 1148231 111 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory BTS Research PO Box 910418 San Diego, CA 92191 Contact: Sami Abunadi 103 Tel: 858.605.5882 Fax: 858.605.5916 Email: [email protected] Website: www.btsresearch.com BTS Research, previously BioTex Sciences and it's subsidiary Bio-Quant, Inc., is your premiere In Vitro & In Vivo preclinical CRO offering research & development services from discovery to IND in small and large molecules in areas of oncology, inflammation, metabolic diseases in rodents, guinea pigs, rabbits, dogs, pigs & NHP's. Calvert Laboratories 130 Discovery Drive Scott Township, PA 18477 Contact: Leslie Maas 312 Tel: 570.586.2411 Fax: 570.586.3450 Email: [email protected] Website: www.calvertlabs.com Calvert is a highly respected and experienced CRO providing a complete spectrum of nonclinical safety studies since 1969 to the pharmaceutical, biotechnology and chemical industries. Services include Acute to Chronic Toxicology, DART studies, General and Safety Pharmacology, Telemetry, Immunology, Immunotoxicology and Pharmacokinetic/ADME. If you are looking for a highly personalized level of flexible, highly-communicative and responsive service, then Calvert is your laboratory of choice. ChanTest Corporation 14656 Neo Parkway Cleveland, OH 44128 Contact: Alissa Mague 215 Tel: 240.453.6331 Email: [email protected] Website: www.chantest.com EXHIBIT INFO ChanTest provides GLP/non-GLP preclinical safety testing for hERG and other cardiac ion channels, APD testing on stem cellderived cardiomyocytes, and in vitro and in vivo QT Prolongation Assays. The company offers the largest panel of ion channel targets and has been recognized as “the most trusted services company” in independent surveys. Charles River 251 Ballardvale Street Wilmington, MA 01887 Contact: Jessica Janiak 301 Tel: 781.222.6548 Email: [email protected] Website: www.criver.com Charles River provides essential products and services to accelerate research and drug development efforts. With flexible solutions, accelerated timelines and customized approaches, we provide clients with exactly what they need to improve and expedite the discovery, early-stage development and safe manufacture of new therapies every step of the way. CiToxLAB101 445 Armand-Frappier Boulevard Laval QC H7V 4B3 Canada Contact: Francois Duguay Tel: 450.973.2240 Email: [email protected] Website: www.citoxlab.com CiToxLAB (sites in France, Canada, Denmark, and Hungary) offers a wide range of GLP-nonclinical and specialty safety services in large and small molecules. CiToxLAB provides expertise in toxicology (general, infusion, inhalation, ocular, dermal, DART, immunotoxicology, carcinogenicity, transgenic, genetic, in vitro, chemical, agrochemical) and in specialty areas (genomics, safety pharmacology, radiation, DMPK). CorDynamics, Inc. 2242 W. Harrison St Chicago, IL 60612 Contact: Peter Senese 200 Tel: 312.421.8876 Email: [email protected] Website: www.cordynamics.com CorDynamics is a preclinical contract research laboratory and consulting group focused on examining the cardiovascular effects of emerging drug candidates with an emphasis on quality, affordability, flexibility, and reliability. 112 113 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory CRC Press - Taylor & Francis Group LLC 315 6000 Broken Sound Parkway NW, Suite 300 Tel: 561.994.0555 Boca Raton, FL 33487 Contact: Charmaine Lowe Email: [email protected] Website: www.crcpress.com CRC Press – Taylor & Francis Group is a global publisher of print and electronic books for medical, scientific and technical communities. Visit our booth #315 to browse our new and bestselling publications in toxicology and take advantage of convention discounts. Register for email alerts at www.crcpress.com. CXR Biosciences Ltd. 2 James Lindsay Place Dundee Scotland DD15JJ United Kingdom Contact: John McManus 212 Tel: +44 1382 432163 Fax: +44 1382 432153 Email: [email protected] Website: www.cxrbiosciences.com CXR Biosciences is a unique CRO combining innovative and standard mechanism-based approached to improve compound development or resolve toxicity issues. We are experts in mechanistic and investigative toxicology and ADME with emphasis on the prediction of animal to human data. Our portfolio includes: toxicogenomics, screening, and mechanistic toxicology protocols. Cyprotex216 313 Pleasant St. Watertown, MA 02472 Contact: Sam Verla Tel: 617.600.4300 Fax: 617.812.0712 Email: [email protected] Website: www.cyprotex.com EXHIBIT INFO Cyprotex is an AIM-listed company (CRX) with headquarters in Macclesfield, UK and laboratories in Watertown, MA, USA (Apredica). The combined company is the leader in predictive toxicology and ADME research, offers several proprietary technologies (CellCiphr, CloePK, gADMETM) and prides itself on custom assay development and fast turnaround times. Data Sciences International (DSI) 119 14th Street NW St. Paul, MN 55112 Contact: Jennifer Seidl 400 Tel: 651.481.7400 Email: [email protected] Website: www.datasci.com DSI offers preclinical physiological monitoring solutions for respiratory, CV, and CNS applications involving acute or chronic studies. Products include data collection and analysis systems, hardwired amplifiers, implantable telemetry, infusion pumps, JET external telemetry, glucose monitoring and respiratory chambers. New offerings include Validation, Surgical and Data Analysis Services. Offices throughout Europe, USA, and Asia provide support and expertise. DRIK107 865 Research Parkway, Suite 415 Oklahoma City, OK 73104 Contact: Kumar Sripathirathan Tel: 405.384.8580 Email: [email protected] Website: www.atdrik.com DRIK, as the name implies shines light on your discovery advancing your innovation. Our focus is on pharmacology and toxicology services for both small and large molecules. DRIK facilitates DMPK studies, both in vivo and in vitro, by conducting early ADME studies to identify lead candidates and their bio-distribution for IND filings. We specialize in 3D slice culture. We also provide PK/PD experiments supporting discovery, pharmacology, and toxicology and compound development strategy. We assess drug safety needs and save our clients' time and money to make safer drugs. 114 Vet Path Services, Inc. Vision • Pride • Service Fully Compliant with GLP’s Histology Laboratory (Paraffin & Plastic) Nine (9) Board-Certified (ACVP) Pathologists Histopathology Evaluation & Reporting Pathology Expert Reports and White Papers Pathology Peer Review Pathology Working Groups Clinical Pathology Interpretation Archiving Vet Path Services, Inc. 6450 Castle Drive Mason, OH 45040 [email protected] www.vetpathservicesinc.com Over 20 years in business with 35+ years combined experience in toxicology consulting. Consulting • Drug • Device • Combination products • Study Placement • Regulatory guidance, writing & filing • Risk Assessment for drugs & medical devices including product contamination, impurities and degradants, leachables and extractables & (Q)SAR Training • (Q)SAR • Drug and Device Development • Combination products • Application of Guidance • Customizable Audits • GLP analytical & preclinical • cGMP No project is too small, able to accommodate urgent requests learn more and meet our team @ www.gadconsulting.com 115 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory emka TECHNOLOGIES, Inc. 307 Annandale Road, Suite 203 Falls Church, VA 22042 Contact: Virginie Brechet 407 Tel: 703.237.9001 Fax: 703.237.9006 Email: [email protected] Website: www.emkatech.com emka TECHNOLOGIES specializes in software and hardware products for preclinical research. We offer a complete range of products for CNS, CV, Pulmonary research, and dedicated services for data analysis and system validation. New and featured products include: - easyMATRIX V2, a direct hardware bridge to implantable telemetry systems.- emkaPACK 4G, a smaller yet more capable version of our renowned non-invasive telemetry. - flexiVENT, precision laboratory instruments for pulmonary research, by SCIREQ. EPL Archives 45610 Terminal Drive Sterling, VA 20166 Contact: Allison Tyler 413 Tel: 703.435.8780 Fax: 703.435.1330 Email: [email protected] Website: www.eplarchives.com EPL Archives, a globally recognized leader in the research archiving and biorepository field, provides secure GLP, cGCP and cGMP storage for research materials. EPL, Inc. EXHIBIT INFO PO Box 169 Sterling, VA 20167 Contact: Paul Sanders 306 Tel: 703.471.7060 Fax: 703.471.8447 Email: [email protected] Website: www.epl-inc.com Experimental Pathology Laboratories, Inc. (EPL) is the world’s largest independent provider of GLP-compliant toxicologic pathology services. Since 1971, EPL has provided necropsy and histology support, pathology evaluation and consultation including pathology peer review and organizing Pathology Working Groups (PWG) for industry and government clients. We are able to customize our services to meet our clients’ specific scientific, regulatory, and management objectives. Experimur211 4045 S. Morgan Street Chicago, IL 60609 Contact: Farah Denahan Tel: 773.254.2700 (Ext. 232) Fax: 773.254.2723 Email: [email protected] Website: www.experimur.com Experimur is a full-service CRO with extensive capabilities in the conduct of preclinical toxicology studies. Place your programs with Experimur and experience the superiority of a team skilled with decades of expertise, top quality and unmatched service. We invite you to visit our spectacular, custom-built, AAALAC-accredited, 54,000 sq ft, state-of-the-art facility and vivarium, including extensive in-house support services for histology, diagnostic pathology, clinical pathology, and analytical chemistry. Gentronix Limited BioHub at Alderley Park Alderley Edge, Cheshire SK10 4TG United Kingdom Contact: Steve Beasley 512 Tel: +44 0 1625 238 700 Fax: +44 (0) 1625 238 701 Email: [email protected] Website: www.gentronix.co.uk Gentronix is a specialist genetic toxicology company providing services and products worldwide. We work to tight deadlines to deliver quality results with expert follow up from product discovery and early screening through to GLP compliant OECD in vitro genetic test battery studies. You can find details of all of our products and services on our website: www.gentronix.co.uk or email us at [email protected] or come talk to us at Booth #512. 116 NO TWO ARE EVER THE SAME. At Covance, we understand that every biologic molecule is uniquely developed. So we’ve created dedicated biologics teams to expertly partner with you. Together, we’ll address the complexities of your molecule to transform data into insight that expedites your molecule’s development. Over the last few years we’ve proudly completed thousands of biologics studies and more than 40 biologics IND/CTA packages. Discover how we can help transform your biologic with Solutions Made Real™. TO LEARN MORE CALL The Americas +1.888.COVANCE | Europe/Africa +00.800.2682.2682 Asia/Pacific +800.6568.3000 | Or go to Covance.com/biologics COVANCE is an independent, publicly held company with headquarters in Princeton, New Jersey, USA. COVANCE is the marketing name for Covance Inc. and its subsidiaries around the world. © Copyright 2014. Covance Inc. 117 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory Histo-Scientific Research Laboratories (HSRL) 5930 Main Street Mt. Jackson, VA 22842 Contact: Patty Schwartz Tel: 540.477.4440 313 Email: [email protected] Website: www.hsrl.org HSRL’s necropsy, histology, pathology, and archiving services are known for expertise, quality and efficiency. We support GLP studies in a responsive and cost-effective manner. Necropsy services are provided at your facility. Our board-certified pathologists have extensive experience. HSRL Archives offers ambient, refrigerated and frozen storage. HistoTox Labs, Inc. 5541 Central Avenue, Suite 150 Boulder, CO 80301 Contact: Jon Bishop 207 Tel: 303.633.5401 Fax: 303.565.3764 Email: [email protected] Website: www.histotoxlabs.com HistoTox Labs is a GLP-compliant contract laboratory performing routine and specialized Histology, Immunohistochemistry and Pathology services for toxicity, arthritis, cancer and inflammation related studies. Additional services include decalcified bone techniques, antibody optimization, full slide scanning, digital image analysis, special stains and frozen techniques. Huntingdon Life Sciences/ Harlan P. O. Box 2360, Mettlers Road East Millstone, NJ 08875 Contact: Melissa Zeier 416 Tel: 732.873.2550 Fax: 732.873.8899 Email: [email protected] Website: www.huntingdon.com EXHIBIT INFO Huntingdon Life Sciences provides complete preclinical drug development services for large and small molecules. With sixty years experience, we are committed to our customers' needs through quality service and scientific expertise. Our services: Toxicology, DART, safety pharmacology, DMPK, genetic tox, immunoassay, bioanalytical CMC, cell-based assays, flow cytometry, environmental risk assessment. IDEXX BioResearch One IDEXX Drive Westbrook, ME 04092 Contact: Melissa Terrano 302 Tel: 207.556.8628 Fax: 207.556.2005 Email: [email protected] Website: www.idexxbioresearch.com IDEXX BioResearch delivers research analyzers and analytical testing services for laboratory animals to the global biomedical research community. Our animal health monitoring, genetic testing, preclinical research services and cell-line quality assurance simplify your ability to conduct quality research. IIT Research Institute (IITRI) 10 W 35th Street Chicago, IL 60616 Contact: Jennifer Van Dinther 405 Tel: 312.567.4911 Fax: 312.567.4924 Email: [email protected] Website: www.iitri.org IIT Research Institute (IITRI) brings together the best of both worlds with the comprehensive, service offering of a large CRO and the personalized collaborations of a small CRO. Specialties include GLP toxicology and safety testing to support IND submissions, cancer drug discovery and development, inhalation toxicology, and efficacy and safety testing of vaccines and antiinfective drugs. 118 Patricia Frank & Associates, Inc. Assisting the Pharmaceutical Industry since 1993 Congratulations to the ACT on another successful Annual Meeting [email protected] 847.864.6528 Are you looking for a Toxicology Consultant? Search through our comprehensive database of Toxicologists around the country. RTC is the oldest and largest consortium of independently practicing toxicologists dedicated to solving the problems of clients on the basis of good science and the expertise gained through many years of industry, government and/or academic experience. Collectively, we work in the following industries: •Pharmaceuticals (drugs) •Chemical •Foods •Consumer Products •Medical Devices •Legal – Expert Witnesses •Environmental Technology / Pesticides •Occupational Exposure / Safety http://www.toxconsultants.com/ Advertisement sponsored by Gad Consulting Services www.gadconsulting.com 119 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory ImmunoTox, a Division of AIBioTech, LLC 601 Biotech Drive Richmond, VA 23235 Contact: John Stradling Tel: 804.648.3820 204 Email: [email protected] Website: www.immunotox.com ImmunoTox, Inc. specializes in studys evaluating the effects of drugs, chemicals, and physical agents on the immune system. In Vivo and In Vitro studys are carried out in well-equipped laboratories specifically designed for immunotoxicological studies. Indigo Biosystems 7820 Innovation Blvd. Suite 250 Indianapolis, IN 46278 Contact: Sasha Bannister 415 Tel: 317.493.2400 Email: [email protected] Website: www.indigobio.com Indigo’s ASCENT software supports that scientist by applying sophisticated software automation to the processing, reviewing and reporting of quantitative mass spectrometry applications. ASCENT dramatically accelerates the overall workflow by highlighting problematic results and outliers for expert review. A rigorous peak processing algorithm reveals the true chromatographic data in complex samples, and a system of automated quality assurance checks provides an overarching and unified quality standard across the lab. The ASCENT system can operate with a wide variety of manufacturer instrumentation, through a web browser, providing a comprehensive data automation suite in which quantified results are available anywhere, and delivered quickly, consistently and reliably. ITR Laboratories Canada Inc. EXHIBIT INFO 19601 Clark Graham Boulevard Baie D Urfe (Montreal) QC H9X 3T1 Canada Contact: Vittoria Badalone 411 Tel: 514.457.7400 Email: [email protected] Website: www.itrlab.com ITR Laboratories, a midsized global CRO, provides a comprehensive array of nonclinical services for the biopharmaceutical industry in support of IND and later-stage regulatory submissions. ITR offers GLP and non-GLP studies in general, inhalation, infusion, genetic and reproductive toxicology, carcinogenicity, and safety pharmacology that are compliant with current international guidelines. KCAS Bioanalytical Services 12400 Shawnee Mission Parkway Shawnee, KS 66216 Contact: Terry Osborn 402 Tel: 847.778.0522 Email: [email protected] Website: www.kcasbio.com KCAS, they know bioanalysis. Their strength is their ability to support your drug development program from non-GLP research through NDA. For more than 30 years their boutique lab has provided a full-spectrum of bioanalytical services, including small molecule analysis, large molecule PK and immunogenicity analysis and predictive toxicology biomarker analysis. Learn more about KCAS and how they can support your toxicology program by visiting the KCAS website. 120 Be part of a scientific community working to advance systems toxicology Network Verification Challenge 2 is now open. sbv IMPROVER stands for Systems Biology Verification combined with Industrial Methodology for PROcess VERification in Research. It is a robust methodology that verifies systems biology approaches using double-blind performance assessment and applies the wisdom of crowds to solve scientific challenges. Call for Action Classical peer review system Are the conclusions supported by the results shown in the publication? SCIENTIFIC QUESTION PUBLICATION DATA ANALYSIS PEER REVIEW CONCLUSIONS & CLAIMS REPRODUCIBILITY ISSUES DATA GENERATION ALL DATA MADE AVAILABLE Are the conclusions supported by the data? INDEPENDENT DATA ANALYSIS DISCUSSION OF RESULTS ROBUST/ TRUSTED/ REPRODUCIBLE CONCLUSIONS sbv IMPROVER • Join the sbv IMPROVER community and expand your network • Participate in the ongoing Network Verification Challenge 2 (February 2014 - April 2015) • Explore the networks at https://bionet.sbvimprover.com • Watch our online training videos and attend the free tutorials https://sbvimprover.com/challenge-3/videos https://sbvimprover.com/challenge-3/tutorials • Gain early access to high-quality, well-curated networks • Become a contributor in network biology for toxicology and drug and biomarker discovery www.sbvimprover.com Use smarter solutions to complement peer review with collaborative crowd-sourcing Network Verification Challenge at a Glance sbv IMPROVER Network Model Construction Boundary Definition + Nature YOU sbv IMPROVER YOU SCIENTIFIC COMMUNITY Online Crowd-Verification Jamboree Preparation Network Jamboree Network Dissemination Continuous Improvement Web-based platform Reputation System Scientific Seminars Further Enhancements Science JBC PNAS Publications Face to face meeting Best Contributors Academia Lung Disease Community Experts Pharma Team Network Building Systems Biologists University Students Biologists Feb 2014 Public Use of Scientifically Accepted Networks Publications Web-based platform Graduate Students Postdocs Evidence Summary Online Forum Moderation by experts Analysis Prioritization Webinars, Training = CausalBioNet Early Access to Networks BEL editor + General Access to Networks Scoring Leaderboard + April 2015 Researchers Students Teachers Pharma Toxicologists The sbv IMPROVER Network Verification Challenge (NVC) aims to verify and enhance existing biological network models. The NVC is expected to increase the networks’ value and promote their use in research applications such as drug discovery, personalized medicine and toxicological risk assessment. CONSENSUS May 2015 Mid 2015 Be part of a scientific community working to advance systems toxicology www.sbvimprover.com The sbv IMPROVER project, the website and the Symposia are part of a collaborative project designed to enable scientists to learn about and contribute to the development of a new crowd sourcing method for verification of scientific data and results. The current challenges, website and biological network models were developed and are maintained as part of a collaboration among Selventa, OrangeBus and ADS. The project is led and funded by Philip Morris International. For more information on the focus of Philip Morris International’s research, please visit www.pmi.com. 121 TM th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory Leadscope, Inc. 1393 Dublin Rd. Columbus, OH 43215 Contact: Michael Conley 208 Tel: 614.675.3768 Fax: 614.675.3732 Email: [email protected] Website: www.leadscope.com Leadscope produces high quality QSAR models and databases. Our Model Applier meets all the requirements of the ICH M7 Guidance on testing for impurities. In addition, our Model Applier produces an ICH M7 consensus prediction as used by the U.S. FDA. Our consensus prediction incorporates the statistical QSAR models; expert alerts results and experimental data. Lhasa Limited Granary Wharf House, 2 Canal Wharf Leeds, LS11 5PS United Kingdom Contact: Lorraine Bowe 417 Tel: +44 (0)113 394 6020 Fax: +44 (0)113 394 6099 Email: [email protected] Website: www.lhasalimited.org Lhasa Limited is an educational charity and leading supplier of expert knowledge based and statistical software and databases including; Derek Nexus: For predicting toxicity, Meteor Nexus: For predicting metabolic fate, Sarah Nexus: For predicting toxicity, Vitic Nexus: Toxicity database and management system, Zeneth: For predicting chemical degradation pathways. Lhasa Limited is an active research organisation with an enviable reputation for collaborative work and data sharing. We work closely with our members in the R&D of software for the chemical and bio-molecular sciences. Members include the world’s top 20 pharmaceutical companies, leading consumer products manufacturers, academics, regulatory bodies and government organisations. Lovelace Respiratory Research Institute (LRRI) EXHIBIT INFO 2425 Ridgecrest Drive SE Albuquerque, NM 87108 Contact: William Bechtold Tel: 505.348.9456 403 Email: [email protected] Website: www.lrri.org The Lovelace Respiratory Research Institute is a private nonprofit biomedical research organization. LRRI offers internationallyrecognized expertise in aerosol science, inhalation exposure technology, respiratory animal models, inhalation toxicology, pharmacokinetics, and clinical trials. Respiratory tract dosimetry is measured using both in vivo and in vitro techniques. Studies routinely performed under GLP standards. Marshall BioResources 5800 Lake Bluff Road North Rose, NY 14516 Contact: Nicole Navratil 318 Tel: 315.587.2295 Fax: 315.587.2109 Email: [email protected] Website: www.marshallbio.com Marshall BioResources provides quality, purpose-bred animals for biomedical research. We supply Marshall Beagles, ferrets, and mixed-breed mongrels and hounds globally. We are also the exclusive North American source of Göttingen Minipigs. Please visit us to discuss your nonrodent animal needs. Millar Inc. 6001-A Gulf Freeway Houston, TX 77023 Contact: Michelle Sanders 206 Tel: 832.667.7000 Fax: 713.714.8497 Email: [email protected] Website: www.millar.com The Millar Telemetry System provides high-resolution, physiologically accurate, long-term recordings for superior data measurements in real time. Unequaled signal quality allows for results in a shorter time frame. The device is fully implantable and recharges wirelessly. Measurable parameters include pressure, temperature, SNA, BP and tissue oxygen. 122 Toxicologic Pathology: Know What to Look for? Functions truly INDEPENDENTLY — unrestricted by Wall Street influences or large corporate bureaucracy, assuring objective, quality toxicologic pathology evaluation and consultation. High degree of sophistication and quality throughout, so every function is performed EXPERTLY — from highly trained and experienced pathologists and technicians to outstanding IT systems, state-of-the-art laboratories, and innovative quality procedures. Extraordinary RESPONSIVENESS to client needs — such as availability of client-site or in-house services, routine or highly specialized procedures, and expedited completion of projects. Large enough to meet your needs…….small enough to care. P.O. Box 169 Sterling, VA 20167-0169 Tel: 703.471.7060 Fax: 703.471.8447 www.epl-inc.com Independent. Expert. Responsive. 123 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory Moltox414 PO Box 1189, 157 Industrial Park Drive Boone, NC 28607 Contact: Ray Cameron Tel: 828.264.9099 Fax: 828.264.0103 Email: [email protected] Website: www.moltox.com Since 1986, Moltox has been providing CRO’s, QC departments and drug discovery scientists with reagents, bacterial strains, metabolic activation products and bacteriological media. As the leading manufacturer of products for use in the Ames Assay, Moltox distributes culture media, tester strains, S9 and positive controls through our partners worldwide. MPI Research 54943 N. Main Street Mattawan, MI 49071-9399 Contact: Katie Kridler 201 Tel: 269.668.3336 Fax: 269.668.4151 Email: [email protected] Website: www.mpiresearch.com MPI Research is a preclinical and early clinical CRO that provides discovery, surgery, safety evaluation, bioanalytical, and analytical services. We exceed expectations through consistency and quality, with a commitment to communication and innovation, delivering benefits throughout all phases of development. Learn how we can go beyond for you at http://www.mpiresearch.com MultiCASE Inc. 23811 Chagrin Boulevard, Suite 305 Beachwood, OH 44122 Contact: Roustem Saiakhov 510 Tel: 216.831.3740 Fax: 216.831.3742 Email: [email protected] Website: www.multicase.com EXHIBIT INFO MultiCASE provides powerful software and QSAR models developed through Research Collaboration (RCA) with the US FDA. Our software/models packages help scientists to assess the potential toxicity of pharmaceuticals, impurities, degradants and metabolites, complying with the ICH M7 Step 2 guideline on testing of impurities using statistical-based QSAR methodology. National Jewish Health 1400 Jackson Street Denver, CO 80228 Contact: David Trollinger 214 Tel: 303.398.1669 Fax: 303.270.2175 Email: [email protected] Website: www.njlabs.com The National Jewish Health Advanced Diagnostic Laboratories offer pre-clinical and clinical testing services under CAP/CLIA/ ISO15189 and FDA GLP. The Complement Laboratory offers the most comprehensive specialized test menu. Offering study design and data analysis based on a proven track record assessing complement with a variety of test systems and test articles. Our experience includes complement activation, cytokines assessment, ADA and immune complex formation for biologics, vaccines, particulars, devices, antibodies, nanoparticles and oligonucleotides. Optivia Biotechnology Inc. 115 Constitution Drive #7 Menlo Park, CA 94025 Contact: David Lustig 314 Tel: 650.324.3177 Email: [email protected] Website: www.optiviabio.com Optivia Biotechnology offers a suite of in vitro transporter assay services to assist drug development companies discover and design better drugs with improved therapeutic responses and safety profiles. Our clients and collaborators include pharmaceutical and biotechnology companies, research institutions and government agencies. 124 dedicated to innovation exceptionalscience nc ie sc gh ro u th s tie ili po WIL Research is dedicated to listening to your specific study requirements. Stop by booth 309 and together we’ll discuss the right approach for you. ib So tell us about your CRO needs. MedImmune leads a movement that demands more out of medicine. With one of the largest and most robust pipelines in the industry, we pioneer the future of science — creating advancements in biotechnology that have a true impact on health. ss TO YOUR EVERY WORD e. We’re hanging on tin g ne w Be sure to attend this continuing education course sponsored by WIL Research. cr ea Toxicology and Pathology of the Respiratory System Sunday, November 9 1:00 p.m. – 4:30 p.m. Let’s start the conversation at booth 309. We have listening down to a science. www.wilresearch.com www.medimmune.com © 2013 MedImmune. All rights reserved. 125 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory OtoScience Labs 401 Timbercreek Drive Jacksonville, IL 62650 Contact: Jeremy Turner 217 Tel: 217.883.1533 Email: [email protected] Website: www.otosciencelabs.com OtoScience Labs specializes in providing comprehensive preclinical contract research and consulting services related to ototoxicity. Our patented OtoCentrix technology helps streamline drug development and safety assessment by rapidly screening rodent models for hearing loss and tinnitus, helping to more efficiently identify toxicity by using the ear as a sentinel system. PDS Preclinical Data Systems, Inc. 100 Valley Road, Suite 204 Mt. Arlington, NJ 07856 Contact: Maro Schuster 307 Tel: 973.398.2800 (Ext. 19) Fax: 815.301.3115 Email: [email protected] Website: www.pds-europe.com PDS, Inc, has been helping accelerate drug development efforts with global clients in Pharma, CRO, Biotech, Academia and Regulatory fields for over 34 years. Our Ascentos™ Preclinical Suite of solutions and TranSEND™will enable your laboratory to complete your studies and prepare your data in SEND format. Stop by for a discussion and demonstration of our Ascentos and TranSend Solutions. Perceptive Instruments Ltd EXHIBIT INFO St. Francis House, Olding Road Bury St Edmunds IP33 3TA United Kingdom Contact: Frances Hall 419 Tel: +44 (0)1284 765566 Email: [email protected] Website: www.perceptive.co.uk Perceptive Instruments supplies image analysis and study management systems which integrate data acquisition, auditing and reporting for genetic toxicology assays. Our flagship products include Comet Assay IV, Sorcerer Colony Counter, Ames Study Manager and Cyto Study Manager. With over 20 years experience, we have a global reputation for delivering highly effective GLPcompliant solutions along with excellent customer service. PointCross Life Sciences, Inc. 1291 E. Hillsdale Blvd, Suite 304 Foster City, CA 94404 Contact: Ana Belo 119 Tel: 650.350.1900 Fax: 650.350.1903 Email: [email protected] Website: www.pointcrosslifesciences.com PointCross Life Sciences offers solutions to standardize, visualize and analyze nonclinical study data. Our DSIMS™ solution is used to review nonclinical studies and package data to comply with the FDA’s implementation of the CDISC SEND standard. It is installed as NIMS at the FDA for regulatory review of NDA and IND study data. We provide data standardization services to the FDA for sponsor submissions to help jump start the regulatory review process. This service is offered to global commercial clients. Companies use SDIS™, our cross-study analytics solution, for R&D purposes. PreClinical Research Services, Inc. 1512 Webster Court Fort Collins, CO 80524 Contact: Patti Waters 500 Tel: 970.232.1122 Fax: 970.232.1126 Email: patti.waters@ preclinicalresearch.com Website: www.preclinicalresearch.com PCRS offers GLP and non-GLP services to support pharmacokinetics, pharmacology, and toxicology studies in addition to osteoarthritis, vascular, and surgical models. Our expertise with purpose-bred small and large animal species allows us to efficiently support drug and medical device development. We provide rapid startup times and turnaround to manage our clients’ needs. 126 127 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory Product Safety Labs 2394 Highway 130, #E Dayton, NJ 08810 Contact: Genevieve Hutchins-Booker 305 Tel: 732.438.5100 (Ext. 262) Email: genahutchinsbooker@ productsafetylabs.com Website: www.productsafetylabs.com For over 40 years PSL has provided Toxicological Research, Analytical Chemistry and other testing services. PSL is able to assist their clients by providing data to support a variety of needs, including custom designed studies, product and discovery development, preclinical safety evaluations, product stewardship, regulatory compliances and risk assessment. PSL prides itself on its commitment to quality, offering competitive pricing and on-time reports delivered through a team of expert, professional staff. Quidel105 12544 High Bluff Drive, #200 San Diego, CA 92130 Contact: Kim Wilson Tel: 858.552.1100 Fax: 858.882.4131 Email: [email protected] Website: www.quidel.com Quidel® MicroVue® products are focused on delivering innovative research and diagnostic tools for identification, development, marketing and sale of biochemical bone markers, immune system monitoring and assays for the assessment of Complement activation. Many of these products are unique in nature and provide researchers and clinicians valuable scientific and diagnostic information. Looking to expand the methodical arsenal of Complement analysis in animals, Quidel marketed the Pan-Specific C3 Reagent kit (RUO product). Microwell kits, related products and core technologies are currently marketed directly and through distribution worldwide under the Quidel® and MicroVue® brands. Quintiles109 EXHIBIT INFO 4820 Emperor Boulevard Durham, NC 27703 Contact: Dominque Talbert Tel: 866.267.4479 Email: [email protected] Website: www.quintiles.com Quintiles (NYSE: Q), a Fortune 500 company, is the world’s largest provider of biopharmaceutical development and commercial outsourcing services. With a network of more than 30,000 employees conducting business in ~100 countries, we helped develop or commercialize all of 2013’s top-100 best-selling drugs on the market. Quintiles’ CardioCheck consists of a panel of in vitro assays that provide rapid insight into the cardiotoxic potential of drugs in pre- and early clinical stages of development. SAGE304 2455 Teller Road Thousand Oaks, CA 91320 Contact: Lisa Lamont Tel: 805.410.7239 Fax: 805.499.0871 Email: [email protected] Website: www.sagepub.com SAGE is a leading international publisher of journals, books, and electronic media for academic, educational, and professional markets. Since 1965, SAGE has helped educate a global community spanning a wide range of subject areas including business, humanities, social sciences, and science, technology, and medicine. SAI Infusion Technologies 276 Park Ave. Lake Villa, IL 60046 Contact: Steve Denault 412 Tel: 847.356.0321 Fax: 847.356.0382 Email: [email protected] Website: www.sai-infusion.com SAI Infusion Technologies designs and manufactures systems and components for preclinical infusion and sampling. SAI surgeons and scientists provide on-site comprehensive training for all of our systems including: Wireless-infusion management software (Axios), pumps, harnesses, access ports, jackets, swivels, catheters, and virtually everything else used in research involving infusion or sampling. 128 SOLVING WHAT MATTERS MOST IN TOXICOLOGY For more than 30 years, industries and government agencies alike have trusted Battelle to solve their most complex toxicology challenges. With expertise spanning dozens of interrelated scientific disciplines, premier test facilities, and objectivity as the world’s largest independent R&D organization, Battelle provides comprehensive toxicology solutions for pharmaceutical, biotechnology, medical device, agrochemical industries and government clients. To solve your most pressing challenges, Think Battelle first. 800.201.2011 ú [email protected] ú www.battelle.org 129 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory sbv IMPROVER 113 Email: [email protected] Website: www.sbvimprover.com The sbv IMPROVER project demonstrated that crowd sourcing is a viable strategy to verify scientific methods and concepts in an industrial context. sbv IMPROVER stands for Systems Biology Verification combined with Industrial Methodology for Process Verification in Research. This approach aims to provide a measure of quality control of industrial research and development by verifying the methods used. It is different from other scientific crowdsourcing approaches as it focuses on the verification of processes in an industrial context, and not just on basic questions regarding science. Visit us at booth #113 to learn more about the challenges. Seventh Wave 743 Spirit 40 Park Drive, Suite 209 Chesterfield, MO 63005 Contact: Jody DeBold 203 Tel: 636.519.4885 Fax: 636.519.4886 Email: [email protected] Website: www.7thwavelabs.com Seventh Wave is a laboratory with the strength and flexibility to maximize your lead development initiatives. We are uniquely qualified to provide you with the scientific expertise you demand and the advice and counsel your project deserves. Our highly collaborative approach means a close working relationship with you, and a measure of flexibility and responsiveness not found with other CRO’s. Simulations Plus, Inc. EXHIBIT INFO 42505 10th Street West Lancaster, CA 93534 Contact: Renee Bouche 408 Tel: 661.723.7723 Fax: 661.723.5524 Email: [email protected] Website: www.simulations-plus.com GastroPlus™ sets the standard for PBPK/PD modeling for different administration routes in humans and animals, plus population simulations and DDI capabilities. The ADMET Design Suite™ mines compound libraries, designs new molecules, and virtually screens structures for ADMET properties. DDDPlus™ and MembranePlus™ offer simulations of in vitro dissolution and permeability experiments. Sinclair Research Center, LLC P.O. Box 658 Columbia, MO 65205 Contact: Carrie Horton 219 Tel: 573.387.4400 Fax: 573.387.4404 Email: [email protected] Website: www.sinclairresearch.com Toxicology, DMPK, and Efficacy studies in 16 species including NHP, acute-chronic, IND enabling studies. TK/PK/ADME with nonradiolabeled compounds. Specialists in dermal, wound healing, ocular, and juvenile toxicology studies. Surgical Services: orthopedic, wound healing, vascular/GI access cannulas in large animals, and many pharmacology models. Animal Models: Type I/ II diabetes, inflammation, ocular, dyslipidemia, obesity, osteoporosis, transdermal delivery, melanoma, cardiovascular. Smithers Avanza 11 Firstfield Rd. Gaithersburg, MD 20878 Contact: Hope Aubin 202 Tel: 240.364.6360 Email: [email protected] Website: www.smithersavanza.com Smithers Avanza Toxicology Services is a GLP-compliant, AAALAC-accredited, USDA-registered, OLAW-assured, DEA-licensed facility located in Gaithersburg, Maryland. Our team plans and conducts safety assessment studies for pharmaceuticals (small molecules and biologics), nutraceuticals, vaccines, and agro and industrial chemicals. Our studies are conducted to the highest scientific standards and in compliance with regulatory requirements.We offer a proven track record of on time delivery of results and reports. 130 GO BEYOND Discovery. Surgery. Imaging. Progress. PreClinical Research Services, Inc. offers GLP and Non-GLP pre-clinical testing for the pharmaceutical and medical-device industries. Experimental Surgery Medical devices Proof of Concept Device V&V Labs Cardiovascular Devices GI/Orthopedic/Thoracic Osteoarthritis Medical imaging Fluoroscopy Angiography MPI Research is dedicated to bringing safer, more effective treatments to the world. With diverse routes of administration, advanced electronic data support, and experience with a wide variety of compounds, our Drug Safety experts go beyond to move your program through the development pathway. Ultrasound/Echo Toxicology Pilot/Acute/MTD/Subacute/Chronic Multiple species and administration routes Go beyond with us in booth 201 and receive a unique gift. Pharmacokinetics (PK/PD/TK) www.PreClinicalResearch.com 131 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory SNBL USA, Ltd. 6605 Merrill Creek Parkway Everett, WA 98203 Contact: Shelby Wilcox 406 Tel: 425.322.2420 Fax: 425.407.8601 Email: [email protected] Website: www.snbl.com SNBL USA offers a unique range of safety assessment services to fulfill its commitment to help free patients from suffering. Managed and operated by a team world renowned for its wide-ranging NHP expertise, SNBL USA offers programs ranging from regulatory tox to customized study designs in multiple drug classes (biologics, small molecules, oligonucleotides, vaccines). Specialized capabilities include DART, radiation, apheresis, TBI, immunotoxicology and carcinogenicity. Southern Research Institute 2000 Ninth Avenue South Birmingham, AL 35205 Contact: Freida Lindsey 213 Tel: 205.581.2000 Fax: 205.581.2044 Email: [email protected] Website: www.southernresearch.org Southern Research Institute is a preclinical contract research organization with over 50 years experience. Services include efficacy testing with special expertise in the areas of oncology and infectious diseases, supported by a complete range of in-house bioanalytical, PCR, immunology, clinical pathology and anatomic pathology facilities. Taconic Biosciences EXHIBIT INFO One Hudson City Center Hudson, NY 12534 Contact: Holly Jordan 514 Tel: 518.697.3900 Email: [email protected] Website: www.taconic.com Taconic Biosciences is a global provider of genetically modified mouse and rat models and services. As a full-service industry leader, founded in 1952, Taconic helps clients acquire, test, develop, breed, cryopreserve, prepare, and distribute highly relevant research lines worldwide. Whether you require custom genetically engineered, humanized or research-ready models, Taconic's scientists will partner with you to rapidly and efficiently obtain the high quality models needed for your discovery or preclinical programs. TSE Systems Inc. 186 Chesterfield Industrial Blvd. Chesterfield, MO 63005 Contact: Jens-Uwe Engler 404 Tel: 636.346.0144 Email: [email protected] Website: www.tse-systems.com TSE Systems develops, manufactures and markets sophisticated life science research instrumentation for preclinical cardiovascular, behavioral, metabolic, physiological and toxicological research since 1886. Featured product: STELLAR TELEMETRY next generation implantable telemetry (Pressure, Biopotentials (ECG/EEG/EMG/EOG), Activity, Temperature). NO receiver platforms needed. Unlimited options in research protocol setups with single/group housed animals monitored with only one receiver. Vet Path Services, Inc. (VPS) 6450 Castle Drive Mason, OH 45040 Contact: Chris Johnson 205 Tel: 513.469.0777 Fax: 513.469.2474 VetPath Services, Inc. is a contract histopathology lab. 132 Email: [email protected] Website: www.vetpathservicesinc.com Build your career and capabilities while transforming patients’ lives around the world for the better. When you embark on a journey to cure at Cubist, innovation is instinctive, collaboration is imperative and your opportunities are endless. Be the difference. See where you can go go with CUBIST Pharmaceuticals @CubistCareers cubist-pharmaceuticals www.cubist.jobs 133 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor Directory VivoPharm518 1214 Research Boulevard, Suite 1050 Hummelstown, PA 17036 Contact: Ralf Brandt Tel: 717.798.9990 Fax: 717.724.5399 Email: [email protected] Website: www.vivopharm.com VivoPharm is a successful and fast-growing contract research organization that provides integrated preclinical services in various disease areas to the biotechnology amd pharmaceutical sectors worldwide. We specialize in planning and conducting tailoried studies to guide with our experience your drug development program, starting from early compound selections and ending with a comprehensive set of in vitro and in vivo data reports, as needed for IND filings. WIL Research 1407 George Road Ashland, OH 44805 Contact: Katie Geitgey 309 Tel: 419.289.8700 Fax: 419.289.3650 Email: [email protected] Website: www.wilresearch.com WIL Research is a global CRO dedicated to listening to customer needs. They custom design product safety toxicological research, bioanalytical, and formulation services for pharmaceutical, biotechnology, chemical, agrochemical, and food companies. With approximately 1,200 scientific, technical, and support personnel located throughout the world, WIL Research offers technological expertise, flexible study design, and quality results. Worldwide Primates, Inc. EXHIBIT INFO P.O. Box 971279 Miami, FL 33197 Contact: John Resuta 502 Tel: 305.378.9585 Fax: 305.232.3838 Email: [email protected] Website: www.wwprimates.com An importer, distributor and breeder of premium quality nonhuman primate models as well as nonhuman primate bio-products. We offer cynomolgus from multiple country origins, rhesus, Caribbean Green monkeys, marmosets, squirrel monkeys, and baboons. We have two facilities in the US, both CDC approved quarantine centers. We provide logistical support for transport, consulting, and delivery to clients. Please contact us for more information on our NHP models. WuXi AppTec 2540 Executive Drive St. Paul, MN 55120 Contact: Mary Trelstad 316 Tel: 651.675.2801 Fax: 651.675.2005 Email: [email protected] Website: www.wuxiapptec.com WuXi AppTec partners with our customers to provide a wide range of IND/NDA enabling toxicology and laboratory services that meet global regulatory standards. As part of our integrated portfolio offering, our preclinical services are designed to shorten the time and lower the cost of drug and medical device R&D. Xenometrics LLC 17745 Metcalf Avenue Stilwell, KS 66085 Contact: Sara Quade 303 Tel: 913.850.5073 Fax: 913.850.5100 Email: [email protected] Website: www.xenometricsllc.com Xenometrics, LLC, of Stilwell, Kansas, is a GLP compliant, USDA registered, AAALAC-accredited contract research organization, providing services to the pharmaceutical, biotech, companion animal health, and industrial- and agro-chemical industries. Xenometrics’ research species includes rodents, rabbits, minipigs, dogs, cats, nonhuman primates, and other research species. Xenometrics’ study services include: General Toxicology, and DART, Safety Pharmacology and DMPK. 134 Can Scientists Bridge the Gap Between Preclinical & Clinical Test Results? They’re getting closer with Taconic’s help. is a unique and evolving portfolio of translational mouse models and in vitro tools & services that improve the predictability of preclinical ADME and Toxicity studies. Find out more at www.taconic.com/tadmet SPONSORED SYMPOSIUM Join Us on Monday, November 10th, for the “Use of Humanized Mouse Models In DMPK and Safety Testing of Compounds” symposium. 135 th 35 American College of Toxicology Annual Meeting Exhibitor Exhibit Hall Directory Map EXHIBIT INFO Grand Cypress Ballroom 136 2014 th 35 American College of Toxicology Annual Meeting 2014 Exhibitor-Hosted Exhibitor Directory Programs Exhibitor-Hosted Programs are commercially supported educational sessions held in conjunction with the ACT Annual Meeting. Programs are open to all meeting attendees. toxicology research as the need for higher throughput toxicity screens has grown. This presentation will focus on the ways in which zebrafish research is changing the field of toxicology. Submitting Standardized SEND to the FDA: Data Fitment and Review Considerations Sunday, November 9 12:00 Noon–12:55 PM Regency Hall #4 Presented by: PointCross Life Sciences, Inc. An overview of the FDA’s e-data submission and JumpStart initiatives for nonclinical studies will be provided. Data fitment requirements, analytics and viewers for reviewing e-data and lessons learned from industry and FDA implementations will be presented. Benefits of standardizing NDA submissions for early engagement with the FDA will be discussed. Maintaining Quality and Regulatory Compliance in a Global Setting Wednesday, November 12 7:00 AM–7:55 AM Grand Cypress Ballroom H Presented by: WuXi AppTec Practicalities of Dermal Administration Wednesday, November 12 7:00 AM–7:55 AM Grand Cypress Ballroom G Presented by: Huntingdon Life Sciences Dermal administration for preclinical evaluations presents logistical challenges. The goal is to mimic as closely as possible the clinical (human) situation using an animal model which may have skin similar to a human's but may be anatomically and behaviorally different from a human. This session will discuss techniques and materials that have been developed to assure that test articles applies to animals (primarily rats and minipigs) remain in place and provide appropriate exposure in preclinical studies. Zebrafish: Reshaping Toxicity Testing Wednesday, November 12 7:00 AM–7:55 AM Grand Cypress Ballroom I Presented by: Charles River Over the past two decades, the zebrafish has transformed from an alternative model to a firmly established workhorse in Wednesday, November 12 12:00 Noon–12:55 PM Grand Cypress Ballroom H Presented by: Leadscope Inc. ICH M7 guidelines require the use of an expert alert-based and a statistical-based QSAR methodology. This seminar will demonstrate the new Leadscope Model Applier – Genetic Toxicity Expert Alerts system. Additionally, we will review our “consensus prediction” using integrated results from our new Genetox Expert Alerts system and statistical-based Genetox QSAR models. Evaluation of Electronic Cigarettes in Testing Tobacco-Related Products Wednesday, November 12 12:00 Noon–12:55 PM Grand Cypress Ballroom G Presented by: Battelle E-cigarettes are regarded as less hazardous than traditional combustible tobacco products. However, the health effects from e-cigarettes are not clearly understood. We will present results from the characterization of exposure atmospheres from e-cigarettes and discuss our perspective on how tobacco-related products may be evaluated to comply with the regulations established by the CTP. Seizure Liability, qEEG and Sleep Quantification in Nonclinical Drug Development: Regulatory and Scientific Considerations Wednesday, November 12 12:00 Noon–12:55 PM Grand Cypress Ballroom I Presented by: CiToxLAB Current topics in Neurotoxicology: • EEG strategies to investigate seizure liabilities at early and late stages of drug development. • Regulatory considerations that impact study design, species selection and EEG experimental endpoints. • qEEG: a sensitive tool in drug development? • Quantification of drug effects on sleep patterns. 137 EXHIBIT INFO With the need now to reach larger patient populations faster and quicker, global submissions are an attractive proposition to drug companies. By doing a single program designed to be globally compliant, companies can save time and money as well as reducing the need for repeating studies and thereby reducing animal use. Demonstration of New Expert Alert System for In Silico Assessment of Impurities under ICH M7 Guidelines th 35 American College of Toxicology Annual Meeting 2014 Council Listing 2013–2014 Council President • Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] Councilors • Nancy R. Bordelon, PhD, DABT (2013–2016) Covance Laboratories 671 South Meridian Road Greenfield, IN 46140 Tel: 317.467.2736 Email: [email protected] President-Elect • Alan P. Brown, PhD, DABT (2013–2014) Novartis Institutes for Biomedical Research 100 Technology Square 607-951P Cambridge, MA 02139 Tel: 617.871.3263 Email: [email protected] • Mary Ellen Cosenza, PhD, DABT, RAC Amgen Inc. One Amgen Center Drive Mail Stop 17-2-A Thousand Oaks, CA 91320-1789 Tel: 805.447.6318 Email: [email protected] • David R. Compton, PhD, DABT (2013–2016) Sanofi US 55 Corporate Drive, PO Box 5925 MC: 55B-425A / B-4-4421 Bridgewater, NJ 08807-5925 Tel: 908.981.3272 Email: [email protected] Vice President • Hanan N. Ghantous, PhD, DABT US FDA CDER/OAP/DAVP 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 410.458.2930 Email: [email protected] Secretary REFERENCE Treasurer • Jerry F. Hardisty, DVM (2011–2014) Experimental Pathology Laboratories, Inc. PO Box 12766 Research Triangle Park, NC 27709 Tel: 919.423.1148 Email: [email protected] • Grace M. Furman, PhD, DABT Paracelsus, Inc. 128 Daphne Street Leucadia, CA 92024 Tel: 760.271.2858 Email: [email protected] • Anthony L. Kiorpes, PhD, DVM, DABT (2012–2015) River Bluff Associates LLC 2470 Skyline Drive Bloomington, MN 55425-2188 Tel: 952.854.9060 Email: [email protected] • Alan M. Hoberman, PhD, DABT, ATS Charles River Laboratories Global Development 905 Sheehy Drive Horsham, PA 19044 Tel: 215.443.8710 Email: [email protected] • Timothy J. McGovern, PhD (2011–2014) US FDA CDER/Office of New Drugs 10903 New Hampshire Avenue WO22 Silver Spring, MD 20993 Tel: 240.402.0477 Email: [email protected] Past President • Robin C. Guy, MS, DABT, RQAP-GLP Robin Guy Consulting, LLC PO Box 830 Preclinical Toxicology & GLP Training Lake Forest, IL 60045-0830 Tel: 847.295.9250 Email: [email protected] 138 th 35 American College of Toxicology Annual Meeting 2014 Council Listing 2013–2014 Council (continued) Councilors (continued) Editor-In-Chief • Sandra L. Morseth, PhD (2012–2015) Morseth Consulting, LLC 4804 Old Middletown Road Jefferson, MD 21755 Tel: 240.422.7699 Email: [email protected] • Mary Beth Genter, PhD, DABT University of Cincinnati 160 Panzeca Way Kettering Lab, Room #129 Cincinnati, OH 45267 Tel: 513.558.6266 Email: [email protected] • Melissa C. Rhodes, PhD, DABT (2013–2016) GlaxoSmithKline 5 Moore Drive Research Triangle Park, NC 27709 Tel: 919.483.6908 Email: [email protected] Executive Director • Nancy Rollman 1821 Michael Faraday Drive, Suite 300 Reston, VA 20190 Tel: 703.547.0875 Email: [email protected] • Patricia C. Ryan, PhD (2012–2015) MedImmune, LLC One MedImmune Way Gaithersburg, MD 20878 Tel: 301.398.4387 Email: [email protected] 2014–2015 Incoming Council President Secretary President-Elect Past President • Mary Ellen Cosenza, PhD, DABT, RAC Amgen Inc. One Amgen Center Drive Mail Stop 17-2-A Thousand Oaks, CA 91320-1789 Tel: 805.447.6318 Email: [email protected] • Hanan N. Ghantous, PhD, DABT US FDA CDER/OAP/DAVP 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 410.458.2930 Email: [email protected] • Timothy J. McGovern, PhD US FDA CDER/Office of New Drugs 10903 New Hampshire Avenue WO22 Silver Spring, MD 20993 Tel: 240.402.0477 Email: [email protected] • Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] Vice President 139 • Holly D. Dursema, MS, DABT Boehringer Ingelheim R9-5900 Ridgebury Rd., Box 368 Ridgefield, CT 06877-0368 Tel: 203.798.5694 Email: [email protected] • Kenneth J. Olivier Jr, PhD, BS Merrimack Pharmaceuticals One Kendall Square Suite B7201 Cambridge, MA 02139 Tel: 774.219.3130 Email: [email protected] • Michael S. Orr, PhD US FDA 10903 New Hampshire Avenue Silver Spring , MD 20993-002 Tel: 301.796.1604 Email: [email protected] REFERENCE • Tracey Zoetis, MS SciLucent, LLC 585 Grove Street Suite 300 Herndon, VA 20170 Tel: 703.342.8747 Email: [email protected] Councilors (2014–2017) th 35 American College of Toxicology Annual Meeting 2014 Committee Listings 2013–2014 Committees Awards Committee • Chair, Hanan N. Ghantous, PhD, DABT US FDA 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 410.458.2930 Email: [email protected] • William J. Brock, PhD, DABT, ATS Brock Scientific Consulting, LLC 19909 Hamil Circle Montgomery Village, MD 20886 Tel: 301.519.3666 Email: [email protected] • Stephen B. Harris, PhD, FATS, FSB Stephen B. Harris Group 6109 Madra Avenue San Diego, CA 92120 Tel: 619.469.7886 Email: [email protected] • A. Wallace Hayes, PhD, DABFE, DABT, FATS, FIBiol, ERT Harvard University School of Public Health 298 South Main Street Andover, MA 01810 Tel: 978.749.3085 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Lisa B. Biegel, PhD Covance Laboratories, Inc. 3301 Kinsman Blvd.–15 Madison, WI 53704 Tel: 608.245.7093 Email: [email protected] • Holly D. Dursema, MS, DABT Boehringer Ingelheim 900 Ridgebury Rd., Box 368 Ridgefield, CT 06877-0368 Tel: 203.798.5694 Email: [email protected] • Kate E. Lane, PhD, DABT Cubist Pharmaceuticals Inc. 65 Hayden Ave Lexington, MA 02421 Tel: 781.860.1026 Email: [email protected] • Melissa C. Rhodes, PhD, DABT GlaxoSmithKline 5 Moore Drive Research Triangle Park, NC 27709 Tel: 919.483.6908 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] Education Committee • Chair, Jerry F. Hardisty, DVM EPL, Inc. PO Box 12766 Research Triangle Park, NC 27709 Tel: 919.423.1148 Email: [email protected] • Co-Chair, Patricia Ryan, PhD MedImmune, LLC One MedImmune Way Gaithersburg, MD 20878 Tel: 301.398.4387 Email: [email protected] • Ilona Bebenek, PhD US FDA 10903 New Hampshire Ave. Bldg 22 Room 6237 Silver Spring, MD 20993 Tel: 240.402.3843 Email: [email protected] REFERENCE Education Committee: Webinar Subcommittee • Chair, Ilona G. Bebenek, PhD US FDA 10903 New Hampshire Ave. Bldg 22, Room 6237 Silver Spring, MD 20993 Tel: 240.402.3843 Email: [email protected] • Lisa D. Beilke, MSPH, PhD, DABT Toxicology Solutions, Inc. 7015 Schilling Ave San Diego, CA 92126 Tel: 650.504.2547 Email: [email protected] • Nancy R. Bordelon, PhD, DABT Covance Laboratories 671 South Meridian Road Greenfield, IN 46140 Tel: 317.467.2736 Email: [email protected] • Angélique Braen, PhD, DABT Ikaria, Inc. 53 Frontage Road, POB 9001 Hampton, NJ 08827 Tel: 908.238.6471 Email: [email protected] 140 • Hanan N. Ghantous, PhD, DABT US FDA 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 410.458.2930 Email: [email protected] th 35 American College of Toxicology Annual Meeting 2014 Committee Listings 2013–2014 Committees (continued) Education Committee: Webinar Subcommittee (continued) • Anthony Ndifor, PhD Janssen R&D 3210 Merryfield Row San Diego, CA 92121 Tel: 858.784.3260 Email: [email protected] • Kenneth Olivier Jr., PhD, BS Merrimack Pharmaceuticals One Kendall Square Suite B7201 Cambridge, MA 02139 Tel: 774.219.3130 Email: [email protected] • Adam Woolley, DABT, MS, FRCPath, ERT, ATS ForthTox Ltd. PO Box 13550 Linlithgow, West Lothian, EH49 7YU Tel: 44 1506 844036 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Hanan N. Ghantous, PhD, DABT US FDA 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 410.458.2930 Email: [email protected] • Michael P. Holsapple, PhD, ATS Covance Laboratories Inc. 3301 Kinsman Blvd Madison, WI 53704-2523 Tel: 608.218.0299 Email: [email protected] • Theresa Sweeney, PhD, DABT Nektar Therapeutics 455 Mission Bay Blvd, South San Francisco, CA 94158 Tel: 415.482.5681 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Anthony L. Kiorpes, PhD, DVM, DABT River Bluff Associates LLC 2470 Skyline Drive Bloomington, MN 55425-2188 Tel: 952.854.9060 Email: [email protected] • Dennis J. Naas, BS, PMP ProDev Consulting Services, Ltd. 14244 Silver Ridge Road Poway, CA 92064 Tel: 858.883.2992 Email: [email protected] • Deborah L. Novicki, PhD, DABT Novartis Vaccines 350 Massachusetts Avenue 45 SS 5103C Cambridge, MA 02139 Tel: 510.703.2713 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] Finance Committee • Chair, Alan M. Hoberman, PhD, DABT, ATS Charles River 905 Sheehy Drive Horsham, PA 19044 Tel: 215.443.8710 Email: [email protected] • Lisa D. Beilke, MSPH, PhD, DABT Toxicology Solutions, Inc. 7015 Schilling Ave San Diego, CA 92126 Tel: 650.504.2547 Email: [email protected] Membership Committee 141 REFERENCE • Chair, Timothy J. McGovern, PhD US FDA 10903 New Hampshire Ave WO22 Silver Spring, MD 20993 Tel: 240.402.0477 Email: [email protected] • Nancy R. Bordelon, PhD, DABT Covance Laboratories 671 South Meridian Road Greenfield, IN 46140 Tel: 317.467.2736 Email: [email protected] • Anne H. Chappelle, PhD, DABT Chappelle Toxicology Services, LLC 3850 Rotherfield Lane Chadds Ford, PA 19317 Tel: 484.844.7662 Email: [email protected] th 35 American College of Toxicology Annual Meeting 2014 Committee Listings 2013–2014 Committees (continued) Nominating Committee • Chair, Robin C. Guy, MS, DABT, RQAP-GLP Robin Guy Consulting, LLC PO Box 830 Lake Forest, IL 60045-0830 Tel: 847.295.9250 Email: [email protected] • Carol S. Auletta, MBA, DABT, RAC Huntingdon Life Sciences PO Box 2360 Mettlers Road East Millstone, NJ 08875-2360 Tel: 732.873.2550 (2960) Email: [email protected] • Tracey L. Spriggs, PhD, DABT GlaxoSmithKline Consumer Healthcare 1500 Littleton Road Parsippany, NJ 07054-3884 Tel: 973.889.2503 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Kenneth Olivier Jr., PhD, BS Merrimack Pharmaceuticals One Kendall Square Suite B7201 Cambridge, MA 02139 Tel: 774.219.3130 Email: [email protected] • Tracy Williams, PhD, DABT Eli Lilly and Company Lilly Corporate Center Indianapolis, IN 46285 Tel: 317.277.4197 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Florence Burleson, PhD Burleson Research Technologies, Inc. (BRT) 120 First Flight Lane Morrisville, NC 27560 Tel: 919.719.2500 Email: [email protected] • Hanan N. Ghantous, PhD, DABT US FDA 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 410.458.2930 Email: [email protected] • Jerry F. Hardisty, DVM EPL, Inc. PO Box 12766 Research Triangle Park, NC 27709 Tel: 919.423.1148 Email: [email protected] • Wafa A. Harrouk, PhD-DABT/D US FDA 10903 New Hampshire Avenue Silver Spring, MD 20993 Tel: 301.796.0908 Email: [email protected] • David Hobson, PhD, DABT LoneStar PharmTox LLC 613 Pleasant Valley Drive N. Boerne, TX 78006 Tel: 210.269.6169 Email: [email protected] • Norman Kim, MS, DABT Biogen Idec Inc. 14 Cambridge Center Cambridge, MA 02142 Tel: 617.679.4995 Email: [email protected] • Patrick D. Lilly, DABT ALCS 601 East Jackson St Richmond, VA 23219 Tel: 804.335.2670 Email: [email protected] • Michael Moore, PhD AZ–Pharma Consulting LLC 16419 S 16th Ave Phoenix, AZ 85045-1721 Tel: 301.943.3325 Email: [email protected] Outreach Committee • Chair, Robin C. Guy, MS, DABT, RQAP-GLP Robin Guy Consulting, LLC PO Box 830 Lake Forest, IL 60045-0830 Tel: 847.295.9250 Email: [email protected] • Florence Burleson, PhD Burleson Research Technologies, Inc. (BRT) 120 First Flight Lane Morrisville, NC 27560 Tel: 919.719.2500 Email: [email protected] REFERENCE Program Committee • Chair, Mary Ellen Cosenza, PhD, DABT, RAC Amgen Inc. 1 Amgen Center Drive Mail Stop 17-2-A Thousand Oaks, CA 91320-1789 Tel: 805.447.6318 Email: [email protected] • Nancy R. Bordelon, PhD, DABT Covance Laboratories 671 South Meridian Road Greenfield, IN 46140 Tel: 317.467.2736 Email: [email protected] • Angélique Braen, PhD, DABT Ikaria, Inc. 53 Frontage Road, POB 9001 Hampton, NJ 08827 Tel: 908.238.6471 Email: [email protected] 142 th 35 American College of Toxicology Annual Meeting 2014 Committee Listings 2013–2014 Committees (continued) Program Committee (continued) • Sandra Morseth, PhD Morseth Consulting, LLC 4804 Old Middletown Rd Jefferson, MD 21755 Tel: 301.473.4730 Email: [email protected] • Deborah L. Novicki, PhD, DABT Novartis Vaccines 350 Massachusetts Avenue 45 SS 5103C Cambridge, MA 02139 Tel: 510.703.2713 Email: [email protected] • Thulasi Ramani, PhD Huntingdon Life Sciences 100 Mettlers Road Somerset, NJ 08873 Tel: 732.873.2550 (4329) Email: [email protected]. com • David Serota, PhD, DABT MPI Research 54943 N. Main Street Mattawan, MI 49071 Tel: 269.668.3336 (1338) Email: [email protected] • Suzanne Wolford, PhD, DABT Covance, Inc. 3301 Kinsman Blvd Madison, WI 53704 Tel: 608.242.2721 Email: [email protected] • Tracey Zoetis, MS SciLucent, LLC 585 Grove Street Suite 300 Herndon, VA 20170 Tel: 703.342.8747 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Shayne C. Gad, PhD, DABT Gad Consulting Services 102 Woodtrail Lane Cary, NC 27518 Tel: 919.233.2926 Email: [email protected] • Kenneth L. Hastings, DrPH, DABT, ATS Hastings Toxicology Consulting, LLC 3860 Turf Court South Mount Airy, MD 21771 Tel: 301.829.5663 Email: [email protected] • Alan Hoberman, PhD, DABT, ATS Charles River 905 Sheehy Drive Horsham, PA 19044 Tel: 215.443.8710 Email: [email protected] • David Hobson, PhD, DABT LoneStar PharmTox LLC 613 Pleasant Valley Drive N. Boerne, TX 78006 Tel: 210.269.6169 Email: [email protected] • Tracey Zoetis, MS SciLucent, LLC 585 Grove Street Suite 300 Herndon, VA 20170 Tel: 703.342.8747 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Patricia Frank, PhD Patricia Frank & Associates, Inc. 417 Dewey Avenue Evanston, IL 60202 Tel: 847.864.6535 Email: [email protected] • Robin C. Guy, MS, DABT, RQAP-GLP Robin Guy Consulting, LLC PO Box 830 Lake Forest, IL 60045-0830 Tel: 847.295.9250 Email: [email protected] • Kenneth Hastings, DrPH, DABT, ATS 3860 Turf Court South Mount Airy, MD 21771 Tel: 301.829.5663 Email: [email protected] Publications Committee • Editor-In-Chief, Mary Beth Genter, PhD, DABT, ATS University of Cincinnati 160 Panzeca Way Kettering Lab, Room #129 Cincinnati, OH 45267 Tel: 513.558.6266 Email: [email protected] • Mary Ellen Cosenza, PhD, DABT, RAC Amgen Inc. 1 Amgen Center Drive Mail Stop 17-2-A Thousand Oaks, CA 91320-1789 Tel: 805.447.6318 Email: [email protected] • Grace Furman, PhD, DABT Paracelsus, Inc. 128 Daphne Street Leucadia, CA 92024 Tel: 760.271.2858 Email: [email protected] Resource Committee 143 REFERENCE • Chair, David Serota, PhD, DABT MPI Research 54943 N. Main Street Mattawan, MI 49071 Tel: 269.668.3336 (1338) Email: [email protected] th 35 American College of Toxicology Annual Meeting 2014 Committee Listings 2013–2014 Committees (continued) Resource Committee (continued) • David Hobson, PhD, DABT LoneStar PharmTox LLC 613 Pleasant Valley Drive N. Boerne, TX 78006 Tel: 210.269.6169 Email: [email protected] • Robert Osterberg, PhD, ATS Osterberg Pharm-Tox Consulting LLC 4617 Morgan Drive Chevy Chase, MD 20815 Tel: 301.951.0338 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] • Ex Officio, Alan Hoberman, PhD, DABT, ATS Charles River 905 Sheehy Drive Horsham, PA 19044 Tel: 215.443.8710 Email: [email protected] Website Committee • Chair, David Compton, PhD, DABT Sanofi US 55 Corporate Drive, PO Box 5925 MC: 55B-425A, B-4-4421 Bridgewater, NJ 08807-5925 Tel: 908.981.3272 Email: [email protected] • Grace Furman, PhD, DABT Paracelsus, Inc. 128 Daphne Street Leucadia, CA 92024 Tel: 760.271.2858 Email: [email protected] • William Mattes, PhD, DABT US Food and Drug Administration National Center for Toxicological Research Room 50-333 Jefferson, AR 72079 Tel: 870.543.7681 Email: [email protected] • Tracey Spriggs, PhD, DABT GlaxoSmithKline Consumer Healthcare 1500 Littleton Road Parsippany, NJ 07054-3884 Tel: 973.889.2503 Email: [email protected] • Ric Stanulis, PhD, DABT Acorda Therapeutics Inc 420 Saw Mill River Road Ardsley, NY 10502 Tel: 914.326.5376 Email: [email protected] • Alan Stokes, PhD, DABT GlaxoSmithKline RD9-2015 Five Moore Drive Res Triangle Pk, NC 27709-3398 Tel: 919.315.2598 Email: [email protected] • Ex Officio, Drew A. Badger, PhD, DABT Amgen Inc. One Amgen Center Drive Thousand Oaks, CA 91320 Tel: 805.447.5875 Email: [email protected] 2014–2015 Incoming Committee Members REFERENCE Awards Committee • Elaine V. Knight, PhD National Cancer Institute, NIH Toxicology and Pharma Branch 9609 Medical Center Drive RM 4-W108, MSC 9734 Bethesda, MD 20892 Tel: 240.276.5939 Email: [email protected] Education Committee • Pam Marone, PhD 1804 Keelingwood Lane Virginia Beach, VA 23454 Tel: 757.450.7191 Email: [email protected] • Laine Peyton Myers, PhD US FDA CDER / Div of Antiviral Products 10903 New Hampshire Ave Silver Spring, MD 20993 Tel: 301.796.2217 Email: [email protected] 144 Finance Committee • Jeff Tepper, PhD, DABT Tepper Nonclinical Consulting 197 Glasgow Lane San Carlos, CA 94070 Tel: 510.717.1413 Email: [email protected] Membership Committee • Elliot B. Gordon, PhD, DABT Elliot Gordon Consulting, LLC 55 Lillie Street Princeton Junction, NJ 0855 Tel: 609.936.1977 Email: [email protected] th 35 American College of Toxicology Annual Meeting Nominating Committee • David Serota, PhD, DABT MPI Research 54943 N. Main Street Mattawan, MI 49071 Tel: 269.668.3336 (1338) Email: [email protected] • Jerry F. Hardisty, DVM EPL, Inc. PO Box 12766 Research Triangle Park, NC 27709 Tel: 919.423.1148 Email: [email protected] 2014 Outreach Committee • Harry M. Olson, PhD, DVM STA Preclinical Services LLC 720 Dartmouth Avenue Silver Spring, MD 20910 Tel: 860.304.5114 Email: [email protected] ACT Headquarters 1821 Michael Faraday Drive, Suite 300, Reston, Virginia 20190 Tel: 703.547.0875 • Fax: 703.438.3113 Email: [email protected] • Website: www.actox.org Executive Director Meetings Manager Nancy Rollman Email: [email protected] Ext. 1404 Elsa Cannon Email: [email protected] Ext. 1890 Program Manager Program Coordinator Elisa Turner Email: [email protected] Ext. 1650 Jordan Ballance Email: [email protected] Ext. 1425 REFERENCE 145 th 35 American College of Toxicology Annual Meeting 2014 ACT Presidents 1979–1980 M. Selikoff* 1997–1998 Christopher P. Chengelis, PhD 1980–1981 Myron Mehlman, PhD 1998–1999 David W. Hobson, PhD, DABT 1981–1982 Yula Bingham, PhD 1999–2000 Merrill R. Osheroff, PhD, DABT 1982–1983 Arthur Furst, PhD, ScD* 2000–2001 Suzanne C. Fitzpatrick, PhD, DABT 1983–1984 Gary Flamm, PhD 2001–2002 Robert E. Osterberg, PhD 1984–1985 Ronald W. Hart, PhD 2002–2003 John E. Atkinson, PhD, DABT 1985–1986 Marshall Steinberg, PhD* 2003–2004 Robert Snyder, PhD, ATS 1986–1987 Gordon W. Newell, PhD 2004–2005 Patricia Frank, PhD 1987–1988 Robert M. Diener, DVM 2005–2006 Leigh Ann Burns Naas, PhD, DABT 1988–1989 Richard M. Hoar, PhD 2006–2007 Stephen B. Harris, PhD, FATS, FSB 1989–1990 Carol M. Henry, PhD, DABT 2007–2008 A. Wallace Hayes, PhD, DABT, FATS 1990–1991 Shayne C. Gad, PhD, DABT 2008–2009 Kenneth L. Hastings, DrPH, DABT, ATS 1991–1992 Mildred S. Christian, PhD, ATS* 2009–2010 Carol S. Auletta, MBA, DABT 1992–1993 Karen M. MacKenzie, PhD, DABT 2010–2011 Russette M. Lyons, PhD 1993–1994 Richard D. Thomas, PhD, DABT 2011–2012 David G. Serota, PhD, DABT 1994–1995 Sharon J. Northup, PhD 2012–2013 Robin C. Guy, DABT, MS, RQAP–GLP 1995–1996 Sidney Green, PhD 2013–2014 Drew A. Badger, PhD, DABT 1996–1997 John A. Thomas, PhD REFERENCE *Deceased 146 th 35 American College of Toxicology Annual Meeting 2014 ACT Award Recipients Distinguished Scientist Award ACT Service Award 2013 Albert E. Munson 2013 John A. Thomas 2012 A. Wallace Hayes 2012 Mary Ellen Cosenza 2011 Glenn Sipes 2011 David Hobson 2010 Elizabeth Weisburger 2010 Jerry F. Hardisty 2009 John Casida 2009 Patricia Frank 2008 Ron Estabrook 2008 Shayne Gad 2007 Gerald Wogan 2007 Harihara Mehendale 2006 Joseph Rodricks 2006 Robert Diener 2005 Bernard Goldstein 2004 Mildred Christian 2004 Peter Guengerich 2001 Arthur Furst 2003 Jack Dean ACT Student Travel Awards 2002 Carol Henry 2001 Curtis Klaassen 2000 Bernard Schwetz 1999 John Thomas 1998 Robert Scala 1997 Joe Borzelleca 1996 John Doull 1995 Kenneth Olden (formerly Distinguished Service Award) 2014 Anthony D. Dayan (formerly Lifetime Contribution Award) 2014 Norman N. Kim 2013 Shailender Singh Chauhan Chand Basha Davuljigari Heidi Hsieh Chitrada Kaweeteerawat ACT Young Professional Award 2014 Ilona G. Bebenek 2013 Lisa D. Beilke 2012 Kenneth J. Olivier Jr. 2011 Melissa C. Rhodes 2010 Nancy Bordelon ACT Unsung Hero Award 2014 Kok-Wah Hew 2013 Eve Kagan 2012 Carol C. Lemire REFERENCE 147 th 35 American College of Toxicology Annual Meeting 2014 Corporate Members Alcon Research, Ltd. Gilead Sciences, Inc. Fort Worth, Texas Foster City, California Allergan, Inc. GlaxoSmithKline Irvine, California Research Triangle Park, North Carolina Amgen Inc. Huntingdon Life Sciences, PRC Astellas Pharma US, Inc. Janssen R&D (Johnson & Johnson) Thousand Oaks, California East Millstone, New Jersey Northbrook, Illinois Spring House, Pennsylvania BASi MPI Research West Lafayette, Indiana Mattawan, Michigan Biogen Idec Inc. Pfizer Inc. Cambridge, Massachusetts Andover, Massachusetts BioReliance Corporation Philip Morris International R&D Boehringer Ingelheim Purdue Pharma, LP Rockville, Maryland Neuchâtel, Switzerland Ridgefield, Connecticut Cranbury, New Jersey Bristol-Myers Squibb Company R. J. Reynolds Tobacco Co Princeton, New Jersey Winston-Salem, North Carolina Charles River Sanofi US Wilmington, Massachusetts Bridgewater, New Jersey Covance Laboratories, Inc. Takeda Global R&D Eli Lilly and Company Vertex Pharmaceuticals Inc. Experimental Pathology Laboratories, Inc. WIL Research Madison, Wisconsin Deerfield, Illinois Indianapolis, Indiana Cambridge, Massachusetts Sterling, Virginia Ashland, Ohio REFERENCE Genentech, Inc. South San Francisco, California 148 th 35 American College of Toxicology Annual Meeting 2014 Welcome New Members Melanie Abongwa Anna Adamou Chidozie Amuzie Mark Bauter Conney Berger Terri Blake Christopher Brynczka Steven Bulera Julie Castaneda Hao Chen Paul Ciaccio David Clarke Roger Clemens Frances Crofts Alison Easter Ikram Elayan Peter Emau Marc Fariss James Fishback Marie Fortin Clay Frederick Brenda Gannon Ronald Gerson Shubham Goyal Arielle Hinds Heidi Hsieh Mohd Jaafar William Jo Laura Kaufman Chitrada Kaweeteerawat Noreen Khan-Mayberry Yumee Kim Andrea Kim Sheri Klas Mark Kleven Seva Kostrubsky Dan Krietlow Raluca Kubaszky Theresa Lansdell Jinyoung Lee Betina Lew Patrick Lilly Zhiwei Liu Norbert Makori Andrea Mason Dani Matsushima Ray Matulka Tanya McDonnell Carrie McMahon Tiffany Messerli Mark Miller David Miller Lance Molnar Paramita Mookherjee Nadia Moore Brian Mulhern Stefan Nechev Louise Neilson Tin Ngo Wei Niu Michael Orr Gopinath Palanisamy Chris Papagiannis David Peters Jessica Phillips Jason Pinkstaff Kara Polhamus Bindu Prabhakar Priti Prasad Larry Rodman Shakil Saghir Natalie Scholpa David Scoville Flemming Simonsen Phillip Smiraldo Graeme Smith Chelsea Snyder Robert Spaet Donald Stump Timothy Teague Michael Templin Donald Thompson Alessandro Venosa W. Mark Vogel Lorraine Webster John Wise David Woolley M. Keith Wyatt Li Yang ACT Charter Members (ACT Founded in 1977) Ed Kerfoot Sandra Reiss Bruce Bernard Mearl Kilmore Harry Salem Morris Cranmor Perry Kurtz Ceinwen Schreiner Mike Farrow Walter Melvin Van Seabaugh William George Robert Osterberg D.P. Sinha Robert Hinderer Matthew Palazzolo Robert Szot Edward Jackson Richard Parent William Watt John Johnnidis John Pollock ACT Distinguished Fellows A. Wallace Hayes (2013) Harry Salem (1998) Myron Mehlman (1989) Joe Borzelleca (2012) Gary Flamm (1989) Joseph Arcos (1985) John Thomas (2003) Art Furst (1989) 149 REFERENCE Mohamed Abou-Donia American College of Toxicology www.actox.org 36thth ANNUAL ANNUAL MEETING MEETING November 8–11, 2015 Red Rock Resort • ummerlin, Nevada s Call for Proposals Do you or a colleague have an idea for a scientific Symposium Session, a speaker, or a Continuing Education course? If so, consider submitting a 2015 Session Proposal for the 36th Annual Meeting to be held in Summerlin, Nevada. Your ideas are invaluable to the College and the success of the Annual Meeting. The deadline for submission is Friday, February 20, 2015. Visit ACT’s website to obtain 2015 Session Proposal information. www.actox.org 150 Reserve Your Exhibit Booth Today for 2015! To reserve your 2015 booth visit the ACT Registration Desk or contact [email protected] Tel: 703.547.0875 American College of Toxicology 36th Annual Meeting November 8–11, 2015 Red Rock Resort, Summerlin, Nevada www.actox.org 151 American College of Toxicology 36 th th ANNUAL ANNUAL MEETING MEETING November 8–11, 2015 Red Rock Resort summerlin, Nevada www.actox.org 152 The American College of Toxicology would like to thank these meeting sponsors. Silver $2,500–$4,999 BASi Patricia Frank & Associates, Inc. Vet Path Services, Inc. MPI Research PreClinical Research Services, Inc. WIL Research Bronze $1,000–$2,499 Calvert Laboratories CiToxLAB Eisai, Inc. Gilead Sciences, Inc. Lovelace Respiratory Research Institute Quidel Corporation Seventh Wave Laboratories Society of Toxicology Southern Research Tox Path Specialists (TPS), LLC Shire Consultant Sponsors Bill Brock Brock Scientific Consulting Chris Chengelis Chengelis Scientific Consulting Services Charles Lindamood CLIII Consulting, LLC Stephen Montgomery Sandra Morseth Morseth Consulting, LLC Michael J. Taylor NonClinical Safety Assessment Grace Furman Stephen Harris Stephen B. Harris Group Robert Susick Paracelsus, Inc. Susick Consulting, LLC John Budny Marian Glynn Consultant in Nonclinical Safety Assessment PharmaCal, Ltd. Adam Woolley ProDev Consulting Services, Ltd. ForthTox Limited Robin Guy Robin Guy Consulting Dennis Naas Anthony Kiorpes River Bluff Associates Tox Clarity Alan Katz ToXcel, LLC The American College of Toxicology would like to thank these meeting sponsors. Platinum $10,000+ Gold $5,000–$9,999 Altria