You are on page 1of 116

10th

A N N IV E R SA RY

Program & Abstracts Book


10th ASM Biodefense and Emerging Diseases Research Meeting
The Omni Shoreham Hotel | Washington, DC
February 26 - 29, 2012

www.asmbiodefense.org
w w w.i ca ac .o rg
www.icaac.org

ICAAC, the premier conference on antimicrobial agents and infectious diseases, showcases the
latest-breaking science and lectures from top researchers from around the globe. Join 10,000 of the
world’s foremost physicians, clinical microbiologists, researchers, pharmacists, and other health care
professionals in San Francisco, California, for timely information that will improve health care and
the management of infectious diseases.

Abstracts
Abstract Submission Closes: May 7, 2012, 11:59 EST
Travel Grant Application Submission Closes: June 29, 2012

Meeting Highlights
Opening Keynote Session | Sunday, September 9, 2012
Emerging New Issues in the Management of Hepatitis C Infection
Charles M. Rice, PhD; The Rockefeller University, New York, NY
The Impact of Emerging New Diagnostic Laboratory Techniques on Clinical Infectious Disease
Franklin R. Cockerill, III, MD; Mayo Clinic, Rochester, MN
Prospects for New Antibiotics
Christopher T. Walsh; Harvard Medical School, Boston, MA

ICAAC Lecture | Monday, September 10, 2012


Fungal Disease in the 21st Century
Arturo Casadevall, MD, PhD; Albert Einstein College of Medicine of Yeshiva University, Bronx, NY
W E LCO M E LE T T E R

Dear Colleagues,
Welcome to the 10th American Society for Microbiology’s Biodefense and Emerging Diseases Research
Meeting in Washington, DC. The ASM Biodefense Research Program Committee has assembled a pro-
gram that includes experts in multiple relevant fields presenting in several different session formats.
The 2012 program is intended to provide each attendee with the opportunity to participate in educa-
tional programming that has the most value and relevance to their individual professional needs. For
this year’s meeting we have brought together individuals who are carrying out research to defend
against the growing threats of bioterrorism and emerging infectious diseases and decision makers
Paul Keim, Ph.D. shaping the future of the biodefense research agenda.
The meeting opens Sunday at noon with four in-depth Focus Sessions followed by the Keynote Session
Sunday afternoon. This year’s Keynote Speaker, Paul Offit, M.D., from the Children’s Hospital of
Philadelphia, will present “Hard Knocks: Communicating Vaccine Science to the Public.” Dr. Offit is
one of the country’s foremost advocates for vaccines. We cordially invite all meeting attendees to
the Opening Reception in the Exhibit Hall immediately following the conclusion of Dr. Offit’s talk.
Beginning Monday morning, daily Plenary Sessions include “Outbreaks, Response, and Hindsight,”
“Targeting Virulence Factors in Acute Infection,” and “Immunity to Pulmonary Pathogens.” Diverse
concurrent symposia are also scheduled for Monday and Tuesday afternoon. In addition, for the first
time we have added new early morning sessions that will focus on current international security
challenges and policy issues. These will include a panel discussing the recent experiments modifying
Marshall Bloom, M.D.
the transmissibility of H5N1 avian influenza in mammals and the resulting implications for biosecurity
policy. High profile speakers have been recruited for these early morning sessions, including Dr. Bruce
Alberts, Dr. Michael Osterholm, Dr. Anthony Fauci, Dr. Ron A.M. Fouchier, Assistant Secretary Thomas
Countryman, and Dr. Alan Rudolph. Please check the details in this Program and Abstracts book to
identify the presentations that most appeal to you.
From over 180 accepted abstract submissions, the Program Committee has organized 2 Poster
Sessions and 6 Highlighted Oral Abstract Sessions. Poster presentations will be presented in the
Exhibit Hall, Monday and Tuesday afternoons. Poster presenters will be available from 1:00 – 3:00 PM
to answer your questions. The Exhibit Hall also features 36 exhibitors with the latest information on
new products. The Highlighted Oral Abstract Sessions occur Monday and Tuesday from 3:00 – 4:00 PM.
Please check the details in this Program and Abstracts book.
Again, the Program Committee welcomes you and looks forward to meeting you as we continue to
work together in addressing the challenges that lie ahead.

Sincerely,

Paul Keim, Ph.D. Marshall Bloom, M.D.


ASM Biodefense Research Meeting Co-Chair ASM Biodefense Research Meeting Co-Chair

BIODEFENSE PROGRAM & ABSTRACTS 1


Table of Contents

1 Welcome Letter
2 Program Committee and ASM Officers
10th ASM Biodefense and Emerging 3 Program-at-a-Glance
Diseases Research Committee 4 Daily Schedule-at-a-Glance
5 Program Highlights
Paul Keim, Ph.D., Co-Chair 6 Student Postdoctoral Fellow Travel
Northern Arizona University, Flagstaff, AZ Grant Recipients
Marshall Bloom, M.D., Co-Chair 7 Biodefense Exhibits
NIH/NIAID, Hamilton, MT
8 Poster Presentation Hall Map—Monday
Catharine Bosio, Ph.D. 9 Poster Presentation Hall Map—Tuesday
NIH/NIAID, Hamilton, MT
10 Omni Shoreham Hotel Map
Elisabeth Carniel, M.D., Ph.D. 11 General Information
Institut Pasteur, Paris, France
13 Scientific Program
Miles Carroll, Ph.D.
33 Abstracts
Health Protection Agency Center for Emergency Preparedness & Response,
Wiltshire, United Kingdom 97 Keyword Index
Colleen Jonsson, Ph.D. 101 Author Index
Center for Predictive Medicine for Biodefense and Emerging Infectious
Disease Clinical & Transitional, Louisville, KY
Karl Klose, Ph.D.
University of Texas, San Antonio, TX
Carol Linden, Ph.D.
Office of the Biomedical Advanced Research & Development Authority,
Washington, DC
C. Rick Lyons, M.D.
Colorado State University, Fort Collins, CO ASM Officers
Petra Oyston, Ph.D. David C. Hooper, Ph.D., President
DSTL, Wiltshire, United Kingdom Massachusetts General Hospital, Boston, MA
Jean Patterson, Ph.D. Jeffery F. Miller, M.D., President-Elect
Texas Biomedical Research Institute, San Antonio, TX University of California, Los Angeles, CA
Theodore Pierson, Ph.D. Joseph Campos, Ph.D., Secretary
NIH/NIAID, Bethesda, MD Children's National Medical Center, Washington, DC 
Anne Rimoin, Ph.D. James Tiedje, Ph.D., Treasurer
UCLA School of Public Health, Los Angeles, CA Michigan State University, East Lansing, MI
Leonard Smith, Ph.D.
US Army Medical Research Institute, Fort Detrick, MD ASM Headquarters
American Society for Microbiology
Meetings and Exhibits Department
1752 N Street, NW
Washington, DC 20036-2904
Telephone: (202) 942-9248
Fax: (202) 942-9340
E-mail: biodefense@asmusa.org
Website: www.asmbiodefense.org

ISBN: 978-1-55581-840-1
©2012 American Society for Microbiology

2 10th ASM Biodefense and Emerging Diseases Research Meeting


P R O G R A M - AT- A - G L A N C E

10th ASM Biodefense and Emerging Diseases Research Meeting

SUNDAY MONDAY TUESDAY WEDNESDAY


February 26 February 27 February 28 February 29

Registration 9:00 AM – 7:00 PM 6:30 AM – 5:00 PM 6:30 AM – 3:30 PM 6:30 AM – 8:30 AM

Focus Sessions 12:00 PM – 2:00 PM


2:15 PM – 4:15 PM

Keynote Session 4:30 PM – 6:00 PM

Global Threats, Collaborative Solutions 7:15 AM – 8:15 AM 7:15 AM – 8:15 AM

H5N1 Research Discussion 7:15 AM – 8:15 AM

Plenary Sessions 8:30 AM – 12:00 PM 8:30 AM – 12:00 PM 8:30 AM – 12:00 PM

Lunch 12:00 PM – 1:00 PM 12:00 PM – 1:00 PM

Exhibits & Poster Hall 6:00 PM – 8:00 PM 12:00 PM – 4:15 PM 12:00 PM – 4:15 PM
(Exhibits Only)

Opening Reception 6:00 PM – 8:00 PM

Poster Sessions 1:00 PM – 3:00 PM 1:00 PM – 3:00 PM

Highlighted Oral Abstract Presentations 3:00 PM – 4:00 PM 3:00 PM – 4:00 PM

Concurrent Symposia 4:15 PM – 6:15 PM 4:15 PM – 6:15 PM

Roundtable Discussion 6:30 PM – 7:30 PM 6:30 PM – 7:30 PM

BIODEFENSE PROGRAM & ABSTRACTS 3


10th ASM Biodefense and Emerging Diseases Research Meeting
Daily Schedule-at-a-Glance
This section provides an at-a-glance look of the entire Biodefense program. Please reference the page numbers listed
under each session title to view the session’s description, moderators, and speakers.

SUNDAY, February 26 Highlighted Oral Abstract Poster Session


Presentations 1:00 PM - 3:00 PM | See Page 23
Focus Sessions 3:00 PM - 4:00 PM | See Page 20 018. Tuesday Poster Session
12:00 PM - 2:00 PM | See Page 13 009. Environmental Detection
001. Animal Models 010. Viral Agents Highlighted Oral Abstract
12:00 PM - 2:00 PM | See Page 13 011. Bacterial Agents Presentations
002. What’s Next? Pathogen 3:00 PM - 4:00 PM | See Page 27
Discovery from the Field to Symposia 019. Immune Response
the Laboratory
4:15 PM - 6:15 PM | See Page 21 020. Therapeutics
2:15 PM - 4:15 PM | See Page 14 012. Biodefense at the Dinner 021. Diagnostics and Vaccines
003. Burkholderia pseudomallei: Table: Emerging Foodborne
Understanding Pathogenesis Illnesses
and Immunity to Guide the Symposia
Next Generation of Medical 4:15 PM - 6:15 PM | See Page 21 4:15 PM - 6:15 PM | See Page 28
Countermeasures 013. Persistence and Inactivation 022. Host Genetic Resistance
of Biothreat Agents and to Infections
2:15 PM - 4:15 PM | See Page 14 Emerging Pathogens
004. Non-Coding RNAs and 4:15 PM - 6:15 PM | See Page 29
the Regulation of Virulence 4:15 PM - 6:15 PM | See Page 22
023. Dynamic Pathogens Make
014. Taking the Bite Out of Difficult Targets
Insect Bites: Biodefense
Keynote Session Implications of Vector-Borne 4:15 PM - 6:15 PM | See Page 29
4:30 PM - 6:00 PM | See Page 15 Infections 024. Visual Discovery of
005. Hard Knocks: Microbe-Host Interactions
Communicating Vaccine Roundtable Discussion
Science to the Public
6:30 PM - 7:30 PM | See Page 22 Roundtable Discussion
015. Recapping the 2011 6:30 PM - 7:30 PM | See Page 30
Biological Weapons Convention 025. Global Status of Strategies
MONDAY, February 27 Review Conference (BWC for Addressing Intersection of
RevCon) Science and Security
Global Threats, Collaborative
Solutions
7:15 AM - 8:15 AM | See Page 15 TUESDAY, February 28 WEDNESDAY, February 29

Plenary Session Global Threats, Collaborative Solutions H5N1 Research Discussion


8:30 AM - 12:00 PM | See Page 15 7:15 AM - 8:15 AM | See Page 23 7:15 AM – 8:15 AM | See Page 31
007. Outbreaks, Response, and
Hindsight Plenary Session Plenary Session
8:30 AM - 12:00 PM | See Page 23 8:30 AM - 12:00 PM | See Page 31
Poster Session 017. Targeting Virulence Factors 027. Immunity to Pulmonary
1:00 PM - 3:00 PM | See Page 16 in Acute Infection Pathogens
008. Monday Poster Session

4 10th ASM Biodefense and Emerging Diseases Research Meeting


Program Highlights

Keynote Session
Sunday, February 26, 4:30 PM – 6:00 PM | Regency Ballroom
Hard Knocks: Communicating Vaccine Science to the Public

PAUL A. OFFIT, M.D., The Children’s Hosp. of Philadelphia, Philadelphia, PA


A major and critical goal in the area of biodefense and emerging diseases is the development
of safe and effective vaccines. However, due to often deliberate misinformation about vaccine
complications and poor news reportage, public acceptance of even currently available vaccines
is waning. If new vaccines are to be effectively deployed, it is critical that the biomedical and
research community be cognizant of the hurdles facing vaccination science. Dr. Offit is an
acknowledged expert in the area of vaccine science and one of the foremost advocates for
vaccines in the United States. He has written books and articles about vaccine science and
the controversies about vaccines, and has appeared numerous times in the public media. As a result, his keynote
lecture will provide timely and important information for anyone interested in the topic of vaccines.

Global Threats, Collaborative Solutions


Monday, February 27, 7:15 AM – 8:15 AM | Palladian Ballroom
Assistant Secretary Thomas Countryman, U.S. Department of State Bureau of International
Security and Nonproliferation, Washinton, DC
Assistant Secretary Thomas Countryman will share his vision for U.S. efforts to prevent the spread
of biological weapons, through cooperative engagement, international treaties and diplomacy.

Tuesday, February 28, 7:15 AM - 8:15 AM | Palladian Ballroom


Dr. Alan Rudolph, Director for Chemical and Biological Technologies Directorate, Research and
Development Enterprise, Defense Threat Reduction Agency (DTRA), Fort Belvoir,VA
Dr. Alan Rudolph will provide a unique look at DoD’s efforts to safeguard America and its allies
from weapons of mass destruction, drawing from his experiences in driving innovative technolo-
gies to reduce, eliminate, and counter biological threats.

H5N1 Research Discussion


Wednesday, February 29, 7:15 AM – 8:15 AM | Palladian Ballroom
Moderated by the Chair of the National Science Advisory Board for Biosecurity (NSABB),
Paul Keim, Ph.D., this newly added session at the ASM Biodefense and Emerging Diseases
Research Meeting will include discussions of the NSABB’s recommendations for the publication
of the controversial H5N1 research. This session will also provide an open forum for attendees to
give their feedback on such policy issues as the appropriate mechanisms to allay public concerns
about the safety of dual use research.
continued on next page...

BIODEFENSE PROGRAM & ABSTRACTS 5


PROGRAM H IG H LIG HTS

Presentations:

NSABB Recommendations
Michael T. Osterholm, Ph.D., MPH
University of Minnesota School of Public
Health, Minneapolis, MN
Director, Center for Infectious Disease
Research and Policy (CIDRAP) Live Video Stream
Government Response to the H5N1 Research Discussion
Recommendations Wednesday, February 29 | 7:15 AM – 8:15 AM
Anthony S. Fauci, M.D.
ASM will be hosting a free live video stream of the
Director, National Institute of Allergy and
H5N1 Research Discussion taking place on February 29,
Infectious Diseases (NIAID)
2012 at that Biodefense Meeting. We encourage you
to share this information with your colleagues that
Science’s Response to the Situation were unable to attend. The link will be available on
Bruce Alberts, Ph.D. this meeting’s website the morning of the session.
Editor-in-Chief of Science If you or your colleagues are unable to participate
in the live event/video stream, you may access the
archived footage of the session that will be posted
to the website at 1:00 PM on Wednesday.
Perspective from an Investigator www.asmbiodefense.org
Ron A.M. Fouchier, Ph.D.
Erasmus MC, Rotterdam, Netherlands

Student Postdoctoral Fellow Andrew Clohessey


Rowan University, Glassboro, NJ
Travel Grant Recipients Session 008; Presentation 102 (J)
Susann Dupke
Robert Koch Institute, Berlin, Germany
Congratulations to our 10th ASM Biodefense and Session 008; Presentation 042 (B)

Emerging Diseases Research Meeting Student Manoshi Gayen


Uniformed Services, University of Health Sciences, Bethesda, MD
Postdoctoral Fellow Travel Grant Recipients! Session 018; Presentation 199 (H)

The ASM Student Travel Grant Program was Yanyan Li


University of Maryland, Baltimore, MD
established to support the travel of students to ASM Session 011; Presentation 116

meetings. Recipients are selected by the Biodefense Kevin McCormick


University of Pittsburgh, Pittsburgh, PA
Research Program Committee on the basis of the Session 008; Presentation 031 (A)
scientific quality of the submitted abstracts. Emilio Ortiz Riano
University of Rochester Medical Center, Rochester, NY
Session 018; Presentation 144 (A)

6 10th ASM Biodefense and Emerging Diseases Research Meeting


E X H I B I T S I N F O R M AT I O N

Biodefense Exhibits Information


Exhibit Hall Information Opening Reception in the Exhibit Hall
Scientific exhibits are located in the Exhibit Hall, Level 1B, Sunday, February 26 | 6:00 PM – 8:00 PM
co-located with poster sessions.
Network with colleagues, investigate the amazing variety of
The Exhibit Hall will be open: exhibits, and visit with industry representatives at your leisure,
Sunday, February 26 ...................6:00 PM – 8:00 PM all while enjoying light hors d’oeuvres and drinks.
Monday, February 27..................12:00 PM – 4:15 PM Name badges required.
Tuesday, February 28 .................12:00 PM – 4:15 PM
Additionally, lunch will be provided in the Exhibit Hall from
12:00 PM – 1:00 PM on Monday and Tuesday. Attendees can lis- Exhibits, posters, lunch and the Opening Reception
ten to poster presentations, then get the most current informa- will be held in the Exhibit Hall.
tion on scientific products and services relating to Biodefense.

The American Society for Microbiology welcomes the following exhibitors:

A2LA American Association for emka Technologies inc Reagent Proteins


Laboratory Accreditation Booth 43 | www.emkatech.com Booth 36 | www.reagentproteins.com
Booth 16 | www.A2LA.org
Germfree Labs, Inc. Southern Research
Advanced Instruments, Inc. Booth 45 | www.germfree.com Booth 25 | www.southernresearch.org
Booth 24 | www.aicompanies.com
Idaho Technology, Inc. Tetracore, Inc.
ASM Press Booth 20 | www.bio-surveillance.com Booth 39 | www.tetracore.com
Booth 21 | estore.asm.org
IIT Research Institute The Howard T. Ricketts Regional
BEI Resources Booth 29 | www.iitri.org Biocontainment Laboratory
Booth 40 | www.atcc.org Booth 26 | www.ntrl.uchicago.edu
InnovaPrep
Bertin Technologies/Bertin Corp. Booth 35 | innovaprep.com UTMB-National Biocontainment
Booth 1 | www.bertin.fr Training Center
(JPM-TMT) Joint Project Manager
Booth 22 | www.utmb.edu/nbtc
Biaera Technologies, LLC Transformational Medical Technologies
Booth 42 | www.biaera.com Booth 44 | www.jpmtmt.mil United States Army Healthcare Recruiting
Booth 17 | www.goarmy.com
Bio-Synthesis, Inc. List Biological Laboratories, Inc
Booth 37 | www.biosyn.com Booth 31 | www.listlabs.com
Center for Predictive Medicine, Lovelace Respiratory Research Institute
University of Louisville Booth 32 | www.LRRI.org
Booth 19 | www.louisville.edu/predictivemedicine
Meso Scale Defense
Certek Booth 46 | www.mesoscale.com
Booth 28 | www.certekinc.com
Nanologix
CH Technologies (USA) Inc. Booth 27 | www.nanologix.com
Booth 41 | www.inhalation.org
National BioSafety and Biocontainment
Data Sciences International Training Program
Booth 30 | www.datasci.com Booth 2 | nbbtp.org
Defense Threat Reduction Agency National Counterproliferation Center (NCP)
Booth 18 | www.dtra.mil Booth 33 | www.phe.gov/s3
Division of Select Agents & Toxins, CDC NIH Office of Biotechnology Activities
Booth 14 | www.selectagents.gov Booth 15 | www.oba.od.nih.gov
DynPort Vaccine Company LLC OpGen, Inc
Booth 38 | www.csc.com/dvc Booth 47 | www.OpGen.com

BIODEFENSE PROGRAM & ABSTRACTS 7


E X H I B I T S I N F O R M AT I O N

Exhibits and Posters


Monday, February 27, 2012

Monday Poster Key


Category Presentation Number
A 027 (A) - 033 (A)
B 034 (B) - 061 (B)
D 062 (D) - 069 (D)
E 070 (E) - 075 (E)
F 076 (F) - 079 (F)
G 080 (G) - 083 (G)
H 084 (H) - 093 (H)
I 094 (I) - 098 (I)
J 099 (J) - 102 (J)
K 103 (K) - 107 (K)

8 10th ASM Biodefense and Emerging Diseases Research Meeting


E X H I B I T S I N F O R M AT I O N

Exhibits and Posters


Tuesday, February 28, 2012

Exhibits and Posters


Tuesday, February 28, 2012

Tuesday Poster Key


Category Presentation Number
A 140 (A) - 146 (A)
B 147 (B) - 174 (B)
D 175 (D) - 180 (D)
E 181 (E) - 186 (E)
F 187 (F) - 191 (F)
G 192 (G) - 196 (G)
H 197 (H) - 209 (H)
I 210 (I) - 214 (I)
J 215 (J) - 216 (J)
K 217 (K) - 220 (K)

BIODEFENSE PROGRAM & ABSTRACTS 9


LOWER LEVEL
2B
Health
Veranda Empire Club
Room

10th ASM Biodefense and Emerging Diseases Research Meeting


Robert’s
Private
Robert’s Restaurant Dining Palladian Diplomat
WEST LOBBY Room Room Room
Cabinet Room
Congressional A&B Room
Council Room Sales Conference
Empire Foyer
Executive Room Room
Forum Room
Sales Conference Room
Senate Room
West Registration Bird Cage Walk
Women’s Lounge ADA Lift to
(Lower Level)
BALLROOMS (West Lobby) Men’s
Ambassador and
Clothing Executive Regency Ball Rooms
Ambassador Ballroom ADA Elevator Store Room Telephones
Diplomat Room to Roberts Restaurant Committee Level 1B
Director’s Room
Empire Room and Palladian Room WEST LOBBY West Room
Elevators
Palladian Room Coat Check Stairs
Regency Room AM
BA
MAIN LOBBY Jewelry Men’s Business West BAL SSAD
Store
Restroom Center Registration LRO OR
Women’s Men’s
OM
Telephones Restroom Restroom
(Lower Level)
Front Desk Reg

Congressional
Council

Marquee Lounge
& Reception
Room Senate
Room
ency G

Room
alle
ry
Forum
Room
O M N I S H O R E H A M H OT E L

West
Conference
Cabinet Center
Room REG
ENC
Y BAL
LRO
OM
MAIN ENTRANCE
ADA Ramp For Access to Diplomat ballrooms
To Lobby Please use elevators on the West Side
and go to level 1B.
For Access to the Empire Ballroom
OMNI SHOREHAM HOTEL
and Health Club/Outdoor Pool
Please use elevators on the West Side
of the Hotel and go to level 2B.

10
G E N E R A L I N F O R M AT I O N

General Information

Badge Policies Certificate of Attendance


For security reasons, all attendees are required to wear a badge Certificates of Attendance are available at the Registration Desk
to attend sessions or enter the Exhibit Hall. Please note that during registration hours beginning Tuesday, February 28th.
badges are the property of ASM and must be relinquished at Certificates of Attendance do not list session information.
any time at the request of ASM staff or security.
Child Safety
Attendee Badges
Children are not permitted into session rooms. Also, children
■ Access into all scientific sessions under the age of 16 are not permitted on the Exhibit Hall
■ Access to the Exhibit Hall and Poster Sessions during floor.
operating hours
Contact Information
Guest Badges
To contact ASM staff on site at the conference, please call
■ Access to the Exhibit Hall during operating hours the Omni Shoreham Hotel, at (202) 234-0700, and ask
■ Access to the Opening Reception and lunch on Monday to be transferred to ASM Headquarters located in the
and Tuesday Director’s Room.

Attendees wearing guest badges will not be permitted into scientific Food and Beverage
session rooms.
There are many dining options within walking distance of the
Exhibitor Badges Omni Shoreham Hotel. A list of restaurants can be obtained
from the hotel concierge.
■ Entrance into the Exhibit Hall during installation and
dismantling hours Please Note: Lunch will be provided Monday, February 27th,
and Tuesday, February 28th from 12:00 PM – 1:00 PM in the
■ Entrance into the Hall one hour before the Hall opens
Exhibit Hall, Level 1B
and one hour after it closes
Attendees wearing exhibitor badges will not be permitted into Lost and Found
scientific session rooms. Unattended personal belongings will be removed and taken
to hotel security.
Business Center
A full-service business center is located in the Lobby Level Meeting Announcements
of the Omni Shoreham Hotel, and is available 24 hours. A bulletin board will be available in the West Lobby
registration area for those attendees wishing to post one-page
Cameras and Recording flyers announcing upcoming meetings.
Digital recorders, cameras (including camera phones), and video
cameras (including video phones) are prohibited in the poster Messages
hall and session rooms. Anyone found photographing, videotap- All mail and communications to meeting registrants should
ing or recording in the prohibited areas will be asked to surren- be directed to the individual at the hotel where he or she is
der their badge immediately and leave the conference. No staying. The phone number for the Omni Shoreham Hotel
refund will be provided. This rule is strictly enforced. is (202) 234-0700.

BIODEFENSE PROGRAM & ABSTRACTS 11


G E N E R A L I N F O R M AT I O N

ASM Press Symposia


ASM Press, the book publishing division of the Symposia consist of 4-5 faculty, invited by the Biodefense
American Society for Microbiology, will be exhibiting Research Program Committee, who present lectures on a
its broad selection of texts and references in molecular specific topic followed by a moderated Q&A period.
biology and the microbiological sciences. Be sure to
shop during the meeting to receive special “meeting Poster Sessions
only” discount. All accepted abstracts are scheduled for poster presentation
based on the December 2011 review of all abstract submis-
sions by the Biodefense Research Program Committee.
Posters presentations will take place in the Exhibit Hall
Registration
Monday, February 27th, and Tuesday, February 28th. Each
Registration will be located at the West Registration Desk from presenter will be available at his or her poster to answer
Sunday, February 26 – Wednesday, February 29. questions from 1:00 PM – 3:00 PM either Monday or
Registration Hours: Tuesday as scheduled in this Program and Abstracts book.

Sunday, February 26 . . . . . . . . . . . 9:00 AM - 7:00 PM Focus Sessions


Monday, February 27. . . . . . . . . . . 6:30 AM – 5:00 PM Focus sessions are in-depth educational activities that highlight
Tuesday, February 28 . . . . . . . . . . . 6:30 AM – 3:30 PM a specific topic area. These sessions precede the Keynote session
Wednesday, February 29. . . . . . . . . 6:30 AM – 8:30 AM and are an opportunity for attendees to explore new and
emerging issues with a group of faculty experts.
Smoking Policy
Smoking is prohibited in all areas of the Omni Shoreham Hotel.
Plenary Sessions
Daily Plenary Sessions will be held Monday, Tuesday, and
Speaker Ready Room Wednesday mornings from 8:30 AM to 12:00 PM. These
sessions provide all attendees an opportunity to collaborate on
The Speaker Ready Room will be located in the Director’s
presentations of science, policy, and public health issues. The
Room on the Lobby Level.
speakers were invited by the Biodefense Program Committee
Speaker Ready Room Hours: based on their professional expertise and achievement. Plenary
Sunday, February 26 . . . . . . . . . . . 9:00 AM - 4:30 PM Sessions may have a moderated Q&A period.

Monday, February 27. . . . . . . . . . . 6:30 AM - 6:30 PM


Highlighted Oral Abstract Sessions
Tuesday, February 28 . . . . . . . . . . . 6:30 AM - 6:30 PM
These sessions highlight research submitted for the ASM
Wednesday, February 29. . . . . . . . . 6:30 AM -12:00 PM Biodefense and Emerging Diseases Research Meeting. Authors
Oral presenters are requested to bring their PowerPoint presen- are invited by topic to give a 10 to 15 minute presentation of
tations on a "thumb drive" or CD-ROM to the Speaker Ready their research followed by a moderated Q&A period.
Room at least one hour prior to their session to be uploaded. A For specific presentation times and additional details, please
technician will be available to help ensure your presentation is see the session information listed by date in this
properly uploaded. Program and Abstracts book.

Special Needs Roundtable Discussion Sessions


Please contact an ASM staff member if you require special One hour sessions dedicated to providing attendees with infor-
accommodations while you attend the meeting. mation about selected government programs to include how to
apply for grants and other financial support.

12 10th ASM Biodefense and Emerging Diseases Research Meeting


SCIENTIFIC PROGRAM

Sunday, February 26, 2012 Focus Session


002. What’s Next? Pathogen Discovery from the
Focus Session Field to the Laboratory
001. Animal Models Sunday, February 26, 2012 | 12:00PM – 2:00PM
Sunday, February 26, 2012 | 12:00PM – 2:00PM Palladian Ballroom
Diplomat Ballroom Pathogen discovery is at the front line of predicting and identifying
Animal studies are the cornerstone of obtaining regulatory approval emerging infections. Here we will cover pathogen discovery from sam-
for products to protect against biodefense agents and emerging pling and surveillance in the field, through development and applica-
threats. To date, guidance from the FDA regarding these studies has tion of tools to interrogate samples, and descriptions of the discovery
been minimal and sometimes contradictory. To bring a single product of novel pathogens. The viewers will learn cutting edge technology of
to point where it can move forward for regulatory approval requires viral discovery and where the field work is headed.
the cooperation of a large community, to include government agen-
MODERATORS:
cies, contract research organizations and the biodefense pharma-
ceutical industry. Animal studies have to be designed and executed in Jean Patterson; Texas Biomedical Res. Inst., San Antonio, TX.
a manner that minimizes animal use and distress, while allowing for Anthony Griffiths; Texas Biomedical Res. Inst., San Antonio, TX.
studies to be scientifically valid and comparable across research sites. PRESENTATIONS:
In order for data to be comparable between laboratories, clear well- 12:00PM
defined ethical study endpoints have to be established for each in-
fectious agent in each appropriate animal model. This requires an 007 Early Detection of Novel Biothreats Through International
Scientific Engagement and Medical Diplomacy
early understanding, During initial model development, of what re-
quirements will be needed to evaluate efficacy of medical counter- Joseph Fair; Global Viral Forecasting Initiative, San Francisco, CA.
measures. 12:30PM
MODERATORS: 008 Viral Discovery Using Pan-Pathogen Microarrays and Deep
Sequencing
Louise M. Pitt; USAMRIID, Frederick, MD.
Charles Chiu; Univ. of California Sch. of Med., San Francisco, CA.
Catharine Bosio; NIAID/Rocky Mountain Lab., Hamilton, MT.
1:00PM
PRESENTATIONS:
009 Discovery and Characterization of Novel Viruses
12:00PM
David Wang; Washington Univ. Sch. of Med., St. Louis, MO.
001 Introduction and Setting the Stage
Louise M. Pitt; USAMRIID, Frederick, MD. 1:30PM
010 Using Targeted Wildlife Surveillance to Predict Emerging
12:20PM Zoonoses
002 Process and Implications of Animal Model Qualification Jonathan H. Epstein; EcoHlth. Alliance, New York, NY.
Rosemary Roberts; CDER, FDA, Silver Spring, MD.

12:40PM
003 Issues in Animal Model Harmonization
Judith Hewitt; DMID/NIAID, Bethesda, MD.

1:00PM
004 Evaluating Study Endpoints and Euthanasia Considerations
for Animal Protocols
Estella Jones; FDA, Silver Spring, MD.

1:20PM
005 Exploring Approaches to Consistent Veterinary Care and
Euthanasia Criteria
Jim Swearengen; NBACC, Fort Detrick, MD.

1:40PM
006 Animal Rule Requirements vs. Expectations: How Can the
Developers of Medical Countermeasures Succeed?
Elizabeth Leffel; PharmAthene, Inc., Annapolis, MD.

BIODEFENSE PROGRAM & ABSTRACTS 13


S U N DAY S C I E N T I F I C P RO G R A M

Focus Session Focus Session


003. Burkholderia pseudomallei: Understanding 004. Non-Coding RNAs and the Regulation of
Pathogenesis and Immunity to Guide the Next Virulence
Generation of Medical Countermeasures Sunday, February 26, 2012 | 2:15 PM - 4:15 PM
Sunday, February 26, 2012 | 2:15 PM - 4:15 PM Diplomat Ballroom
Palladian Ballroom Non-coding RNAs are gaining increased attention as important mole-
This session will emphasize the tie from basic research into the biol- cules involved in bacterial gene regulation, particularly of those genes
ogy of a microbial pathogen to the development of medical counter- involved in virulence. Topics to be discussed in this session include
measures against it. B. pseudomallei is particularly problematic in small RNA identification in bacterial pathogens, the regulatory mech-
that it is genetically diverse, there is a limited understanding of its anisms exerted by noncoding RNAs and their chaperones, the evolu-
pathogenesis, the current therapeutic strategies are quite extensive, tion of pathogens as mediated through changes in small RNA content
and there are no viable vaccine candidates. Therefore, this session or expression, and the contributions to virulence of non-coding RNAs
will focus on the recent findings in Burkholderia genomics and how in biodefense in emerging infectious diseases.
this microbe interacts with its host, as well as how this information
MODERATOR:
could be used to develop novel countermeasures.
Wyndham Lathem; Northwestern Univ., Chicago, IL.
MODERATORS: PRESENTATIONS:
Daniel Wolfe; Defense Threat Reduction Agency, Fort Belvoir, VA. 2:15PM
Paul Keim; Northern Arizona Univ., Flagstaff, AZ.
015 Small RNAs and the Evolution of Yersinia Species
PRESENTATIONS: Wyndham W. Lathem; Northwestern Univ., Chicago, IL.
2:15PM 2:40PM
011 Melioidosis: Clinical and Therapeutic Issues 016 sRNA Regulation of EHEC Virulence
Bart J. Currie; Royal Darwin Hosp., Casuarina, Australia.
Vanessa Sperandio; Univ. of Texas Southwestern Med. Ctr., Dallas, TX.
2:45PM 3:05PM
012 Population Genetics and Evolution of Burkholderia 017 Regulation of Francisella tularensis Virulence by Small Non-
pseudomallei Coding RNAs
David Wagner; Northern Arizona Univ., Flagstaff, AZ.
John Gunn; Ohio State Univ., Columbus, OH.
3:15PM 3:30PM
013 Elucidation of Type 6 Secretion System as a Critical Virulence 018 Legionella pneumophila Small Regulatory RNAs and
Determinant of Burkholderia pseudomallei Intracellular Multiplication
David Deshazer; USAMRIID, Fort Detrick, MD.
Sebastien P. Faucher; McGill Univ., Ste-Anne-de-Bellevue, Canada.
3:45PM 3:55PM
014 Burkholderia pseudomallei Antibiotic Resistance 019 A Unique Intergenic RNA Thermosensor Controls Yersinia
Mechanisms: Impact on Therapeutic Strategies Virulence
Herbert P. Schweizer; Colorado State Univ., Fort Collins, CO.
Petra Dersch; Helmholtz Ctr. for Infection Res., Braunschweig, Germany.

14 10th ASM Biodefense and Emerging Diseases Research Meeting


S U N DAY / M O N DAY S C I E N T I F I C P RO G R A M

Monday, February 27, 2012


Keynote Session
005. Keynote Session
Sunday, February 26, 2012 | 4:30 PM – 6:00PM 006. Global Threats, Collaborative
Regency Ballroom Solutions
A major and critical goal in the area of biodefense and emerging dis- Monday, February 27, 2012 | 7:15 AM – 8:15AM
eases is the development of safe and effective vaccines. However, due Palladian Ballroom
to often deliberate misinformation about vaccine complications and
This new program will provide insights into current interna-
poor news reportage, public acceptance of even currently available
tional security challenges from notable thought leaders, pol-
vaccines is waning. If new vaccines are to be effectively deployed, it
icy advisors, and senior government officials.
is critical that the biomedical and research community be cognizant of
the hurdles facing vaccination science. Dr. Offit is an acknowledged MODERATOR:
expert in the area of vaccine science and one of the foremost advo- Dr. Jason Rao; ASM Director of International Affairs, Washington, DC.
cates for vaccines in the United States. He has written books and ar-
ticles about vaccine science and the controversies about vaccines, PRESENTATION:
and has appeared numerous times in the public media. As a result, Assistant Secretary Thomas Countryman; U.S. Department of State
his keynote lecture will provide timely and important information for Bureau of International Security and Nonproliferation, Washington, DC.
anyone interested in the topic of vaccines.

MODERATORS:
Marshall E. Bloom; DIR/NIAID/NIH, Hamilton, MT.
Paul S. Keim; Northern Arizona Univ., Flagstaff, AZ.
PRESENTATION:
4:30PM
020 Hard Knocks: Communicating Vaccine Science to the Public
Paul A. Offit; The Children’s Hosp. of Philadelphia, Philadelphia, PA.
Plenary Session
007. Outbreaks, Response, and Hindsight
Monday, February 27, 2012 | 8:30 AM – 12:00PM
Regency Ballroom
The session is designed to provide an insight into outbreaks of
infectious disease, how they occur, how agencies responded
and what we learned about them to prevent recurrence. By cov-
ering both viral and bacterial pathogens associated with recent
outbreaks of disease this will highlight how effective current
regulations and health frameworks are in different countries.
The session will also demonstrate the impact of new technol-
ogy in characterizing pathogens. Finally, since these are recent
outbreaks, this session will provide useful scientific informa-
tion from those with first-hand experience for the first time to
the audience and whether we are improving in our ability to
deal with such incidents.

MODERATORS:
Miles Carroll; Hlth. Protection Agency, Salisbury, United Kingdom.
David L. Heymann; Hlth. Protection Agency, London, United Kingdom.
8:30AM
021 Outbreaks: A Global Perspective
David L. Heymann; Hlth. Protection Agency, London, United Kingdom.

BIODEFENSE PROGRAM & ABSTRACTS 15


MO N DAY S C I E N T I F I C P RO G R A M

9:00AM 032 (A) Predicting the Timing of Peak Pandemic Influenza


022 Swine Flu Emergence in Mexico Activity: ED Volume, School Absenteeism, and Community
Reported ILI
Edgar E. Sevilla-Reyes; Natl. Inst. of Respiratory Diseases, Mexico City,
Mexico. T. Bell1, M. Richardson2; 1West Texas Influenza Ctr., TTUHSC, Amarillo, TX,
2
Amarillo Publ. Hlth. Dept., Amarillo, TX.
9:30AM
033 (A) The Role of Rare Variants in Coronavirus Cross-Species
023 Swine Flu in the United Kingdom Transmission Events
Gail Thomson; Hlth. Protection Agency, Salisbury, United Kingdom. M. Borucki, J. Allen, H. Harris-Chen; Lawrence Livermore Natl. Lab, Liver-
10:00AM more, CA.

Coffee Break 034 (B) Transient Bacterial Lipopolysaccharide-Induced


10:30AM Resistance to Inhalational Bacillus anthracis Infection in the New
Zealand White Rabbit Model
024 Overview of E. coli Outbreaks in the USA
S. B. Yee, D. N. Dyer, N. A. Twenhafel, M. L. M. Pitt; USAMRIID, Fort Detrick,
Robert Tauxe; CDC, Atlanta, GA. MD.
11:00AM
035 (B) Small Quinolone Resistance Plasmids: A Model for
025 E. coli O104:H4: The Outbreak Experience from Germany Evolution Mediated by Site-Specific Recombination (SSR) at oriT
Reinhard F. Burger; Robert Koch Inst., Berlin, Germany. and Xer Sites
T. Tran1,2, P. Andres3, A. Petroni3, A. Soler-Bistué4, E. Albornoz3,
11:30AM
A. Zorreguieta4, R. Reyes-Lamothe2, D. Sherratt2, A. Corso3, M. Tolmasky1,2;
026 Molecular Epidemiology and Characterization of the E. coli 1
California State Univ., Fullerton, CA, 2Univ. of Oxford, Oxford, United King-
O104:H4 Outbreak Strain dom, 3INEI-ANLIS, Buenos Aires, Argentina, 4Fundación Inst. Leloir, Buenos
Alexander Mellmann; Univ. Hosp. Muenster, Muenster, Germany. Aires, Argentina.

036 (B) Antibacterial Activity of Alpha-MSH Alone and in


Poster Session Combination with Other Antibiotics against Methicillin-Resistant
Staphylococcuus aureus
008. Monday Poster Session M. Singh, K. Mukhopadhyay; Jawaharlal Nehru Univ., New Delhi, India.
Monday, February 27, 2012 | 1:00PM – 3:00PM
037 (B) Enhanced Aminoglycoside Sensitivity of Ribonuclease
Exhibit Hall Mutants of Escherichia coli
PRESENTATIONS: A. D. Frazier, W. S. Champney; East Tennessee State Univ., Johnson City, TN.
027 (A) Viral Acute Respiratory Tract Infections in Non-Winter 038 (B) A Model of Inhalation Glanders in African Green Monkeys
Time J. Yeager, P. Facemire, D. Nyakiti, L. Pitt; USAMRIID, Frederick, MD.
M. EL-BARRAWY; Alexandria Univ., Alexandria, Egypt.
039 (B) Genes Required for the Trafficking of S-Layer Proteins in
028 (A) Laboratory Surveillance for Non-Influenza Respiratory the Envelope of Bacillus anthracis
Viruses in Maryland S-M. Nguyen-Mau1,2, D. Missiakas1,2, O. Schneewind1,2; 1Univ. of Chicago,
W. A. Murtaugh1,2, J. Tan1, H. Peters2, B. Healey2, M. Tran2, M. Ziese2, B. An- Chicago, IL, 2Howard Taylor Ricketts Lab., Argonne, IL.
derson2, M. P. Carlos1,2; 1Johns Hopkins Univ., Baltimore, MD, 2Lab. Admin.
Maryland Dept. of Hlth. and Mental Hygiene, Baltimore, MD. 040 (B) Development of a Selective Enrichment Media for
Brucella Species Isolation
029 (A) Natural History Studies for Ebola Virus Zaire Strain in M. Perry, A. Kidney, M. Peck, C. Kelly-Cirino, C. Egan; NYS Dept. of Hlth., Al-
Hartley Guinea Pigs bany, NY.
A. Nalca, H. Bloomfield, S. Frederick, R. Baker, R. Williams, J. Yeager, E.
Zumbrun; USAMRIID, Frederick, MD. 041 (B) Periplasmic YopR Promotes Type III Needle
Polymerization
030 (A) Experimental Respiratory Marburg Angola Virus Infection B. Blaylock, O. Schneewind; Univ. of Chicago, Chicago, IL.
in Hartley Guinea Pigs
A. Nalca, H. Bloomfield, S. Frederick, R. Baker, R. Williams, J. Yeager, E. 042 (B) Virulence Analysis of B. cereus biovar anthracis
Zumbrun; USAMRIID, Frederick, MD. S. Dupke1, T. Franz1, A. Lander1, R. Grunow1, P. L. Goossens2, C. Brezillon2,
S. R. Klee1; 1Robert Koch Inst., Berlin, Germany, 2Inst. Pasteur, Paris, France.
031 (A) A Cytopathic Effect-Based High-Throughput Screening
Assay Identified Four Novel Compounds that Inhibit Dengue 043 (B) Burkholderia thailandensis is a Better Surrogate for B.
Infection pseudomallei Growth and Persistence than B. pseudomallei Bp82
K. D. McCormick; Univ. of Pittsburgh, Pittsburgh, PA. M. A. Firmani, S. Lehman, S. Dieringer-Boyer, K. Bower, B. Dorsey, D. Strat-
ton, K. Delp, D. K. R. Karaolis; Natl. Biodefense Analysis and Countermea-
sures Ctr., Frederick, MD.

16 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY S C I E N T I F I C P RO G R A M

044 (B) WITHDRAWN 054 (B) Interspecies Interaction of Francisella tularensis and
045 (B) Efficacy Testing of Orally Administered Antibiotics against Pseudomonas aeruginosa
an Inhalational Bacillus anthracis Infection in BALB/CMice S. N. Dean, M. L. van Hoek; George Mason Univ., Manassas, VA.
G. J. Hatch, S. R. Bate, J. Fretwell, J. Vipond, S. G. P. Funnell, A. D. G. 055 (B) A Method for Detection of Bacillus anthracis Spores in
Roberts; Hlth. Protection Agency, Salisbury, United Kingdom. Complex Matrices
046 (B) Human Anthrax Outbreaks in Georgia in 2011 O. Stephansson1,2, P. Ågren3,2, S. Byfors3,2, S. Ehrs1,2, M. Klint1,2, M. Lavan-
1 1 1 1 1 der3,2, C. Nilsson3,2, A. Lundin Zumpe3,2; 1Natl. Vet. Inst., Uppsala, Sweden,
L. Malania , N. Tsertsvadze , N. Abazashvili , M. Ramishvili , M. Broladze , 2
Swedish Joint Lab. for Food Safety and Biopreparedness, Uppsala, Swe-
S. Tsanava1, N. Avaliani1, R. J. OBISO2, P. Imnadze1; 1The Natl. Ctr. for Dis-
den, 3Natl. Food Agency, Uppsala, Sweden.
ease Control and Publ. Hlth., Tbilisi, Georgia, 2The Microbe Company, Chris-
tiansburg, VA. 056 (B) Comparative Genomic MGE Finding Process Streamlines
047 (B) Proteomic Analysis of Atypical Y. pestis Strains Isolated Mobile Element Search
from Natural Foci in Georgia S. Y. Choi1,2, N. A. Hasan1,2, J. Chun3, M. Hoq4, A. Huq1, T. A. Cebula5,2, R. R.
1 11 1 2 2 Colwell1,5,2; 1Univ. of Maryland, College Park, MD, 2CosmosID Inc., College
R. Solomonia , M. Nozadze , E. Mikeladze , E. Zhgenti , G. Chanturia , M.
Park, MD, 3Sch. of Biological Sci. and Inst. of Microbiol., Seoul, Korea, Re-
kekeklidze2, R. J. Obiso3, S. Francesconi4, L. Bakanidze2; 1Life Sci. Res. Ctr.,
public of, 4Univ. of Dhaka, Dhaka, Bangladesh, 5Johns Hopkins Univ., Balti-
Ilia State Univ., Tbilisi, Georgia, 2The Natl. Ctr. for Disease Control and Publ.
more, MD.
Hlth., Tbilisi, Georgia, 3The Microbe Company, Christiansburg, VA, 4Naval
Med. Res. Ctr., Frederick, MD. 057 (B) A Literature-Based System for Selecting Burkholderia
048 (B) Clinical and Epidemiologic Assessment of Brucellosis pseudomallei Strain Panels for Animal Model Testing Under the
in Georgia FDA Animal Rule
K. E. Van Zandt1, A. Tuanyok2, H. C. Gelhaus, Jr.1, K. T. Brittingham1, S. A.
T. Onashvili1, E. Mamisashvili11, R. J. Obiso2, M. Nikolaishvili, M. Za-
Sarrazine1, P. S. Keim2, R. L. Warren1; 1Battelle, Columbus, OH, 2Northern
kareishvili, I. Beradze; M. Donduashvili, M. Kokhreidze, N. Vepkhvadze;
1 Arizona Univ., Flagstaff, AZ.
Lab. of Ministry of Agriculture (LMA) Tbilisi, Georgia, 2The Microbe
Company, Christiansburg, VA. 058 (B) Paratransgenic and Transgenic Analysis of Cellular
049 (B) Members of the FPT Subfamily of MFS Transporters are Interactions Between Ixodes Ticks, Mammalian Cells, and
Important for Pathogenesis of Francisella tularensis and Rickettsiae: Lighting the Way with Fluorescent Proteins
Contribute to Modulation of the Host Cytokine Response T. J. Kurtti, J. D. Oliver, N. Y. Burkhardt, R. F. Felsheim, M. J. Herron, U. G.
1 2 1 1 1 1 Munderloh; Univ. of Minnesota, St. Paul, MN.
M. Marohn , A. Santiago , K. Shirey , S. Vogel , E. Barry ; Univ. of Maryland
Sch. of Med., Baltimore, MD, 2Univ. of Virginia Sch. of Med., Charlottesville, 059 (B) Comparison of Challenge Outcomes with Three Different
VA. Bacillus anthracis Isolates in New Zealand White Rabbits
050 (B) Testing Diagnostic and Potential Therapeutic Values of A. Belle1, L. Schaefer1, A. E. Boyer2, M. Gallegos-Candela2, J. R. Barr2,
Bacteriophages Capable of Lysing Yersinia pestis and Yersinia M. de la Garza3, J. Nunneley3, R. Carrion, Jr.3, K. Wycoff1; 1Planet Biotechnol-
pseudotuberculosis ogy Inc., Hayward, CA, 2CDC, Atlanta, GA, 3Texas BioMed. Res. Inst., San
Antonio, TX.
A. A. Filippov, K. V. Sergueev, Y. He, M. P. Nikolich; WRAIR, Silver Spring,
MD. 060 (B) Genotyping of Mycobacterium tuberculosis Strains from
051 (B) Biosynthesis of Lipoteichoic Acid in Bacillus anthracis a Third Level Hospital in Lima, Peru
C. A. Tello, Sr., N. A. Borja, R. Garcia de la Guardia; Sand Marcos Univ.,
G. Garufi1,2, A. Hendrickx1,2, S. G. Richter1,2, K. Beeri1, J. Kern1, O.
Lima, Peru.
Schneewind1,2, D. Missiakas1,2; 1 Univ. of Chicago, Chicago, IL, 2Howard
Taylor Ricketts Lab., Chicago, IL. 061 (B) Evaluation of a Commercially Available Disposable
052 (B) Resistance of Francisella to Fosmidomycin Associated Nebulizer for Use in Nose-Only Pneumonic Plague, Tularensis,
with Mutations in the Glycerol-3-Phosphate Transporter and Melioidosis Murine Models
W. C. Lin, K. Siefkas, S-C. Hu, D. Sullivan, B. Gingras, L. Holland; IIT Res.
R. S. Mackie1, E. S. McKenney2, M. L. van Hoek1,3; 1George Mason Univ.,
Inst., Chicago, IL.
Manassas, VA, 2Univ. of Virginia Hlth. System, Charlottesville, VA, 3George
Mason Univ., Manassas, VA. 062 (D) A PCR Platform Comparison Study to Support
053 (B) Control of Pigment Production in a White Phenotypic Sustainability in Resource-Limited Settings
Variant of Staphylococcus aureus A. Ongarbaev1, A. Khodiev1, A. Smagin2, V. Garib2, G. Abdukhalilova2, G.
1 2 1 1 1 Ibadova2, R. Obiso3, S. Francesconi4, A. Baccetty5, J. Callahan6, D. Williams7;
V. Antonic , A. Stojadinovic , M. Izadjoo , M. R. Alavi ; Henry M. Jackson 1
Tech. Management Company, Tashkent, Uzbekistan, 2Tashkent Inst. of
Fndn. for the Advancement of Military Med., Gaithersburg, MD, 2Combat
Postgraduate Med. Ed., Tashkent, Uzbekistan, 3The Microbe Company,
Wound Initiative Program, Bethesda, MD.
Christiansburg, VA, 4Naval Med. Res. Ctr., Silver Spring, MD, 5Defense
Threat Reduction Agency, Ft. Belvoir, VA, 6Biotechnology and Diagnostic
Solutions Animal Hlth. Life Technologies, Austin, TX, 7Threat Reduction
Support Ctr., Alexandria, VA.

BIODEFENSE PROGRAM & ABSTRACTS 17


MO N DAY S C I E N T I F I C P RO G R A M

063 (D) Development of User-Defined Assays for the Genexpert 075 (E) Multi-User Validation of a Real-Time PCR System for
Platform Detecting Bacillus anthracis Collected by Air Sampling Devices
E. Felder, R. Wölfel; Bundeswehr Inst. of Microbiol., Munich, Germany. in a Liquid Matrix
U. K. Spaulding1, K. M. Clemens1, T. Hadfield2, V. Ryan2, S. Brunelle3, I. M.
064 (D) Production and Qualification of Unrestricted Reagents to Ota1; 1Idaho Technology, Inc., Salt Lake City, UT, 2MRIGlobal, Inc., Palm Bay,
Support Anthrax Assays FL, 3AOAC Intl., Gaithersburg, MD.
K. H. Clement, T. L. Rudge, Jr., N. T. Huber, K. A. Newman, K. A. Sankovich,
R. H. Migliozzi, C. L. Sabourin; Battelle, Columbus, OH. 076 (F) A Hemorrhagic Fever Virus Sequence Database and
Analysis Platform
065 (D) Development of Rapid Diagnostic Platform for Detection C. Kuiken; Los Alamos Natl. Lab., Los Alamos, NM.
of Category A Biothreat Pathogens in the Field
P. Patel1, M. Weidmann2, K. Achazi1, S. Linke1, O. Strohmeier3, D. Mark4, T. 077 (F) Ethical & Social Issues Related to Synthetic Biology: What
van Oordt4, J. Drexler5, M. Eberhard5, F. von Stetten4, M. Niedrig1; 1Robert We Are Doing as Academicians?
Koch Inst., Berlin, Germany, 2Univ. Med. Ctr., Göttingen, Germany, 3Dept. of S. T. HAKIM, S. M. H. Tayyab, S. G. Nadeem; Jinnah Univ. for Women,
Microsystems Engineering (IMTEK), Freiburg, Germany, 4HSG-IMIT, Karachi, Pakistan.
Freiburg, Germany, 5QIAGEN Lake Constance GmbH, Stockach, Germany.
078 (F) Identification and Molecular Characterization of Novel
066 (D) Indirect Phage-Based Detection of Yersinia pestis: Field Group C Orthobunyaviruses Isolated from Mosquitoes Captured
and Clinical Applications in Peru
K. V. Sergueev, M. P. Nikolich, A. A. Filippov; WRAIR, Silver Spring, MD. L. L. Lofts, M. J. Turell, J. R. Kugelman, G. I. Koroleva, G. F. Palacios, C. A.
Whitehouse; USAMRIID, Frederick, MD.
067 (D) Quantification of Anthrax Lethal Factor by Mass
Spectrometry: Application to Clinical Anthrax 079 (F) Development of a High-Throughput Bioinformatics
A. E. Boyer1, A. R. Hoffmaster1, C. P. Quinn1, M. Gallegos-Candela1, R. C. Typing System for the Phylogenomic Analysis of Infectious
Lins2, R. A. Stoddard1, E. Steward-Clark3, P. Maniatis1, C. Marston1, L. X. Category A and B Agents
Cronin1, D. Aranio1, H. Li1, V. Semenova3, K. Isbell2, S. Shadomy1, M. M. Eppinger, S. Agrawal, S. Daugherty, K. Galens, A. Maroo, L. Tallon, H.
Lehman1, D. Blaney1, N. Pesik1, T. L. Smith1, J. R. Barr1; 1CDC, Atlanta, GA, Tettelin, C. Fraser-Liggett, J. Ravel; Univ. of Maryland, Sch. of Med., Balti-
2
Battelle Analytical Services, Atlanta, GA, 3CDC, Chamblee, GA. more, MD.

068 (D) Applying Elastic Light Scatter Technology (BARDOT) to 080 (G) Humanization of Anti-Ricin Neutralizing Monoclonal
Integrate Organism Classification Antibody
H. Motlagh1, A. Giordano1, C. Waters1, E. Bae2, J. G. Thomas1, J. Robinson2; W. Hu, J. Yin, L. Negrych, D. Chau, J. Cherwonogrodzky; Defence R & D,
1
West Virginia Univ., Morgantown, WV, 2Purdue Univ., West Lafayette, IN. Medicine Hat, Canada.

069 (D) Negative Capture for Selective Enrichment of Pathogen- 081 (G) Inhibition of Lipoteichoic Acid Synthesis in
Derived Nucleic Acids in Clinical Samples Staphylococcus aureus and Bacillus anthracis
Z. Bent, S. Langevin, V. VanderNoot, P. Lane, D. Curtis, M. Tran-Gyamfi, S. S. Richter1,2, A. P. A. Hendrickx2, O. Schneewind2,1, D. Missiakas2,1; 1Howard
Branda, T. Lane; Sandia Natl. Labs, Livermore, CA. Taylor Ricketts Lab., Argonne, IL, 2Univ. of Chicago, Chicago, IL.

070 (E) Surveillance on Plague Natural Foci in Georgia 082 (G) Discovery of Targeted and Broad Spectrum Antiviral
L. G. Bakanidze, P. G. Imnadze, S. A. Tsanava, N. S. Tsertsvadze; Natl. Ctr. Countermeasures for Alphaviruses
Disease Control and Publ. Hlth. of Georgia, Tbilisi, Georgia. D. Chung1, B. Beitzel2, J. Arterburn3, J. O. Tret1, J. Smith2, C. Schmaljohn2, G.
Gene Olinger2, C. Scully2, C. B. Jonsson1; 1Univ. of Louisville, Louisville, KY,
071 (E) Performance of Rapid Viability PCR Method for Detection 2
US Army Med. Res. Inst. of Infectious Diseases, Fort Detrick, MD, 3New
of Bacillus anthracis Ames Spores from Sponge-Stick, and Vacuum Mexico State Univ., Las Cruces, NM.
Sock and Filter Sample Types
S. R. Shah1, S. Letant2, G. Murphy2, T. Alfaro2, S. Kane2; 1EPA, Washington, 083 (G) Generation of Biologically Active Nano-Aerosol by an
DC, 2LLNL, Livermore, CA. Electrospray-Neutralization Method
V. N. Morozov1, I. L. Kanev1, N. K. Balabaev1, A. V. Glyakina1, M. L. van
072 (E) An Operational Special Unit to Amend Biological Security Hoek2; 1RAS, Moscow, Russian Federation, 2George Mason Univ.,
M. H. Richter, M-H. Lee, C. Herzog; Robert Koch Inst., Berlin, Germany. Manassas, VA.

073 (E) The Microfluidic Bioagent Autonomous Networked 084 (H) H5N1 Avian Influenza Specific Preventive Maintenance
Detector (M-BAND): An update in the Russia
M. Sanchez, L. Probst, E. Blazevic, B. Nakao, M. A. Northurp; Microfluidic D. Surnev, Yu. Kuznetsov, T. Okovytaya, V. Yelnikov; JSK Pokrov Biological
Systems, Fremont, CA. Plant, Pokrov, Russian Federation.

074 (E) Investigation on Drinking Water Quality of the Bogra


Sadar Upazila: Findings from a Rapid Assessment Lesson in the
Bogra District of Bangladesh
M. Mollik1,2; 1Peoples Integrated Alliance, Dhaka, Bangladesh, 2PreSci.
Trust Funds, Phoenixville, PA.

18 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY S C I E N T I F I C P RO G R A M

085 (H) Immunogenicity and Efficacy of a Recombinant Plague 096 (I) Burkholderia pseudomallei Induces IL-1Beta and IL-6
Vaccine (rF1V) in Swiss Webster Mice Secretion Via Activation of MAPK and STAT3 in Differentiated THP-
P. Fellows1, J. Price1, W. Lin2, A. Rom2, S. Marshall3, L. Holland2, L. Wolfraim1; 1 Cells
1
Dynport Vaccine Co., LLC, Frederick, MD, 2IIT Res. Inst., Chicago, IL, 3Bio- M-H. Cho1, Y-W. Shin1,2, J-H. Chun1, K-J. Hong1, G-E. Rhie1; 1Korea CDC,
Stat Solutions, Inc., Mount Airy, MD. Chungbuk, Korea, Republic of, 2Yonsei Univ., Seoul, Korea, Republic of.

086 (H) Ribosomal Protein TgP0 in the Invasion of Host Cells by 097 (I) Increased Protection of African Green Monkeys Vaccinated
Toxoplasma gondii Tachyzoites with Increasing Doses of a Recombinant F1/V Plague Vaccine
S. Rajagopal, S. Sharma; Tata Inst. of Fundamental Res., Mumbai, India. K. A. Overheim1, S. Lemoine1, K. Gallegos1, K. Brooks1, I. Fisher1, A. Monier1,
C. Schneider1, M. W. Valderas1, J. Wilder1, R. Russell1, R. L. Sherwood1, R.
087 (H) Advancing Clinical Grade rV10-2 Plague Vaccine Toward LeClaire2; 1Lovelace Respiratory Res. Inst., Los Lunas, NM, 2Lovelace Respi-
Licensure ratory Res. Inst., Albuquerque, NM.
L. E. F. Quenee, N. A. Ciletti, D. Elli, T. M. Hermanas, O. Schneewind; Univ. of
Chicago, Argonne, IL. 098 (I) Ebola Virus Secreted Glycoprotein Can Function as a
Decoy for Antibodies
088 (H) Tularemia Vaccine Trials in Nonhuman Primates G. S. Mohan, Y. Chinglai, L. Ye, R. Compans; Emory Univ., Atlanta, GA.
R. Tempel1, R. Shirley2, A. Legasse1, F. Heffron1, M. Axthelm1; 1Oregon Hlth.
& Sci. Univ., Portland, OR, 2Virogenomics, Inc., Portland, OR. 099 (J) Bioactivity of an Indigenous Plant Used in the Treatment
of Malaria against Anopheles Mosquito Larvae in South West
089 (H) A Clinical Phase II Study Confirming the Safety and Nigeria
Immunogenicity of One or Two Doses of IMVAMUNE (MVA-BN) F. O. Omoya, B. E. Boboye; Federal Univ. of Technology, Akure, Nigeria.
Smallpox Vaccine in Vaccinia-Experienced Elderly Subjects
R. N. Greenberg for the POX-MVA-024 Sites1, P. Chaplin2; 1Univ. of Kentucky 100 (J) Formaldehyde Disinfection Using Porcine parvovirus as
Sch. of Med., Lexington, KY, 2Bavarian Nordic GmbH, Martinsried, Germany. Indicator
E. Emmoth, U. A. Bengtsson, I. Dergel, C. Hultén; Natl. Vet. Inst., Uppsala,
090 (H) LC16M8, an Attenuated Smallpox Vaccine Will be a Useful Sweden.
Countermeasure against Bioterrorism with Smallpox
H. Yokote1, Y. Shinmura1, C. Uemura1, T. Kanehara1, S. Maruno1, H. Matsui1, 101 (J) Bactericidal Activity of Antimicrobial Peptides Immobilized
K. Ohkuma1, K. Yokoi1, A. Funatsu1, S. Gordon2, G. Franchini2, S. on Planar Surfaces
Hashizume3; 1Kaetsuken, Kumamoto, Japan, 2Natl. Cancer Inst., Bethesda, S. Arcidiacono, J. Uzarski, L. Doherty, W. Muller, C. Mello; U.S. Army Natick
MD, 3Univ. of Chiba, Chiba, Japan. Soldier Ctr., Natick, MA.

091 (H) Development of a Guinea Pig Passive Transfer Efficacy 102 (J) Investigating the Dispersion and Decontamination of
Model Using Immunoglobulin Derived from Clinical Trial Anthrax Spores within Indoor Work Areas
Volunteers Vaccinated with Recombinant Botulinum Vaccine A/B A. Clohessey, S. Hoque; Rowan Univ., Glassboro, NJ.
S. Martin1, W. Swiderski1, K. Metcalfe1, N. Niemuth2, M. Vassar2, J. Shearer1;
1
DynPort Vaccine Company, Frederick, MD, 2Battelle BioMed. Res. Ctr., 103 (K) Preparing Biodefense Professionals: MS in Biotechnology
Columbus, OH. Concentration in Biodefense and Certificate in National Security
Studies
092 (H) Increased Dose of Recombinant F1/V Plague Vaccine K. M. Obom1, A. I. Roth2, P. J. Cummings3; 1Johns Hopkins Univ., Rockville,
Results in Increased Survival in Plague-Challenged Cynomologus MD, 2Johns Hopkins Univ., Washington, DC, 3Johns Hopkins Univ., Balti-
Macaques more, MD.
K. A. Overheim, S. Lemoine, K. Gallegos, F. Romero, I. Fisher, A. Monier, C.
Schneider, M. W. Valderas, J. Wilder, R. Russell, R. L. Sherwood, R. LeClaire; 104 (K) Recruiting First Responders from the Dental Profession
Lovelace Respiratory Res. Inst., Albuquerque, NM. R. J. Boylan, D. L. Glotzer; NYU Coll. of Dentistry, New York, NY.

093 (H) Generation of Protective Immunity against Fatal 105 (K) Tungiasis (Jigger Infestation) in Rural Kenya: An Emerging
Rickettsial Disease Infectious Disease
S. P. Riley1,2, M. M. Cardwell1,2, R. Hillman1,2, L. Quenee1,2, J. J. Martinez1,2; N. N. N. N. Ngomi, Jr.; Jomokenyatta Univ., Nairobi, Kenya.
1
Univ. of Chicago, Chicago, IL, 2The Howard Taylor Ricketts Lab., Lemont, IL.
106 (K) Essential Oils of Moroccan Thymus munbyanus as
094 (I) Determination of Efficacious Praziquantel Dose in Natural Product for Nosocomial Infection Prevention
Different Mouse Strains: BALB/C and Swiss Mice for Treatment of M. Fatima Zohra, Mohammed Khatouf, Badiaa Lyoussi, Abdelfattah Abdel-
Schistosoma mansoni laoui; Faculty of Sci. Dhar Mhraz, Fes, Morocco.
M. P. M. N. Na1, D. Y. S. N. Yole2; 1Mombasa Polytechnique Univ. Coll.,
Nairobi, Kenya, 2Kenya Polytechnique Univ. Coll., Nairobi, Kenya. 107 (K) Biosafety and Biosecurity: The Case of a Third World
Country and the One Health Spectrum
095 (I) A Novel Mechanism by Which Human Neutrophils Kill D. B. Ndumu1, R. Heckert2, C. S. Rutebarika1, R. W. Kaboyo3, M. Busuulwa4,
Bacillus anthracis P. Atimnedi5, S. Balinandi6, P. Delarosa7; 1Ministry of Agriculture, Animal In-
G. Ramachandran1, L. Baillie2, P. Tsai3, G. Rosen3, A. Cross1; 1Univ. of Mary- dustry and Fisheries, Entebbe, Uganda, 2Robert Heckert Consulting LLC,
land Med. Sch., Baltimore, MD, 2Welsh Sch. of Pharmacy, Cardiff, United Bowie, MD, 3Ministry of Hlth., Kampala, Uganda, 4AFENET, Kampala,
Kingdom, 3Dept. of Pharmaceutical Sci., Baltimore, MD. Uganda, 5Uganda Wildlife Authority, Kampala, Uganda, 6Uganda Virus Res.
Inst., Entebbe, Uganda, 7Booz Allen Hamilton, Middletown, DE.

BIODEFENSE PROGRAM & ABSTRACTS 19


MO N DAY S C I E N T I F I C P RO G R A M

Highlighted Oral Abstract Presentations 3:15PM


113 Distinct Differences in Disease Presentation after Aerosol
009. Environmental Detection Exposure to Rift Valley Fever Virus Dependent on Inbred Rat Strain
Monday, February 27, 2012 | 3:00PM – 4:00PM J. M. Bales, D. Powell, L. M. Bethel, D. Reed, I. Pandrea, A. L. Hartman; Univ.
of Pittsburgh, Pittsburgh, PA.
Diplomat Ballroom
MODERATOR: 3:30PM
Miles Carroll; Hlth. Protection Agency, Wiltshire, United Kingdom. 114 Interplay between RVFV and the Host NF-KappaB Signaling
Cascade
PRESENTATIONS:
A. Narayanan, K. Kehn-Hall, L. Hill, S. Senina, R. VanDuyne, I. Guendel, R.
3:00PM Das, A. Baer, C. Bailey, F. Kashanchi; George Mason Univ., Manassas, VA.
108 Case-Clusters of Melioidosis Associated with Climate 3:45PM
Conditions and Geographical Topography in Southern Taiwan
Y-L. Chen1, Y-S. Chen2, H-S. Lin3, P-J. Liu1; 1Natl. Kaoshiung Normal Univ.,
115 Interactions of Venezuelan Equine Encephalitis Virus with
Kaohsiung, Taiwan, 2Kaohsiung Veterans Gen. Hosp., Kaohsiung, Taiwan,
Host MicroRNA Processing Machinery
3
E-DA Hosp., Kaohsiung, Taiwan. C. A. King, N. Shafagati, I. Guendel, F. Kashanchi, K. Kehn-Hall; George
Mason Univ., Manassas, VA.
3:15PM
109 Environmental Contamination of Goat Farms with Coxiella
burnetii During a Q Fever Outbreak
Highlighted Oral Abstract Presentations
G. J. Kersh, K. A. Fitzpatrick, A. V. Kondas, A. J. Roche, J. S. Self, R. A. Priest- 011. Bacterial Agents
ley, A. Bjork, A. D. Anderson; CDC, Atlanta, GA.
Monday, February 27, 2012 | 3:00PM – 4:00PM
3:30PM Palladian Ballroom
110 Rapid and Sensitive On-Site Identification of Biothreat MODERATOR:
Agents Using Electrochemical Biochips and a Portable Detection
Platform Petra F. Oyston; DSTL, Salisbury, United Kingdom.
T. Elssner, C. Pöhlmann; Bruker Daltonik GmbH, Leipzig, Germany. PRESENTATIONS:

3:45PM 3:00PM
111 Detection of Botulinum Neurotoxin Serotypes A and B Using 116 Temperature-Regulated Membrane Remodeling is Essential
Chemiluminescence and Electrochemiluninescene Immunoassays for Francisella Pathogenesis
in Food and Serum Matrices Y. Li1, D. A. Powell1, S. A. Shaffer2, D. A. Rasko3, D. R. Goodlett2, C. R. H.
L. W. Cheng, L. H. Stanker; USDA, Albany, CA. Raetz4, X. Wang5, R. K. Ernst1; 1Univ. of Maryland, Baltimore, MD, 2Univ. of
Massachusetts Med. Sch., Worcester, MA, 3Univ. of Maryland Sch. of Med.,
Baltimore, MD, 4 Duke Univ. Med. Ctr., Durham, NC, 5State Key Lab. of Food
Highlighted Oral Abstract Presentations Sci. and Technology, Wuxi, China.

010. Viral Agents 3:15PM


Monday, February 27, 2012 | 3:00PM – 4:00PM 117 Development of a Luciferase Reporter System to Monitor
Plague Infection
Ambassador Ballroom
Y. Sun, J. Pennington, M. G. Connor, J. M. Warawa, M. B. Lawrenz; Univ. of
MODERATORS: Louisville, Louisville, KY.
Jean Patterson; Texas Biomedical Res. Inst., San Antonio, TX. 3:30PM
Colleen Jonsson; Univ. of Louisville, Louisville, KY.
118 A Variant of LcrV, the Plague Protective Antigen and Needle
PRESENTATIONS: Cap Protein of Yersinia pestis that Blocks Type III Secretion
3:00PM K. Given1, A. Mitchell2, N. Miller1, O. Schneewind1; 1Univ. of Chicago, Ar-
gonne, IL, 2Univ. of Chicago, Chicago, IL.
112 First Evidence of Hantavirus Circulation in Terrestrial Small
Mammals From Madagascar 3:45PM
J-M. Heraud1, N. Razafindralambo1, T. andrianaivo2, M-M. Olive1, V. L. Soari- 119 Hide and Seek: Changes in Francisella Surface Proteins
malala2, S. M. Goodman2, J-M. Reynes3; 1Inst. Pasteur de Madagascar, An- During Infection Reveal Ideal Vaccine Targets
tananarivo, Madagascar, 2Association Vahatra, Antananarivo, Madagascar,
3 J. Huntley; Univ. of Toledo Coll. of Med., Toledo, OH.
Inst. Pasteur, Lyon, France.

20 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY S C I E N T I F I C P RO G R A M

Symposium Session Symposium Session


012. Biodefense at the Dinner Table: Emerging 013. Persistence and Inactivation of Biothreat
Foodborne Illnesses Agents and Emerging Pathogens
Monday, February 27, 2012 | 4:15 PM - 6:15 PM Monday, February 27, 2012 | 4:15 PM - 6:15 PM
Diplomat Ballroom Palladian Ballroom
This symposium will explore mechanisms of pathogenesis of a num- Following the attacks of 2011, it was quickly realized that persistence
ber of foodborne viral and bacterial pathogens. A key goal in under- and inactivation data on biothreat agents was lacking. Very few stud-
standing enteric infections is elucidating the mechanisms by which ies in this area had been conducted since the 1960s. In addition to
epithelial barriers are evaded, and the impacts that systemic co-in- summarizing the progress that has been made over the last decade,
fections can exert. Understanding these phenomena will inform the research presented will inform the research microbiologists of the
strategies to prevent and treat foodborne infections. The scope in the need for additional like studies to form risk assessments, remediation
symposium will be from clinical and molecular epidemiology studies efforts and biopreparedness planning. A better understanding of the
to molecular genetics, cell biology and ultrastructure. persistence and inactivation parameters necessary for remediation will
improve response timelines associated with contamination events.
MODERATORS:
Marshall E. Bloom; DIR/NIAID/NIH, Hamilton, MT. MODERATORS:

Melita A. Gordon; Univ. of Liverpool, Liverpool, United Kingdom. Gene Rice; EPA, Cincinnati, OH.
Tonya Nichols; EPA, Washington, DC.
PRESENTATIONS:
4:15PM
4:15PM
124 Persistence of Category A Select Agents
120 Mechanisms of Murine Norovirus Interaction with the Chuck Gerba; Univ. of Arizona, Tucson, AZ.
Intestinal Epithelium
Christiane Wobus; Univ. of Michigan Med. Sch., Ann Arbor, MI. 4:45PM

4:45PM 125 Persistence and Inactivation of Vaccinia Virus


Joseph Wood; EPA, Research Triangle Park, NC.
121 Picking the Lock: Stealth Invasion of Epithelial Barriers by
Enteric Pathogens 5:15PM
Manuel R. Amieva; Stanford Univ. Sch. of Med., Stanford, CA. 126 Inactivation of Bacterial Biothreat Agents in Water
5:15PM Laura Rose; CDC, Atlanta, GA.

122 Multi-Dimensional Analysis of E. coli Colonization of 5:45PM


Mammalian Cells and Green Leafy Vegetables 127 Decontamination of Bacillus Spores from Water Using
Jorge A. Giron; Univ. of Florida, Gainesville, FL. Germinant
5:45PM Jeff Szabo; USEPA/NHSRC, Cincinnati, OH.

123 Clinical and Molecular Epidemiology of Invasive Salmonella


Disease in Africa Adults and Children
Melita Gordon; Univ. of Liverpool, Liverpool, United Kingdom.

BIODEFENSE PROGRAM & ABSTRACTS 21


MO N DAY S C I E N T I F I C P RO G R A M

Symposium Session Roundtable Discussion


014. Taking the Bite Out of Insect Bites: Biodefense 015. Recapping the 2011 Biological Weapons
Implications of Vector-Borne Infections Convention Review Conference (BWC RevCon)
Monday, February 27, 2012 | 4:15 PM - 6:15 PM Monday, February 27, 2012 | 6:30 PM - 7:30 PM
Ambassador Ballroom Palladian Ballroom
The scope of this session is to focus on the impact of arthropod vec- Awareness of safety and security issues is fundamental to the re-
tors on the kinetics of bacterial, viral and parasitic infections affecting sponsible conduct of work in the life sciences. This session will con-
global health and implications for emerging and biodefense-related tribute to the overall effectiveness of the microbiologist who attends
diseases. The topics will broadly explore changes in distribution of this meeting by increasing awareness of the Biological Weapons Con-
vectors and disease prevalence; effect of ecological changes and its vention, with particular attention to international efforts directly rel-
impact on vector-borne infections; novel preventive strategies against evant to the life of the microbiologist, including those related to
vector-borne infections; and biome to genome tweets: Vector- laboratory biosecurity and biosafety, regulation, and efforts to engage
pathogen interactions. These areas are at the cutting-edge of vector- the scientific community in discussions of education, awareness-rais-
borne disease research. An understanding of the current trends in ing, and self-governance.
these areas will help in developing strategies to combat several
emerging diseases with significant impact on global health. Strate- MODERATOR:
gies to prevent vector-borne diseases using novel vector-specific TBD
chemotherapeutics would add to the list of prevention strategies for PANELISTS:
vector-borne infections.
132 Christopher Park; U.S. Dept. of State, Washington, DC.
MODERATORS: 133 Gregory Koblentz; George Mason Univ., Fairfax, VA.
Janakiram Seshu; Univ. of Texas, San Antonio, TX. 134 Gerald L. Epstein; U.S. Dept. of Homeland Security, Washington, DC.
Karl E. Klose; Univ. of Texas, San Antonio, TX.
PRESENTATIONS:
4:15PM
128 Borrelia burgdorferi-Tick Interactions: New Insight into
Ixodes Proteins Modulating Pathogen Persistence in the Vector
Utpal Pal; Univ. of Maryland, College Park, MD.

4:45PM
129 Dissecting the Mechanisms of Tick Transmission from
Anaplasma to Francisella
Susan M. Noh; USDA-ARS Animal Disease Res. Unit, Pullman, WA.

5:15PM
130 Characterizing 24-48h Tick Saliva Proteins to Understand
Early Stage Tick Feeding Biology
Albert Mulenga; Texas A&M Univ., College Station, TX.

5:45PM
131 Tweets from Ticks: Modulation of Vector-Specific Gene
Expression by BadR in the Agent of Lyme Disease
Janakiram Seshu; Univ. of Texas, San Antonio, TX.

22 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY S C I E N T I F I C P RO G R A M

10:30AM
Tuesday, February 28, 2012 138 Targeting Cyclic Di-GMP Signaling to Prevent Biofilm
Formation
Chris Waters; Michigan State Univ., East Lansing, MI.
016. Global Threats, Collaborative Solutions
11:00AM
Tuesday, February 28, 2012 | 7:15AM – 8:15AM
139 Identification of Inhibitors of DNA Adenine Methylation as
Palladian Ballroom Novel Therapeutics
This new program will provide insights into current international se- Petra Oyston; DSTL, Salisbury, United Kingdom.
curity challenges from notable thought leaders, policy advisors, and
senior government officials.
Poster Session
MODERATOR:
018. Tuesday Poster Session
Dr. Jason Rao; ASM Director of International Affairs, Washington, DC.
Tuesday, February 28, 2012 | 1:00PM – 3:00PM
PRESENTATION:
Exhibit Hall
Dr. Alan Rudolph; Director for Chemical and Biological Technologies Direc-
torate, Research and Development Enterprise, Defense Threat Reduction PRESENTATIONS:
Agency (DTRA), Fort Belvoir, VA. 140 (A) Molecular Epizootiology of Street Rabies Virus Isolates
in Ukraine
I. Polupan, N. Ivanov, O. Deryabin, V. Skripnik; Inst. of Vet. Med., Kyiv,
Ukraine.

Plenary Session 141 (A) Utility of a New Real-Time RT-PCR Assay for the Detection
of Crimean-Congo Haemorrhagic Fever Virus in Pakistan
017. Targeting Virulence Factors in Acute Infection B. Atkinson1, Z. Hasan2, A. Samreen2, B. Jamil2, T. Moatter2, J. Chamberlain1,
N. Cook1, C. Logue1, C. Bruce1, S. Dowall1, R. Hewson1; 1Hlth. Protection
Tuesday, February 28, 2012 | 8:30AM - 12:00PM
Agency UK, Salisbury, United Kingdom, 2The Aga Khan Univ., Karachi, Pak-
Regency Ballroom istan.
Antibiotic resistance is a global crisis caused by antibiotic overuse, 142 (A) Development of Differential and Broad Spectrum
misuse and a dearth of new targets under development. This session Immunological Reagents for Identification and Characterization
will examine which approaches have been successful and brought to of Filovirus Sub-Types
the clinic (toxin inhibitors, type three secretion, for example) and C. Bruce, K. Richards, L. Easterbrook, H. Love, L. Hudson, J. Plank, H. Tolley,
which have not yet succeeded and the underlying difficulties for these R. Hewson, A. Robers; Hlth Protection Agency, Salisbury, United Kingdom.
relative failures (e.g. reaching intracellular microbial targets within
tissues, specificity, redundancy, etc.). 143 (A) Fighting Fire with Fire: The Development of Antiviral AAVs
K. K. Wong, Jr.; City of Hope Natl. Med. Ctr., Duarte, CA.
MODERATORS:
144 (A) Arenavirus Reverse Genetics for Vaccine Development
Petra F. Oyston; DSTL, Salisbury, United Kingdom.
E. Ortiz Riano1, B. Y. H. Cheng1, J. C. de la Torre2, L. Martinez-Sobrido1; 1
Nancy Connell; UMDNJ-New Jersey Med. Sch., Newark, NJ. Univ. of Rochester Med. Ctr., Rochester, NY, 2The Scripps Res. Inst., La Jolla,
PRESENTATIONS: CA.
8:30AM 145 (A) New Select Agent Regulations
135 Antimicrobial Peptide Modulation of Chemokine and Pro- J. Garrett, V. Sutton, H. Carson; Texas Tech Univ. Sch. of Law, Lubbock, TX.
Inflammatory Cytokine Responses: It’s all in the Timing
146 (A) Development of a Murine Model for Aerosolized Filovirus
Kim A. Brogden; Univ. of Iowa, Iowa City, IA.
Infection
9:00AM E. E. Zumbrun, H. A. Bloomfield, T. B. Chance, D. K. Nichols, A. Nalca; US-
136 Anthrax Diagnostic Tools (Nonclinical and Clinical) for AMRIID, Fort Detrick, MD.
Medical Countermeasure Development Under the Animal Rule 147 (B) Antibiotic Profile of Nosocomial Bacterial Pathogens
Elizabeth Leffel; PharmAthene, Inc., Annapolis, MD. N. N. Eduok1, I. P. Udoh2, C. I. Eleazar2, B. O. Ogeneh2; 1Ministry of Sci.
9:30AM and Technology, Uyo, Nigeria, 2Univ. of Nigeria, Enugu, Nigeria.

137 148 (B) Comparative Transcriptomics of Three Species of Fleas


Christian Baron; Univ. de Montreal, Montreal, Canada. (Siphonaptera: Pulicidae)
C. A. Whitehouse1, G. Koroleva1, J. R. Kugelman1, S. K. Williams2, S. W.
10:00AM
Bearden2, K. L. Gage2, G. F. Palacios1; 1USAMRIID, Frederick, MD, 2CDC,
Coffee Break Fort Collins, CO.

BIODEFENSE PROGRAM & ABSTRACTS 23


T U E S DAY S C I E N T I F I C P RO G R A M

149 (B) Development of a Coxiella burnetii Aerosol Infection 159 (B) Anthrax Surveillance System and Canonical SNP Typing
Model in Rodents of Bacillus anthracis Strains Isolated in the Country of Georgia
K. R. Bewley, J. K. Pitman, D. J. Atkinson, G. McLuckie, M. Charlwood, G. 2009-2011
Hatch, J. Vipond, S. G. P. Funnell, A. Roberts; Hlth. Protection Agency, G. Chanturia1, E. Khmaladze1, E. Zhgenti1, L. Malania1, M. Zakalashvili1,
Salisbury, United Kingdom. N. Tsertsvadze1, M. Kekelidze1, P. Imnadze1, S. Tsanava1, R. J. Obiso2, S.
Francesconi3; 1The Natl. Ctr. for Disease Control and Publ. Hlth., Tbilisi,
150 (B) Automated Extraction of Genomic DNA from Food- Georgia, 2The Microbe Company, Christiansburg, VA, 3Naval Med. Res. Ctr.,
Associated Pathogens and Environmental Samples Frederick, MD.
P. Williams, T. Parrish, M. Duggan, J. Chapman, W. Burke, J. Birkenholz;
Evogen, Lenexa, KS. 160 (B) Diversity of Bacteriophages Active to B. anthracis
Bacterial Strains Isolated in Georgia
151 (B) Monoclonal Antibody against Capsule of Bacillus S. Rigvava1, M. Natidze2, L. Gubeladze1, L. Kvachadze1, R. J. Obiso3,
anthracis Protects Mice from Enhanced Lethal Toxin Activity Due M. Kutateladze1, R. Adamia1; 1G. Eliava Inst. of Bacteriophages, Microbiol.
to Capsule and Anthrax Spore Challenge and Virology, Tbilisi, Georgia, 2G, Tbilisi, Georgia, 3The Microbe Company,
G-E. Rhie1, J. Jang1, M. Cho1, J-H. Chun1, J. Park2, K. Hong1; 1Korea NIH, Christiansburg, VA.
Chungbuk 363-951, Korea, Republic of, 2Hankuk Univ. of Foreign Studies, Yon-
gin, Korea, Republic of. 161 (B) Characterisation of Inhalational Infection with
Burkholderia pseudomallei in BALB/c mice
152 (B) The Cat Flea as a Laboratory Model for Understanding S. G. P. Funnell, G. Hatch, J. A. Kane, S. Bates, K. M. Tonks, C. H. Tsang,
Yersinia pestis Colonization of its Insect Vector J. Fretwell, H. Shuttleworth, J. Vipond, A. Roberts; Hlth. Protection Agency,
T. M. Hermanas, L. E. Quenee, O. Schneewind; Univ. of Chicago, Chicago, Salisbury, United Kingdom.
IL.
162 (B) Reaerosolization of Bacillus Spores During the Bio-
153 (B) Bacillus anthracis Acetyl-Transferases PatA1 and PatA2 Response Operational Testing and Evaluation
and Secondary Cell Wall Polysaccharide-Mediated Assembly of S- S. C. Taft1, R. A. Lordo2, D. J. Chappie2, E. Silvestri1, T. L. Nichols1, J. A. Rosati
Layer Proteins Rowe1; 1Natl. Homeland Security Res. Ctr., Cincinnati, OH, 2Battelle Mem.
J. M. Lunderberg1,2, S-M. Nguyen-Mau1,2, G. S. Richter1,2, Y-T. Wang1,2, J. Inst., Columbus, OH.
Dworkin3, D. M. Missiakas1,2, O. Schneewind1,2; 1Univ. of Chicago, Chicago,
IL, 2Howard Taylor Ricketts Lab., Argonne, IL, 3Columbia Univ., New York, 163 (B) Regulation of the Yersinia Type Three Secretion Pathway
NY. K. Kopaskie1,2, J. McCool3, K. Aly1, B. Blaylock1,2, O. Schneewind1,2; 1Univ. of
Chicago, Chicago, IL, 2Howard Taylor Ricketts Lab., Argonne, IL, 3Boston
154 (B) Autotransported (Type V Secreted) Proteins of Univ., Boston, MA.
Burkholderia pseudomallei and their Role in Biofilm Formation,
Serum Resistance and Pathogenesis 164 (B) Real-Time Monitoring of Bacillus thuringiensis as a
N. R. Lazar Adler1, R. E. Dean2, M. P. Stevens3, J. L. Prior2, T. P. Atkins2,4, E. E. Surrogate for Bacillus anthracis in Potable Water Distribution
Galyov1; 1Univ. of Leicester, Leicester, United Kingdom, 2Defence Sci. and System
Technology Lab., Salisbury, United Kingdom, 3Univ. of Edinburgh, Easter S. P. Sherchan, I. Pepper; Univ. of Arizona, Tucson, AZ.
Bush, United Kingdom, 4Univ. of Exeter, Exeter, United Kingdom.
165 (B) Achromobacter Xylosoxidans: An Emerging Pathogen
155 (B) S-Layer Assembly in Bacillus cereus G9241 Requires csaB Carrying Different Elements Involved In Horizontal Genetic
Y-T. Wang1,2, S-Y. Oh1,2, A. P. Hendrickx1, O. Schneewind1,2; 1Univ. of Transfer
Chicago, Chicago, IL, 2Howard Taylor Ricketts Lab., Argonne, IL. G. Traglia1, C. Adams2, M. Almuzara3, C. Vay3, D. Centrón1, M. Ramirez2,1;
1
Sch. of Med., Buenos Aires, Argentina, 2California State Univ., Fullerton,
156 (B) Antibody Interactions with the Capsule of Bacillus CA, 3Hosp. de Clinicas, Buenos Aires, Argentina.
anthracis
M. A. Hubbard, P. Thorkildson, W. Welch, D. AuCoin, T. Kozel; Univ. of Ne- 166 (B) Increase in Temperature of Cultivation Above 37°C
vada Sch. of Med., Reno, NV. Influenced Production of Yersinia pestis Major Protective Antigens
F1 and LcrV
157 (B) Salmonella Enteritidis Gene SEN1140 Contributes to V. A. Feodorova1, T. I. Polyanina1, M. A. Khizhnyakova1, A. M. Lyapina1, O. V.
Accelerated Type III Secretion System-2 Dependent Early Ulianova2,1, V. L. Motin3; 1Saratov State Vet. Inst., Saratov, Russian Federa-
Inflammation Kinetics in Mouse Colitis Model tion, 2Saratov State Univ., Saratov, Russian Federation, 3Univ. of Texas Med.
V. Vishwakarma1, B. Periaswamy2, N. B. Pati1, W-D. Hardt2, M. Suar1; 1Sch. Branch, Galveston, TX.
of Biotech., Bhubaneswar, India, 2Inst. of Microbiol., Zürich, Switzerland.
167 (B) Infection and Dissemination, Francisella tularensis
158 (B) Activity of Two Novel Antibacterial Agents XF-70 & XF-73 holarctica in Mosquito Vector
against Multidrug-Resistant Acinetobacter baumannii S. Backman; CBRN Defence and Security, Umea, Sweden.
K. L. Lohman1, J. Coffin1, W. Rhys-Williams2, I. Hayter2, W. G. Love2; 1MRI
Global, Rockville, MD, 2Destiny Pharma Ltd., Brighton, United Kingdom.

24 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY S C I E N T I F I C P RO G R A M

168 (B) Phylogeography of Bacillus anthracis Isolates from the 179 (D) Development of Single Domain Antibodies for Bacillus
Country of Georgia anthracis Spore Proteins
E. Khmaladze1, D. Birdsell2, E. Zhghenti1, G. Chanturia1, M. Kekelidze1, S. A. Walper1, P. Lee2, J. Liu3, G. Anderson3, E. Goldman3; 1Natl. Academy of
M. Nikolich3, P. Imnadze1, S. Tsanava1, L. Malania1, N. Tsertsvadze1, Sci., Washington, DC, 2Nova Res. Inc., Alexandria, VA, 3Naval Res. Lab.,
S. Beckstrom-Sternberg4, J. Beckstrom-Sternberg4, T. Pearson2, D. Wagner2, Washington, DC.
P. Keim2; 1Natl. Ctr. for Disease Control and Publ. Hlth., Tbilisi, Georgia,
2
Northern Arizona Univ., Flagstaff, AZ, 3Walter Read Army Inst. of Res., Fort 180 (D) Zeptomole/ML Quantification of Anthrax Edema Factor by
Detrick, MD, 4Translational Genomics Res. Inst., Phoenix, AZ. LC-ESI-MS/MS: Activity Enhancement by Monoclonal Antibodies
R. C. Lins1, Z. Kuklenyik2, C. P. Quinn2, K. Bedi2, J. Goldstein2, A. Lyons2,
169 (B) Polymorphisms in the lcrv Gene of Yersinia enterocolitica M. Gallegos2, U. Ansari1, R. Pitts1, D. Bagarozzi1, K. Isbell1, C. E. Leysath3,
Do Not Provide for Escape from Plague Protective Immunity Z. Chen3, S. H. Leppla3, J. R. Barr2, A. Boyer2; 1Battelle CDC, Atlanta, GA,
2
N. C. Miller, L. Quenee, D. Elli, N. Ciletti, O. Schneewind; Univ. of Chicago, CDC, Atlanta, GA, 3NIH, Bethesda, MD.
Chicago, IL.
181 (E) Simultaneous Detection of Ricin and Abrin DNA by Real-
170 (B) Multiple-Locus Variable Number Tandem Repeat Analysis Time PCR
of Brucella melitensis from Azerbaijan Using Eight Markers I. Mossbrugger, E. Felder, R. Wölfel; Bundeswehr Inst. of Microbiol.,
X-Z. Huang, E. B. Zelazowska, M. P. Nikolich; Walter Reed Army Inst. of Res., Munich, Germany.
Silver Spring, MD.
182 (E) Low-Cost, Highly Efficient Airborne Pathogen Collection
171 (B) Rapid Single Nucleotide Repeat Sequence-Based and Detection System for Field Use
Detection and Identification of B. anthracis Using Pyrosequencing J. Gordon1,2, P. Gandhi1; 1Inspirotec LLC, Chicago, IL, 2Northwestern Univ.,
Technology Evanston, IL.
K. R. Hahn, E. Valle, M. Thomas, T. Janzen, M. Shields, N. Goji, K. Amoako;
Canadian Food Inspection Agency, Lethbridge, Canada. 183 (E) Evaluation of Automated and Manual Commercial Nucleic
Acid Extraction Methods for Detection of Ricinus communis DNA
172 (B) Beef and Chicken Contamination with Resistant by Real-Time PCR
Escherichial coli O157 from City Abattoirs in Southwest Nigeria A. S. Hutchins1, M. Astwood2, J. R. Saah1, L. A. Dauphin2; 1North Carolina
O. Olatoye, A. E. Amosun; Univ. of Ibadan, Ibadan, Nigeria. State Lab. of Publ. Hlth., Raleigh, NC, 2 CDC, Atlanta, GA.

173 (B) Contribution of OxyR to Burkholderia mallei Survival and 184 (E) Development of Multiplex Rapid Tests Allowing Warfare
Virulence In Vitro and In Vivo Agents Detection
D. A. Revelli, J. Boylan, F. Gherardini; Rocky Mountain Lab., NIAID/NIH, T. Berthelot, J. Credou, H. Volland; CEA, Gif sur Yvette, France.
Hamilton, MT.
185 (E) Validation of Unique Signature Discovery in Select Agents
174 (B) Growth Inhibition of Burkholderia pseudomallei by Onion and an Emerging Pathogen
Isolates of Burkholderia cepacia B. J. Haley1, E. Taviani1, A. Chen1, A. Phillippy2, A. Huq1,3, R. R. Colwell1,4,5,
J. M. Inamine, T. M. Eckstein; Colorado State Univ., Fort Collins, CO. I. Knight3,6; 1Maryland Pathogen Res. Inst., College Park, MD, 2Ctr. for Bioin-
formatics and Computational Biology, College Park, MD, 3Maryland Inst. for
175 (D) Quality Assurance for Select Agents: An Australian Applied Environmental Hlth., College Park, MD, 4Univ. of Maryland Inst. for
Approach Advanced Computer Studies, College Park, MD, 5CosmosID, College Park,
T. Theis1, D. Liu1, J. Gray1, P. Roffey2, W. Rawlinson3,1; 1RCPA BioSecurity MD, 6Canon U.S. Life Sci., Inc., Rockville, MD.
QAP, Surry Hills, Australia, 2Australian Federal Police, Weston, Australia,
3
SEALS Microbiol. Prince of Wales Hosp., Randwick, Australia. 186 (E) Novel Surface Chemistry For The Development of
Multiplex Immunochromatographic Tests
176 (D) Broad-Range Microbial DNA Isolation from Clinical J. Credou, H. Volland, T. Berthelot; CEA, Gif sur Yvette, France.
Specimens for Universal PCR Diagnosis
C. Disqué1, M. Linow1, N. Murphy2; 1Molzym GmbH & Co.KG, Bremen, 187 (F) Virus Pathogen Resource (ViPR): An Open Bioinformatics
Germany, 2CaerusBio Inc., Downingtown, PA. Database and Analysis Resource for Human Virus Pathogen
Research
177 (D) Y. Zhang1, B. Pickett1, E. Sadat1, J. Noronha1, R. B. Squires1, V. Hunt1,
WITHDRAWN M. Liu2, S. Kumar3, S. Zaremba3, Z. Gu3, L. Zhou3, C. Larson4, J. Dietrich3, E.
Klem3, R. H. Scheuermann1; 1Univ. of Texas Southwestern Med. Ctr., Dallas,
178 (D) Laboratory Surveillance of Tick Born Causing Agents and TX, 2Southern Methodist Univ., Dallas, TX, 3Northrop Grumman Hlth. Solu-
Other Relevant Pathogens in the State of Maryland tions, Rockville, MD, 4Vecna Technologies, Greenbelt, MD.
M. Dembele1, L. Trivedi2, T. Lawson2, S. Montgomery2, M. P. Carlos2,1; 1Johns
Hopkins Sch. of Publ. Hlth., Baltimore, MD, 2Maryland Dept. of Hlth. and 188 (F) Effects on Host Responses of Air-Borne and Aerosolized
Mental Hygiene Lab. Admin., Baltimore, MD. Rabbitpox Infections in New Zealand Rabbits
L. DaSilva1, A. Porter1, A. Nalca1, R. Settlage2, C. L. Galindo3, L. McIver2,
K. McMahon2; 1USAMRIID, Frederick, MD, 2Virginia Bioinformatics Inst. at
Virginia Polytechnic Inst., Blacksburg, VA, 3Eagle Eye Data Analysis, Hous-
ton, TX.

BIODEFENSE PROGRAM & ABSTRACTS 25


T U E S DAY S C I E N T I F I C P RO G R A M

189 (F) Analysis of Next-Generation Sequencing for Detecting 200 (H) Longevity of Vaccine Protection against Pneumonic
Rare Variants in Bacillus anthracis Plague in BALB/C Mice, Part 2
R. E. Colman1, J. Schupp1, J. Gillece1, A. Rawat1, D. M. Engelthaler1, J. Fos- Z. N. Llewellyn1, F. Koide2, L. E. Bowen1, P. William2, S. Lin2, J. A. Boydston1,
ter2, P. Keim2,1; 1Translational Genomics Res. Inst., Flagstaff, AZ, 2Northern N. Harman-Richardson3, P. M. Silvera2; 1Southern Res. Inst., Birmingham,
Arizona Univ., Flagstaff, AZ. AL, 2Southern Res. Inst., Frederick, MD, 3Alpha StatConsult, LLC,
Damascus, MD.
190 (F) Nested Amplification for Improved Genotyping of Coxiella
burnetii 201 (H) Dose Response of IMVAMUNE, a Third Generation
R. Vipond, A. Pepler, G. Vincent, J. Latham; Hlth. Protection Agency, Salis- Smallpox Vaccine Candidate, in a Lethal Monkeypox Virus
bury, United Kingdom. Nonhuman Primate Inhalation Model
T. Brasel, K. Agans, K. Dearen, T. Bailey, A. Russo, S. Storch, R. Sherwood;
191 (F) Utility and Challenges Associated with Use of Next Lovelace Respiratory Res. Inst., Albuquerque, NM.
Generation Sequencing in Ultra-Rare Variant Detection
H. Koshinsky1, V. Fofanov2, J. Liu2; 1Eureka Genomics, Hercules, CA, 2Eureka 202 (H) Stabilization Technology for Biodefence Vaccines:
Genomics, Houston, TX. Exemplification Using a New Viral Vaccine and an Adjuvanted
Vaccine
192 (G) PANACEA Broad-Spectrum Antiviral Therapeutics J. Drew, C. Andrews, E. Abbott, S. Ward; Stabilitech Ltd., London, United
T. H. Rider; Massachusetts Inst. of Technology, Cambridge, MA. Kingdom.

193 (G) Efficacy of a Doxycycline Treatment Regimen in Rhesus 203 (H) A Marmoset Model for Lethal Orthopox Virus Infection
Macaques Aerosol Challenged with Brucella melitensis Enables Study of Vaccines
L. Henning, E. McGuinness, D. Pak, S. Miller, C. Briscoe, M. Anderson, C. Yue1, M. Kramski1,2, C. Stahl-Hennig3, K. Mätz-Rensing3, F-J. Kaup3, G.
R. Warren; Battelle, Columbus, OH. Pauli1, H. Ellerbrok1; 1Robert Koch Inst., Berlin, Germany, 2Univ. of Mel-
bourne, Parkville, Australia, 3German Primate Ctr., Goettingen, Germany.
194 (G) Host-Directed Antimicrobial Therapeutics for Intracellular
Bacterial Pathogens 204 (H) New Adjuvants for Anthrax Vaccine
A. Levi1, D. Arvans2, P. Robins2, L. P. Potluri1, B. Heaton1,2, S. Riley1,2, S. Y. Gauthier1, A. Borel2, Y. Trescos1, J. Mathieu1, B. Verrier2, J-N. Tournier1;
Crosson1,2, J. Martinez1,2, J. E. Gabay1, H. A. Shuman2,1; 1Univ. of Chicago, 1
IRBA, La Tronche, France, 2CNRS-IBCP, Lyon, France.
Chicago, IL, 2Howard Taylor Ricketts Lab., Lemont, IL.
205 (H) Effects of Signal Sequences on the Immunogenicity and
195 (G) Restoring Antibiotic Sensitivity with Antisense Inhibitors Efficacy of an MVA Vectored Plague Vaccine
of Gene Expression J. Brewoo1, T. Powell2, G. Young1, C. Partidos1, D. Stinchcomb2, J. Osorio3;
P. L. Iversen, D. V. Mourich, F. Schnell, B. Geller; AVI BioPharma, Inc., 1
Inviragen, Inc., Madison, WI, 2Inviragen, Inc., Fort Collins, CO, 3Univ. of
Bothell, WA. Wisconsin, Madison, WI.

196 (G) Post-Exposure Administration of a Monoclonal Antibody 206 (H) Second-Generation Inactivated Vaccines Protect Mice
Cocktail is Protective in Orthopoxvirus-Infected Mice and from Lethal Venezuelan Equine Encephalitis Virus Infection
Cynomolgus Macaques K. Spurgers1, S. Honnold1, R. Bakken1, J. Cohen1, L. Rowan1, C. Lind1, P.
J. L. Nordstrom1, G. J. Rainey1, H. Li1, P. Silvera2, F. Koide2, M. Buller3, J. Gupta2, A. Sharma2, R. Maheshwari2, P. Glass1; 1USAMRIID, Frederick, MD,
Schriewer3, A. Dekonenko4, K. Shah1, L. Huang1, V. Ciccarone1, S. Burke1, M. 2
USUHS, Bethesda, MD.
Bowe1, D. Farb1, M. Kadan1, E. Bonvini1, K. Stein1, S. Koenig1, S. Johnson1;
1
MacroGenics, Rockville, MD, 2Southern Res. Inst., Frederick, MD, 3Saint 207 (H) Evaluation of Inactivated and Live-Attenuated Bivalent
Louis Univ., St. Louis, MO, 4Univ. of New Mexico, Albuquerque, NM. Vaccines that Confer Protection against Rabies and Ebola Viruses
in Mice
197 (H) Clinical Development of a Plant-Produced Subunit J. Blaney1, A. Papaneri1, C. Wirblich2, J. Cann1, M. Holbrook1,3, A. Freiberg3,
Vaccine against Bacillus anthracis P. Jahrling1, M. Schnell2; 1NIAID/NIH, Fort Detrick, MD, 2Jefferson Med. Coll.,
V. Yusibov, J. Chichester, S. Manceva, K. Musiychuk, M. Shamloul, A. Rhee, Thomas Jefferson Univ., Philadelphia, PA, 3Univ. of Texas Med. Branch,
J. Trefethen, M. Jones; Fraunhofer USA CMB, Newark, DE. Galveston, TX.

198 (H) Adoptive Transfer of Pulsed BMDC Enhances Specific 208 (H) Improved Efficacy of a Multiagent Equine Encephalitis
Immune Responses and Provides Protection against Lethal Virus Vaccine Candidate
Anthrax Challenge P. J. Glass1, R. R. Bakken1, J. Ennis2, K. B. Spurgers1, K. I. Kamrud3,
I. J. Thompson1, M. G. Stokes1, B. Hallis2, A. J. Simpson1, E. D. Williamson1; K. Alterson3, M. Custer3, J. Turner2; 1USAMRIID, Fort Detrick, MD, 2Defyrus
1
Defense Sci. and Technology Lab., Salisbury, United Kingdom, 2Hlth. Pro- Inc., Toronto, Canada, 3Alphavax, Research Triangle Park, NC.
tection Agency, Salisbury, United Kingdom.
209 (H) Relationship between Vaccine Dosage, Immunogenicity
199 (H) Radioprotective Mn-DP-Pi Complexes Preserve and Survival in rF1V-Vaccinated Cynomolgus Macaques
Immunogenicity of Inactivated VEEV During Gamma Irradiation: P. Fellows1, J. Price1, R. Krile2, H. Lockman2, L. Wolfraim1; 1Dynport Vaccine
A New Approach for Vaccine Development Company LLC, Frederick, MD, 2Battelle, Columbus, OH.
M. Gayen1,2, E. K. Gaidamakova1, P. Gupta1,2, A. Sharma1, P. J. Glass3, M. J.
Daly1, R. K. Maheshwari1; 1Uniformed Services Univ. of Hlth. Sci., Bethesda,
MD, 2Birla Inst. of Tech. and Sci., Pilani, India, 3U.S. Army Med. Res. Inst. of
Infectious Diseases, Frederick, MD.

26 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY S C I E N T I F I C P RO G R A M

210 (I) Peptide Mapping of Anthrax Toxin Protective Antigen Highlighted Oral Abstract Presentations
Using Anti-AVA Human Sera
V. A. Semenova, P. Svoboda, J. Pohl, S. Soroka, C. Quinn; CDC, Atlanta, GA. 019. Immune Response
211 (I) Serum Opsonization Promotes the Efficient Uptake and Tuesday, February 28, 2012 | 3:00PM – 4:00PM
Clearance of Burkholderia pseudomallei by Human Neutrophils Diplomat Ballroom
M. E. Woodman, R. G. Worth, R. M. Wooten; Univ. of Toledo Coll. of Med., MODERATOR:
Toledo, OH.
Catharine Basio; NIAID/Rocky Mountain Lab., Hamilton, MT.
212 (I) Priming of Alveolar Macrophages Augments Inflammatory PRESENTATIONS:
Response on Anthrax Lethal Toxin Stimulation
3:00PM
J. Thomas, L. Xiao, J. W. Christman, J. L. Cook, I. Levitan; Univ. of Illinois
Med. Ctr., Chicago, IL. 221 Ornithine Lipids as Therapeutic Targets for Melioidosis
M. Gonzalez-Juarrero1, R. Bowen1, W. Mwangi2, T. M. Eckstein1; 1Colorado
213 (I) A Quantitative PCR-Based Method for the Evaluation of State Univ., Fort Collins, CO, 2Texas A&M Univ., College Station, TX.
Anti-Vaccinia Neutralization Titers
A. D. Garcia, C. A. Meseda, J. Weir; FDA/CBER, Bethesda, MD. 3:15PM
222 Identification of Host Responses Contributing Towards
214 (I) The Chemokine Receptor CCR7 is Expressed in Cells of the
Neuroinvasion and Neurovirulence by Venezuelan Equine
Human Lung in Association with Bacillus anthracis Spore Infection
Encephalitis Virus
M. Langer1, J. Booth1, E. Duggan1, V. Patel1, T. Veres2, F. Prenzler2, K. Sewald2,
P. Gupta1,2, A. Sharma1, M. Bhomia1,2, J. Han3, A. Yang3, R. K. Puri3,
K. Coggeshall3, A. Braun2, J. Metcalf1; 1Univ. of Oklahoma, Oklahoma City,
R. K. Maheshwari1; 1Uniformed Services Univ. of Hlth. Sci., Bethesda, MD,
OK, 2Fraunhofer Inst. for Toxicology and Experimental Med., Hannover, 2
Birla Inst. of Technology and Sci., Pilani, India, 3Food and Drug Admin.,
Germany, 3Oklahoma Med. Res. Fndn., Oklahoma City, OK.
Bethesda, MD.
215 (J) Forensic Entomology and Biodefense 3:30PM
A. Lindström; Natl. Vet. Inst., Uppsala, Sweden.
223 Dynamic Analysis of Respiratory Immune System Provides
216 (J) Rapid Detection of Endospores by Capillary New Insights on Pulmonary Anthrax
Electrophoresis on a Hybrid Channel-to-Digital Microfluidic D. Fiole1,2, A. Quesnel-Hellmann1, J. D. Mathieu1, J. Douady2, J-N. Tournier1;
Platform for Sterilization Validation 1
Inst. de Recherche Biomédicale des Armées, Grenoble, France, 2Univ.
L. Li, W. Wu, W. Chan, H. Cheong, P. Yung; Chinese Univ., Shatin, Hong Kong. Joseph Fourier Grenoble, Saint Martin d’Hères, France.

217 (K) WMD Training Courses at Dugway Proving Ground, 3:45PM


Dugway, UT 224 Visualizing Early Immune Response: Bacterial Specific
P. B. Brown, J. Montague, J. Sorenson; U.S. Army, Dugway Proving Ground, Reorganization at the Nanoscale
Dugway, UT. J. S. Aaron, Q. B. Smith, B. D. Carson, J. A. Timlin; Sandia Natl. Lab.,
Albuquerque, NM.
218 (K)
WITHDRAWN

219 (K) Biological Security: Identifying Vulnerabilities,


Preventing Damage
C. Uhlenhaut; Robert Koch Inst., Berlin, Germany.

220 (K) Transitional Operations: Guiding Owners Through Start-


Up Operations and Regulatory Compliance
M. L. Weiss, D. C. Sharpe; The WorkingBuildings Companies, Atlanta, GA.

BIODEFENSE PROGRAM & ABSTRACTS 27


T U E S DAY S C I E N T I F I C P RO G R A M

Highlighted Oral Abstract Presentations Highlighted Oral Abstract Presentations


020. Therapeutics 021. Diagnostics and Vaccines
Tuesday, February 28, 2012 | 3:00PM – 4:00PM Tuesday, February 28, 2012 | 3:00PM – 4:00PM
Ambassador Ballroom Palladian Ballroom
MODERATOR: MODERATOR:
Colleen B. Jansson; Univ. of Louisville, Louisville, KY. Elisabeth Carniel; Inst. Pasteur, Paris, France.
PRESENTATIONS: PRESENTATIONS:
3:00PM 3:00PM
225 Novel Strategies to Enhance Current Antibiotic Therapies 232 Construction of a Lateral Flow Immunoassay for the
R. V. D’Elia, T. R. Laws, A. Carter, R. A. Lukaszewsk, G. C. Clark; DSTL, Diagnosis of Melioidosis
Salisbury, United Kingdom. D. Reed1, N. Chantratita2, S. J. Peacock3, M. N. Burtnick4, P. J. Brett5, S. Ray-
chaudhuri6, R. Houghton6, T. R. Kozel1, D. P. AuCoin1; 1Univ. of Nevada Sch.
3:10PM of Med., Reno, NV, 2Mahidol Univ., Bangkok, Thailand, 3Univ. of Oxford, Ox-
226 EV-035: A New Compound Series with Broad Spectrum ford, United Kingdom, 4Univ. of South Alabama, Reno, NV, 5Univ. of South
Antibiotic Activity against Burkholderia pseudomallei and Other Alabama, Mobile, AL, 6InBios Intl., Seattle, WA.
Select Agent Pathogens
3:20PM
S. Goldman1, D. Milligan1, H. Yao1, G. Liu1, G. Salerno2, P. Roussel2, J. Heim2;
1
Evolva, Inc., Palo Alto, CA, 2Evolva SA, Reinach, Switzerland. 233 Immunization with Burkholderia pseudomallei Outer
Membrane Vesicles Protects against Pneumonic and Septicemic
3:20PM Melioidosis
227 A Single PMOplus Oligomer is Effective in a Cynomolgus W. Nieves1, B. Judy2, A. Torres2, C. J. Roy3, L. A. Morici1; 1Tulane Univ. Sch. of
Macaque Marburg Virus Lethal Challenge Model Med., New Orleans, LA, 2Univ. of Texas Med. Branch, Galveston, TX, 3Tulane
P. Iversen1, T. Warren2, A. Shrutleff2, J. Wells2, N. Garza2, K. Stuthman2, S. Natl. Primate Res. Ctr., Covington, LA.
Van Tongeren2, L. Dong2, D. Mourich3, L. Hauck1, J. Kugelman2, B. Beitzel2, 3:40PM
G. Palacios2, S. Bavari2; 1AVI BioPharma, Bothell, WV, 2USAMRIID, Fredrick,
MD, 3AVI BioPharma, Bothell, WA. 234 Sortase-Conjugation Generates a Capsule Vaccine that
Protects Guinea Pigs against Bacillus anthracis
3:30PM O. Schneewind, G. Garufi, Y-T. Wang, S-Y. Oh, H. Maier, D. Missiakas; Univ.
228 Therapeutic Efficacy of ST-246 in Cynomolgous Macaques of Chicago, Chicago, IL.
Challenged with Monkeypox Virus via Aerosol
A. T. Russo, K. Agans, T. Brasel, K. Dearen, T. Bailey, S. Storch, R. Baker;
LRRI, Albuquerque, NM.
Symposium Session
3:40PM 022. Host Genetic Resistance to Infections
229 Rationally Designed Broad-Spectrum Antifungals Agents for Tuesday, February 28, 2012 | 4:15 PM - 6:15 PM
the Treatment of Biodefense Pathogens Diplomat Ballroom
E. Garvey, S. Brand, W. Hoekstra, C. Yates, W. Moore, R. Schotzinger; It is now well established that the major factor accounting for the
Viamet Pharmaceuticals, Morisville, NC.
lethality due to infectious diseases is not environmental, but genetic.
This session will provide state-of-the-art information on genetic de-
terminants that predispose individuals to resist to or to develop dif-
ferent types of infectious diseases. In this symposium, an example of
genetic determinisms to infectious disease caused by a parasite, a
fungus, a bacterium and a virus will be presented. In addition, the mo-
lecular and cellular bases for these susceptibility/resistance pheno-
types will be discussed.

MODERATORS:
Simon J. Foote; Macquarie Univ., Macquarie Park, Australia.
Elisabeth Carniel; Inst. Pasteur, Paris, France.

28 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY S C I E N T I F I C P RO G R A M

PRESENTATIONS:
Symposium Session
4:15PM
235 Host Genetic Resistance to Parasitic Infections: The Case of
024. Visual Discovery of Microbe-Host Interactions
Malaria Tuesday, February 28, 2012 | 4:15 PM - 6:15 PM
Simon J. Foote; Macquarie Univ., Macquarie Park, Australia. Palladian Ballroom
4:45PM Pathogen-host interactions are being unraveled by a wide spectrum of
236 Host Genetic Resistance to Mycobacterial Infections: The techniques ranging from high resolution imaging of pathogens, live
Case of Mycobacterium tuberculosis imaging of host response and interactions with specific cellular com-
Philippe Gros; McGill Univ., Montreal, Canada. ponents to whole animal imaging of pathogenesis. Within this vast
array of technical approaches, the direct visualization of pathogens
5:15PM within cells, tissues and whole animals is revealing novel macro-level
237 Host Genetic Resistance to Gram-Negative Bacterial interactions. Especially when combined with real-time imaging and 3-
Infections: The Case of Yersinia pestis dimensional reconstruction, the field is linking pieces of the patho-
Christian E. Demeure; Inst. Pasteur, Paris, France. genesis puzzle together and helping us understand pathogen biology
more thoroughly.
5:45PM
238 Host Genetic Resistance to Viral Infections: The Case of HIV MODERATORS:
Florencia Pereyra; Brigham and Women’s Hosp. and the Ragon Inst. of Colleen B. Jonsson; Univ. of Louisville, Louisville, KY.
MGH, Boston, MA.
Andrew Hayhurst; Texas Biomedical Res. Inst., San Antonio, TX.

PRESENTATIONS:
Symposium Session
4:15PM
023. Dynamic Pathogens Make Difficult Targets 243 Viral Nesting: Rewiring the Host to Generate Organelle
Tuesday, February 28, 2012 | 4:15 PM - 6:15 PM Platforms for Replication
Ambassador Ballroom Nihal Altan-Bonnet; Rutgers Univ., Newark, NJ.

The remarkable adaptability of viral and bacterial pathogens compli- 4:40PM


cates the development of diagnostics, therapeutics, and vaccines. 244 How Vaccinia Virus Manipulates Signaling and the
Considering how pathogens evolve in the face of changing environ- Cytoskeleton to Enhance its Spread
mental pressures, the host response, or drug therapies is a critical Michael Way; London Res. Inst., London, United Kingdom.
perspective. This session will highlight recent insights into the mo-
lecular mechanisms of pathogen evolution and how this informs the 5:05PM
design of novel more durable interventions. 245 Lighting Up Paramyxovirus Pathogenesis: Is Virus Eradication
a Reality, a Redundant Concept, or a Risk?
MODERATORS: Paul Duprex; Boston Univ. Sch. of Med., Boston, MA.
Ted C. Pierson; NIAID, Bethesda, MD.
5:30PM
Jon Yewdell; NIAID, NIH, Bethesda, MD.
246 Insights into Respiratory Melioidosis through Optical
PRESENTATIONS: Diagnostic Imaging
4:15PM Jonathan M. Warawa; Univ. of Louisville, Louisville, KY.

239 Diversity and Evolution of Pathogenic Escherichia coli: 5:55PM


Lessons Learned from High-Throughput Genome Sequencing 247 Computer-Assisted Quantitative Analysis of Infectious
David A. Rasko; Univ. of Maryland Sch. of Med., Baltimore, MD. Disease Imaging
4:45PM Daniel J. Mollura; NIH, Bethesda, MD.

240 Adaptive Arbovirus Landscapes and Implications for Disease


Emergence
Scott C. Weaver; Univ. of Texas Med. Branch, Galveston, TX.

5:15PM
241 On the Molecular Evolution of Influenza A Surface Proteins
Joshua B. Plotkin; Univ. of Pennsylvania, Philadelphia, PA.

5:45PM
242 Burkholderia pseudomallei: A Bacterial Pathogen with a
Highly Adaptable Genome
Paul Keim; Northern Arizona Univ., Flagstaff, AZ.

BIODEFENSE PROGRAM & ABSTRACTS 29


T U E S DAY S C I E N T I F I C P RO G R A M

Roundtable Discussion
025. Global Status of Strategies for Addressing
Intersection of Science and Security
Tuesday, February 28, 2012 | 6:30 PM - 7:30 PM
Palladian Ballroom
Life sciences research is a vital social undertaking that yields innu-
merable and immeasurably important benefits. At the same time, we
must also recognize that good science can be put to bad uses, and
that even a single misuse of certain information, knowledge, or tech-
nology could have devastating and far-reaching effects. Over the past
ten years, there has been a growing recognition that some information
resulting from life sciences research, in the wrong hands, can be mis-
used to pose a threat to public health and national security. Research
yielding new technologies or information with the potential for both
benevolent and malevolent applications is referred to as “dual use re-
search.” Because the life sciences are a global endeavor, it is impor-
tant to work together to develop effective approaches to minimizing
the risk of misuse, while still fostering the continued progress of the
science that underpins public health and safety. A critical step is en-
gaging microbiologists, who may not be aware of the issue. This ses-
sion provides an opportunity to learn about the dual use issue and
provides a forum for sharing international perspectives and activities
relating to dual use life sciences research. The presenters and partic-
ipants in this session will explore strategies for managing the over-
sight of dual use life sciences research, as well as strategies for
fostering international awareness and engagement on the issue. A
major focus of the session will be on lessons learned from the imple-
mentation of strategies in various countries as well as to hear from
those countries with strategies under development.

MODERATORS:
Stuart Levy; Tufts Univ. Sch. of Med., Boston, MA.
Paul Keim; Northern Arizona Univ., Flagstaff, AZ.
PANELISTS:
248 Stuart Levy; Tufts Univ. Sch. of Med., Boston, MA.
249 Koos Van der bruggen; Independent Researcher and Science Writer,
The Hague, Netherlands.
250 Catherine Jefferson; The Royal Society, London, United Kingdom.
251 Gerald L. Epstein; Biosecurity Analyst, Washington, DC.

30 10th ASM Biodefense and Emerging Diseases Research Meeting


W E D N E S DAY S C I E N T I F I C P RO G R A M

Wednesday, February 29, 2012 Plenary Session


027. Immunity to Pulmonary Pathogens
Research Discussion Wednesday, February 29, 2012 | 8:30AM - 12:00PM
026. H5N1 Regency Ballroom
Wednesday, February 29, 2012 | 7:15 AM – 8:15AM The lung is a dynamic and relatively immunosuppressed organ. Fur-
Palladian Ballroom thermore, immune responses in the lung are often dissimilar to those
that occur following systemic infection. Many pathogens successfully
This newly added session at the ASM Biodefense and Emerging Dis-
use these features of the pulmonary compartment to their advantage
eases Research Meeting will include discussions of the NSABB’s rec-
in establishing disease. Thus, understanding how pathogens take ad-
ommendations for the publication of the controversial H5N1 research.
vantage of the lung environment will increase our ability to design
This session will also provide an open forum for attendees to give
novel therapeutics and vaccines that target this important tissue.
their feedback on such policy issues as the appropriate mechanisms
to allay public concerns about the safety of dual use research. MODERATORS:
Catharine M. Bosio; NIAID/Rocky Mountain Lab., Hamilton, MT.
MODERATOR:
Paul Keim; National Science Advisory Board for Biosecurity Chair, Daniel Voth; Univ. of Arkansas for Med. Sci., Little Rock, AR.
Bethesda, MD. PRESENTATIONS:

PRESENTATIONS: 8:30AM
NSABB Recommendations 252 The Host Immune Response to Pulmonary Virus Infection: It’s
About Self Control
Michael T. Osterholm, PhD., MPH; University of Minnesota School of Public
Health, Minneapolis, MN; Director, Center for Infectious Disease Research Thomas J. Braciale; Univ. of Virginia, Charlottesville, VA.
and Policy (CIDRAP). 9:15AM
Government Response to the Recommendations 253 Requirements for Pulmonary Immunity against Virulent
Anthony S. Fauci, MD; Director, National Institute of Allergy and Infectious Francisella tularensis
Diseases (NIAID). Catharine Bosio; NIAID/Rocky Mountain Lab., Hamilton, MT.

Science’s Response to the Situation 10:00AM


Bruce Alberts, PhD; Editor-in-Chief of Science. Coffee Break
Perspective from a Scientist 10:30AM
Ron A.M. Fouchier, PhD.; Elasmus Med. Ctr., Rotterdam, The Netherlands. 254 Infection of Human Alveolar Macrophages by the Q Fever
Agent, Coxiella burnetii
Daniel Voth; Univ. of Arkansas for Med. Sci., Little Rock, AR.

11:15AM
255 The Protective Role of Surfactant Protein-A During
Mycoplasma pneumoniae Infection in Allergic and Non-Allergic
Airways
Julie G. Ledford; Duke Univ., Durham, NC.

BIODEFENSE PROGRAM & ABSTRACTS 31


NOTE S

32 10th ASM Biodefense and Emerging Diseases Research Meeting


ABSTRACTS

majority of all respiratory infections were male (n=116) and age ≤ 4 (n=111).
Poster Session Amongst those cases associated with mortality (n=11), 7 were male and 7
008. Monday Posters were age ≤ 4. Conclusions: This laboratory surveillance data emphasized the
temporal impact of non-influenza respiratory viruses during the 2009 H1N1
Monday, February 27, 2012 | 1:00 PM - 3:00 PM Influenza Pandemic. Characterizing disease severity, notably mortality, aids
Exhibit Hall in discerning the potential consequences of a pandemic. As the foundation
of respiratory viral surveillance, state laboratories describe the comprehen-
027 (A) sive viral etiology to rapidly implement the necessary control and preventive
measures in case of disease outbreaks and pandemics.
Viral Acute Respiratory Tract Infections in Non-Winter Time
M. EL-Barrawy; Alexandria Univ., Alexandria, Egypt.
Introduction: Acute respiratory tract infection (ARTI) is the most frequent ill- 029 (A)
ness globally and a leading cause of death in the developing world. The most Natural History Studies for Ebola Virus Zaire Strain in Hartley
common cause of an ARTI may be one of over 200 viruses. However, bacte-
Guinea Pigs
ria or fungi can be its cause as well. Although viral respiratory infections are
A. Nalca, H. Bloomfield, S. Frederick, R. Baker, R. Williams, J. Yeager, E. Zum-
relatively considered harmless, in developing countries they are an impor-
brun; USAMRIID, Frederick, MD.
tant cause of mortality, especially when followed by bacterial complications.
Background: Ebola virus (EBOV) is a negative-stranded, enveloped RNA virus
The viruses which are mostly implicated are: adenoviruses, influenza viruses,
and a member of the Filoviridae family. It causes severe viral hemorrhagic
RSV and HSV. Although Respiratory viral infection is commonly peaked in
fevers with possible case-fatality rates of more than 90% depending on the
winter, it still has its heavy impact upon human health rest of the year. Aim
virus and virus strain. The fear of a bioterrorism event involving filoviruses
of the work: was to determine the prevalence of the previous respiratory
has increased the efforts of the scientific community in the development of
viruses in non-winter period among patients with ARTIs. Subjects & Meth-
countermeasures against these viruses. Currently, there are no licensed med-
ods: This study was carried out through the period from March 2010 to No-
ical countermeasures against filoviruses. Therefore, it is important to have a
vember 2010. It included 477 patients attending Alexandria Fever Hospital
small animal model for these viruses especially for aerosol route of exposure
and showing ARTI symptoms. All patients were recruited after having their
which could be highly infectious. Methods: In this study, we exposed two
consents and proper history taking. Throat swabs were collected, transferred
groups of 10 naïve Hartley guinea pigs to target doses of 10 PFU (low dose)
to Naval American Medical Research Unit 3(NAMRU 3) in Cairo, and tested
and 1000 PFU (high dose) of aerosolized guinea pig-adapted Ebola virus Zaire
using PCR techniques. Results: Out of 477 patients, the tested viruses were
strain (ZEBOV). Blood was collected on day -7 and then on days 3, 6, and 9
detected in 158 (33.1%) patients. Single virus infection was detected in
PE to test complete blood count (CBC) and serum chemistries. Results:
144(30.2%). This was as follows: 63(13.2%) influenza-A virus, 54(11.3%)
Guinea pigs started to show clinical signs of the disease including weakness,
adenovirus, 26(5.5%) RSV and one (0.2%) HSV. Meanwhile, 14(2.9%) had
fever, anorexia, and weight loss on day 4 post exposure (PE). While the high-
combined viral infection: 7(1.5%) had a combination of adenovirus and RSV,
dose exposure group started to show increased body temperature on day 4
6(1.3%) had a influenza A virus and adenovirus, and one patient (0.2%) had
PE, the low-dose exposure group did not have increased body temperature
influenza A virus, adenovirus and RSV. Conclusion: Viral infections represent
until day 5 PE. Interestingly, as the guinea pigs white blood cells (WBC), lym-
a substantial cause of ARTIs , even in non-winter time. Influenza A virus was
phocytes, and platelets decreased, their monocytes, basophils and
the most prevalent respiratory virus (14.7%). The viral ARTIs were more
eosinophils increased throughout the study. Increases in liver enzymes such
prevalent in children <15 years. There was no association between the res-
as alkaline phosphatase, alanine transferase, and aspartate transferase, as
piratory viruses’ infections and: smoking, occupation, temperature, and/ or
well as total bilirubin, were observed on day 6 PE. Furthermore, blood urea
existence of any chronic disease.
nitrogen (BUN) and creatinine levels were high in moribund animals. Low-
dose and high-dose exposure groups succumbed to disease on average at
028 (A) days 9.0 ± 0.48 and 8.3 ±1.0 PE, respectively. Conclusion: These data very
closely reflect observations of ZEBOV-infected non-human primates. There-
Laboratory Surveillance for Non-Influenza Respiratory Viruses fore, the guinea pig model for aerosolized ZEBOV is a useful tool to screen
in Maryland medical countermeasures against filoviruses.
W. A. Murtaugh1,2, J. Tan1, H. Peters2, B. Healey2, M. Tran2, M. Ziese2, B. An-
derson2, M. P. Carlos1,2; 1Johns Hopkins Univ., Baltimore, MD, 2Lab. Admin.
Maryland Dept. of Hlth. and Mental Hygiene, Baltimore, MD. 030 (A)
Background: State laboratories routinely identify the viral causes of respira- Experimental Respiratory Marburg Angola Virus Infection in
tory diseases. We provide a summary of non-influenza respiratory viral test-
Hartley Guinea Pigs
ing from the Maryland State Laboratory during the period of the 2009 H1N1
A. Nalca, H. Bloomfield, S. Frederick, R. Baker, R. Williams, J. Yeager, E. Zum-
Influenza Pandemic, and describe the implications of continual monitoring
brun; USAMRIID, Frederick, MD.
of circulating respiratory viral strains. Methods: Between 2008 and 2011,
Background: Marburg virus (MARV) is a causative agent of severe viral hem-
5014 influenza virus negative respiratory specimens were tested for respira-
orrhagic fever and classified as a category A biological agent due to its po-
tory syncytial virus (RSV), adenovirus (AdV), and parainfluenza virus (PIV) 1-
tential use as a bioweapon. The objective of this study to develop small
3 by cell-culture. For three overlapping influenza periods (2008-2009
animal model and characterize natural history of aerosolized Marburg virus
influenza season, 2009 H1N1 Pandemic Influenza, 2010-2011 influenza sea-
infection in Hartley guinea pigs (GPs). Methods: Two groups of 10 GPs were
son), the number of specimens tested, virus identified, and positivity rates
challenged by aerosol route with target doses of 10 (low) and 1000 (high)
were summarized. We performed an analysis of the positive specimens strat-
PFU of GP-adapted Marburg Virus Angola strain on day 0. . Blood was col-
ified by respiratory disease severity (out-patient, hospitalized, deceased),
lected on day -7 and then on days 3, 6, 9, 12, 15 and 18 post exposure (PE)
age and sex. Results: A total of 199 respiratory viruses were detected: RSV
to test complete blood count (CBC) and serum chemistries. Results: It was de-
(56), AdV (66), PIV-1 (30), PIV-2 (21), PIV-3 (26). The testing volume peaked
termined that average presented doses were 55 PFU for low-target dose and
in October 2009 (n=613). RSV and PIV displayed their common seasonal pat-
4400 PFU for high-target dose. Although they were sick and showed the signs
terns in the Northern Hemisphere, while AdV was detected intermittently. The

BIODEFENSE PROGRAM & ABSTRACTS 33


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

of the disease, five GPs from low-dose group survived the challenge with on October 13th while school attendance dropped on October 9th, reaching
aerosolized GP-adapted Marburg virus and all high-dose exposed group GPs a nadir on October 16th. ILI reporting peaked during the same week although
except one succumbed to disease by day 12 PE. Last GP from high-dose data was unavailable until the following Monday (Figure). Although all
group was moribund and euthanized on day 17 PE. Guinea pigs started to datasets showed similar activity, school absenteeism reports were available
show clinical signs of the disease including weakness, fever, anorexia, and on a “same day” basis, whereas reported ILI and rapid antigen test results
weight loss on day 6 PE\. While the high-dose exposure group started to were collated weekly, delaying availability. During the 2009 H1N1 outbreak,
show increased body temperature on day 5 PE, the low-dose exposure group school absenteeism and reported ILI were temporally related to ED patient
did not have increased body temperature until day 6 PE. The white blood cells visit volume. Health officials may be able to monitor influenza activity by fol-
(WBC), lymphocytes, and platelets decreased throughout the study. Except lowing school attendance and predict strain on healthcare resources.
survivors, increases in liver enzymes such as alkaline phosphatase, alanine
transferase, and aspartate transferase, as well as total bilirubin, were ob-
served on day 9 PE. Conclusion: These results suggest that Hartley GPs are
susceptible to aerosolized MARV therefore this animal model might be a use-
ful tool to screen medical countermeasures against filoviruses.

031 (A)
A Cytopathic Effect-Based High-Throughput Screening Assay
Identified Four Novel Compounds that Inhibit Dengue Infection
K. D. McCormick; Univ. of Pittsburgh, Pittsburgh, PA.
To date, no approved vaccine or antiviral therapy exists for dengue virus
(DENV). Based on the observation that DENV infection of hepatocytes in-
duces massive cell death, “cytopathic effect (CPE),” we developed a high-
throughput screening assay(HTS) whereby inhibitors of DENV prevent cells
from dying. In this assay, the viral induced CPE is quantitated using a lu-
ciferase system for monitoring cellular ATP levels, which positively correlates
with cellular viability. This assay simultaneously screens possible inhibitory
compounds while controlling for any cytotoxicity triggered by these com- 033 (A)
pounds. Moreover, it is highly reproducible yielding a screening window co-
efficient, Z-factor, of 0.78±0.12. Our initial screening of a 288 NCI The Role of Rare Variants in Coronavirus Cross-Species Trans-
small-compound library yielded a total of eleven hits that prevented the CPE mission Events
of DENV infection. Further evaluation with an immunofluorescence assay M. Borucki, J. Allen, H. Harris-Chen; Lawrence Livermore Natl. Lab, Livermore,
showed that four of these compounds, streptovitacin A, nagilactone C, bac- CA.
tobolin and trichodermin, are highly potent inhibitors of DENV infection. At ef- A vast majority of the recently discovered human pathogens are viruses that
fective inhibitory doses, they did not appear to be cytotoxic, and they have an animal reservoir. The genetic diversity of a viral population within a
inhibited all four DENV serotypes albeit with varied efficacies. Interestingly, host may allow the virus to adapt to a diverse array of selective pressures
these compounds displayed strong inhibitory effects on hepatitis c virus (also and enable cross-species transmission events. Understanding the role of ge-
belongs to Flaviviridae family) infection, but exerted no or only marginal ef- netic variants within a viral population is a necessary step toward predicting
fect on new castle diseases virus. Altogether, our data show that these com- and treating emerging infectious diseases. Deep genome sequencing was
pounds are possible DENV antivirals. combined with computational analysis to characterize genomic changes in
coronavirus quasispecies during simulated cross-species transmission
events. Three bovine nasal samples naturally infected with bovine coron-
032 (A) avirus were used to infect human and bovine macrophage and lung cell lines.
Virus present in each sample was able to reproduce relatively well in
Predicting the Timing of Peak Pandemic Influenza Activity: ED
macrophages but not in the lung cell lines. Approximately 12 kb of the
Volume, School Absenteeism, and Community Reported ILI
genome was amplified before and after passage and sequenced. The con-
T. Bell1, M. Richardson2; 1West Texas Influenza Ctr., TTUHSC, Amarillo, TX,
2 sensus sequence of viruses passaged in bovine macrophages changed more
Amarillo Publ. Hlth. Dept., Amarillo, TX.
rapidly than those passaged in human cell lines, and the consensus se-
Although influenza is usually a self-limited illness, the disease burden and ra-
quences of spike gene varied the most of any region tested. Many of the pas-
pidity of spread during a pandemic can strain healthcare resources. Rapid
saged samples had a 12 nucleotide insert in the consensus sequence of the
escalation of cases makes monitoring difficult. Monitoring systems should
spike gene, and multiple point mutations were associated with the presence
be 1) available in “real time” 2) utilize existing data or require minimal addi-
of the insert. Deep sequencing revealed that the insert was present but very
tional work and 3) reflect local disease activity. In 2009, a novel influenza
rare in the unpassaged samples but could quickly shift to dominate the pop-
pandemic occurred. Most cases were self-limited, and the greatest strain on
ulation when placed in a different environment. Analysis of the subconsen-
healthcare resources in our community was in Emergency Departments (ED).
sus (quasispecies) data indicated that two distinct genotypes circulated at
As part of our public health response, we asked providers to voluntarily re-
different frequency levels in each sample, and support the hypothesis that
port influenza-like-illness (ILI) to the public health department (PHD). We
the mutations present in passaged strains were “selected” from a pre-exist-
also monitored school absenteeism and influenza testing results from local
ing pool rather than through de novo mutation and subsequent population
providers. We compared these data sets to ED volume to determine which
fixation.
best predicts peak influenza activity. The PHD monitored school attendance
on a daily basis. ILI visits to physicians were reported on a voluntary basis
from sentinel sites. Community providers provided information on influenza
test results. We obtained a daily ED census from hospitals. ED volume peaked

34 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

lution of plasmids and present a model for DNA swapping between plasmid
034 (B) molecules mediated by successive SSR events at oriT and the Xer RS. The
Transient Bacterial Lipopolysaccharide-Induced Resistance to first event involves SSR at oriT, which depends exclusively on oriT site and the
Inhalational Bacillus anthracis Infection in the New Zealand nickase activity, and leads to integration. The cointegrate includes two di-
White Rabbit Model rectly positioned non-identical Xer RSs that may serve as substrate for a sec-
S. B. Yee, D. N. Dyer, N. A. Twenhafel, M. L. M. Pitt; USAMRIID, Fort Detrick, ond SSR event mediated by Xer that leads to cointegrate resolution. The final
MD. products are the original plasmids but they have exchanged the fragments
Recent studies have demonstrated prior bacterial infection induces resist- flanked by oriT and Xer. We propose that the combination oriT-Xer RS could
ance to subsequent infections from gram-negative and gram-positive be considered an element that facilitates plasmid evolution by swaping DNA
pathogens. In the present study, we evaluated whether underlying inflam- regions.
mation would enhance host resistance to inhalational Bacillus anthracis in-
fection in New Zealand white (NZW) rabbits. Accordingly, NZW rabbits
(specific pathogen-free, Bordetella-free) were either pretreated with the in-
036 (B)
flammagen, bacterial lipopolysaccharide (LPS), a component of the outer Antibacterial Activity of Alpha-MSH Alone and in Combination
membrane of gram-negative bacteria, or saline vehicle (VEH). Administration with Other Antibiotics against Methicillin-Resistant Staphylo-
of this noninjurious dose of LPS (60,000 EU/kg) resulted in brief pyrexia and coccuus aureus
a significant increase in the pro-inflammatory cytokine tumor necrosis fac- M. Singh, K. Mukhopadhyay; Sch. of Environmental Sci., Jawaharlal Nehru
tor-alpha confirming LPS-induced inflammation. Approximately 24 h later, Univ., New Delhi, India.
rabbits were exposed to aerosolized B. anthracis spores (Ames strain; ~300 Background: We recently demonstrated that melanocortin peptide alpha-
LD50). Blood samples collected at various time points postchallenge were Melanocyte Stimulating Hormone (α-MSH), possesses antimicrobial activity
cultured for bacteremia. LPS-pretreated, B. anthracis-exposed rabbits com- against Staphylococcus aureus (S. aureus) and causes bacterial membrane
pared to VEH-pretreated, B. anthracis-exposed rabbits exhibited significantly permeabilization. The present study was performed to investigate the in vitro
delayed biomarkers of B. anthracis infection, anthrax-induced pyrexia (66 h antibacterial potential of α-MSH against the methicillin resistant S. aureus
vs. 25 h postchallenge, respectively) and bacteremia (63 h vs. 26 h), and sur- (MRSA) and synergy between α-MSH and four other antibiotics (oxacillin,
vived significantly longer (90 h vs. 41 h). As with the control animals, all LPS- ciprofloxacin, gentamicin and rifampicin) against MRSA strain resistant to all
pretreated, B. anthracis-exposed rabbits exhibited pathology consistent with selected antibiotics and α-MSH. The study had been extended to examine α-
succumbing to inhalational anthrax. No differences in histologic lesions — MSH induced morphological and ultra structural changes in MRSA and he-
including acute necrotizing splenitis, lymphadenitis of the mediastinal lymph molytic effect of α-MSH was also evaluated. Method: An ATCC MRSA and a
node, mediastinitis, pneumonia, adenitis of the adrenal gland, and renal tu- multidrug resistant S. aureus were used in this study. MIC of selected antibi-
bular necrosis—were observed between the two groups. Taken together, otics was determined using broth microdilution method. The staphylocidal
these results suggest that prior or underlying stimulation of the immune sys- activities of α-MSH (10-6 to 10-9M) was assessed by quantitative culture
tem induces transient host resistance to and/or delayed B. anthracis infection after 2h bacterial incubation with α-MSH and compared to peptide free con-
in NZW rabbits. In particular, these results emphasize the importance of using trols. Synergism was checked using the above staphylocidal assay for differ-
animals free of underlying infections to prevent confounding data in inhala- ent antibiotics alone and in combination with α-MSH. Scanning and
tional anthrax characterization and therapeutic studies. transmission electron microscopy was employed to visualize the morpho-
logical and ultrastructural changes. The hemolytic effect was evaluated by
determining hemoglobin release of fresh rat erythrocyte suspension treated
035 (B) with α-MSH at 414nm. Results: α-MSH killed 80.57%±2.01 and 49±1.08%
Small Quinolone Resistance Plasmids: A Model for Evolution ATCC MRSA strain in micromolar and nanomolar concentration respectively.
Mediated by Site-Specific Recombination (SSR) at oriT and Xer 80%±1.2 (p <0.05) increase in killing was observed in case of combination
Sites of ciprofloxacin and α-MSH in comparison of either of agent alone. Similar
T. Tran1,2, P. Andres3, A. Petroni3, A. Soler-Bistué4, E. Albornoz3, A. Zorregui- observations were also obtained for oxacillin and gentamicin. SEM and TEM
eta4, R. Reyes-Lamothe2, D. Sherratt2, A. Corso3, M. Tolmasky1,2; 1California images had shown remarkable alteration in bacterial cell envelope. <10%
State Univ., Fullerton, CA, 2Univ. of Oxford, Oxford, United Kingdom, 3INEI- hemolytic effect was obtained in case of 10-5M of α-MSH. Conclusion: α-
ANLIS, Buenos Aires, Argentina, 4Fundación Inst. Leloir, Buenos Aires, Ar- MSH is strongly active against the MRSA and its synergistic action with other
gentina. antibiotics is indeed encouraging and provides a hope for MRSA infection
Qnr are proteins that mediate resistance to quinolones by protecting DNA treatment.
gyrase. The qnrB19 gene was found in several Enterobacteriaceae isolated
from numerous geographical regions within large plasmids, in ISEcp1C -
based transposons, and in small ColE1-type plasmids (about 3 kbp) lacking
037 (B)
ISEcp1C or any other insertion sequence. In spite of being located in such Enhanced Aminoglycoside Sensitivity of Ribonuclease Mutants
dissimilar elements, the qnrB19 genes share a conserved genetic environ- of Escherichia coli
ment. We found 4 small plasmids harboring qnrB19, pPAB19-1, pPAB19-2, A. D. Frazier, W. S. Champney; East Tennessee State Univ., Johnson City, TN.
pPAB19-3 and pPAB19-4 in a collection of clinical enterobacteria with reduced Background: An important issue for global health is the emergence of bac-
quinolone susceptibility. Nucleotide sequencing and analysis showed that terial resistance to many current antibiotics. This raises the possibility of
they share extensive homology among themselves and with other described using antibiotic resistance for bioterrorism. Consequently, new cellular tar-
small qnrB19-harboring plasmids. The genetic environment of qnrB19 in all gets for the existing antibiotics are needed. Ribosome biosynthesis is a novel
four plasmids is identical to that in those other plasmids and in transposons antibiotic target in bacterial cells and ribonucleases are essential to the pro-
such as Tn2012, Tn5387, and Tn5387-like. These plasmids as well as those cessing and turnover of bacterial rRNA. Previous studies have shown E. coli
previously described showed a variable region characterized by being flanked strains deficient for specific RNases displayed an enhanced sensitivity to
by an oriT locus and a Xer recombination site (RS). Both loci were confirmed azithromycin. It was hypothesized the inhibition of specific RNases in bacte-
to be functional. We propose that this arrangement could play a role in evo- rial cells would lead to an increased sensitivity to aminoglycoside antibiotics.

BIODEFENSE PROGRAM & ABSTRACTS 35


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

Methods: Ten RNase deficient E. coli strains were grown in broth in the pres-
ence or absence of 5μg/mL of neomycin or paromomycin. Viable cell counts
039 (B)
were determined after two cell doublings. The synthesis of 30S and 50S ri- Genes Required for the Trafficking of S-Layer Proteins in the
bosomal subunits in aminoglycoside treated cells was determined by sucrose Envelope of Bacillus anthracis
gradient centrifugation of 3H uridine labeled cell lysates. RNA from these S-M. Nguyen-Mau1,2, D. Missiakas1,2, O. Schneewind1,2; 1Univ. of Chicago,
cells was analyzed by Agilent chip analysis and Northern blotting. Results: Chicago, IL, 2Howard Taylor Ricketts Lab., Argonne, IL.
Mutant strains deficient for RNase E, G, or PH were found to display an in- Many Gram-positive microbes, including Bacillus anthracis— the causative
creased sensitivity to the inhibitory effects of neomycin and paromomycin. In agent of anthrax—assemble proteins into a crystalline lattice on their sur-
each of these mutant strains, cellular viability was significantly reduced in face. S-layer proteins contribute to the envelope assembly of B. anthracis S-
the presence of the aminoglycosides. In the RNase PH deficient cells, growth layer associated proteins (BSLs), which play important roles during the
with the antibiotics present reduced the synthesis of the 30S and 50S sub- pathogenesis of bacterial infections and are known to bind to host cell re-
units by more than 30%. Agilent chip analysis revealed an accumulation of ceptors or scavenge heme-iron from host hemoproteins. Precursors of S-layer
the degradation products of rRNA for each mutant strain. Northern blotting proteins and BSLs harbor N-terminal signal peptides followed by three S-
analysis showed a large increase in the breakdown of 16S and 23S ribosomal layer homology (SLH) domains. In mature polypeptides, the SLH domains are
RNA in strains deficient for RNase E, G, or PH in comparison with a wild type responsible for the non-covalent association of these proteins with pyruvy-
control. Conclusion: This work showed that E. coli mutants lacking RNases E, lated secondary cell wall carbohydrate (SCWP), thereby enabling their S-layer
G, or PH, revealed an enhanced sensitivity to aminoglycoside antibiotics. This deposition on the bacterial surface. Comparably little is known about the
work also suggests the inhibition of essential RNases may be a new drug tar- genes and molecular mechanisms whereby S-layer proteins BSLs are se-
get for small molecules or RNAi sequences. creted and traffic to their site of envelope assembly. Here we report that the
efficient deposition of S-layer proteins depends on a homolog of the secA se-
cretion gene, designated secA2, and a gene encoding acytoplasmic protein,
038 (B) designated slaP (S-layer assembly protein). Mutations in secA2 or slaP re-
A Model of Inhalation Glanders in African Green Monkeys duce the secretion of S-layer proteins Sap and EA1, without affecting the se-
J. Yeager, P. Facemire, D. Nyakiti, L. Pitt; USAMRIID, Frederick, MD. cretion of two S-layer associated proteins, BslA and BslO, or of protective
Burkholderia mallei, the causative agent of glanders, has been nearly eradi- antigen, a secreted polypeptide. SlaP associates with the Sap and EA1 pre-
cated in industrialized nations, but recently has had renewed interest due to cursors in the cytoplasm of B. anthracis, suggesting that it may function as a
The Center for Disease Control classification of it as a Category B select agent. chaperone for S-layer protein secretion via SecA2.
There is a lack of modern clinical cases resulting in limited knowledge of sus-
ceptibility of B. mallei to antibiotics and of its mechanism of virulence. There-
fore an animal model(s) that represent human glanders is needed to evaluate
040 (B)
the natural history and evaluation medical countermeasures. The goal of this Development of a Selective Enrichment Media for Brucella
study was to evaluate African green monkeys (AGM) effectiveness as a model Species Isolation
for glanders. Ten AGMs were challenges to 147±90 LD50 of aerosolized B. M. Perry, A. Kidney, M. Peck, C. Kelly-Cirino, C. Egan; NYS Dept. of Hlth., Al-
mallei (ATCC 23344). Telemetry acquired temperature was collected hourly bany, NY.
and clinical symptoms were monitored daily for 45 days. Each AGM had com- Brucellosis is a zoonotic disease transmittable to humans usually through
plete blood counts, bacteremia and cytokines measured daily for the first direct contact with infected animal tissues/fluids or by the consumption of
seven days post challenge (p.c) and then reduced to weekly. Gross pathology contaminated, unpasteurized dairy products and meat. Due to the fastidious
and histopathology analysis were performed on all animals. A total of 7/10 nature of Brucella and high background levels of competing organisms, con-
AGMs succumbed to glanders between 6 and 32 days p.c. Days 1 to 3 post ventional microbiological methods can take days to weeks for isolation and
challenge a fever (≥38.5°C) or increase in the diurnal temperature was ob- confirmation. To prevent the growth of competing organisms, isolation usu-
served in 10/10 AGMs. On day 2 p.c. 10/10 AGMs had an increase in leuko- ally requires selective media. Current diagnostic isolation recommends Far-
cytes with an increased percentage of neutrophils. Bacteremia was detected rell’s medium (FM) and Modified Thayer-Martin medium (mTM), however they
in 5/10 AGMs ranging from 5 to 45 days p.c. Lesions and nasal discharge are non-selective and can inhibit the growth of several Brucella species. In
were common and typically manifested in the presence of a sustained fever order to facilitate isolation, the Wadsworth Center (WC) has developed a se-
rather than a shift in diurnal temperature. Common gross lesions included lective enrichment media for solid plate agar and broth cultures incorporat-
abscesses in the lungs and skin. Histologically, the abscesses were composed ing both selectivity as well as color identification of Brucella species. The
of necrotizing to necrosuppurative inflammation. Additionally, this inflam- Wadsworth Brucella Selective Media (WBSM) was compared to mTM, FM,
mation was often identified in the spleen, bone marrow and lymph nodes. CITA, Brucella agar (BruA) and 5% sheep blood agar plates (SBAP) for speci-
The three surviving AGMs developed limb weakness to partial paralysis. Here ficity and selectivity. All Brucella spp. exhibited growth on the WBSM, how-
we describe a model of inhalation glanders in AGMs. Much of the clinical in- ever both Brucella canis and neotome colonies appeared smaller on WBSM
dicators of infection are those that have been described previously in human compared to CITA. In addition, growth was not optimal for Brucella abortus,
glanders. This data suggests that AGMs are a reasonable model for studying strain RB51, on either media but the concentration was 225 CFU/100 μL with
glanders, making them valuable in therapeutic treatment studies and future SBAP. CITA and WBSM outperformed all other media when Brucella was
vaccine studies. spiked into food matrices. An added benefit of WBSM is that it incorporates
neutral red with erythritol, creating easily identifiable pinkish colonies. Fur-
thermore, growth of other organisms including E. coli and Salmonella were in-
hibited when plated on the WBSM, demonstrating its selectivity. The
capability of WBSM can be increased when combined with the WC enrich-
ment broth (mF). After overnight enrichment of Brucella in mF, there was a
10 fold increase from 18 to 310 CFU/100 μL in a 103 CFU/mL sample of feta
cheese. A similar increase post enrichment was seen for other matrices, in-
cluding infant formula. This study demonstrates the ability of a newly devel-

36 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

oped media and enrichment, WBSM and mF, to selectively identify Brucella
spp. and allow for more rapid patient treatment.
043 (B)
Burkholderia thailandensis is a Better Surrogate for B. pseudo-
mallei Growth and Persistence than B. pseudomallei Bp82
041 (B) M. A. Firmani, S. Lehman, S. Dieringer-Boyer, K. Bower, B. Dorsey, D. Stratton,
Periplasmic YopR Promotes Type III Needle Polymerization K. Delp, D. K. R. Karaolis; Natl. Biodefense Analysis and Countermeasures
B. Blaylock, O. Schneewind; Univ. of Chicago, Chicago, IL. Ctr., Frederick, MD.
Yersinia pestis, the causative agent of plague, depends absolutely on type Burkholderia pseudomallei is a Category B select agent. B. thailandensis is
III secretion to cause disease. An essential component of the type III secre- closely related and coexists in the same environment. However, B. thailan-
tion machine is a hollow filament that extends from the outer membrane, the densis is not a human pathogen and is a commonly used surrogate for B.
“needle.” The proteins that regulate and assemble needle polymerization are pseudomallei studies. B. pseudomallei Bp82 is a recently developed B.
the earliest substrates of the type III pathway. In Yersinia species, YopR con- pseudomallei 1026b purM mutant that is avirulent and has been exempted
tributes to needle polymerization by a mechanism that is under investiga- as a select agent. In this study, we performed growth kinetics and evaluated
tion. Mutations in yopR result in the abundant secretion of the needle shaft the persistence of both organisms grown in three media conditions. Survival
protein, YscF, but abolish the assembly of this structure. Complementation of over time at four temperatures was evaluated, including survival of log vs
a yopR mutation with alleles that are tagged at the protein’s carboxy termi- stationary cells. In addition, phenotypic comparisons (i.e., antibiotic sus-
nus are secreted but non-functional. YopR variants where the tag is at the ceptibilities, biofilm formation, motility, and protease production) were per-
amino terminus would be expected to obscure the secretion signal; surpris- formed. Our results demonstrate that B. thailandensis biofilm production in
ingly, these alleles are secreted and functional. Sub-cellular fractionation re- stationary phase cells was significantly (P<0.05) higher than log cells. In con-
vealed that only functional YopR alleles were present in the periplasm. trast, Bp82 did not produce a biofilm in log or stationary phase. A key find-
Artificial delivery of YopR to the periplasm by means of the Sec translocation- ing was that Bp82 is not able to grow well in certain media. Using LB-Lennox
dependent PhoA signal sequence also resulted in the restoration of needle media, we were able to perform growth and persistence assays with Bp82. In-
polymerization and was surprisingly re-engaged by the type III pathway. Col- terestingly, persistence studies showed log phase Bp82 cells survive better
lectively, these data suggest a role for YopR at the earliest stages of type III than stationary phase cells, which is in contrast to B. thailandensis. Pheno-
machine assembly at a time when it is still open to the periplasmic contents. typic assays with Bp82 were difficult to perform due to the mutant’s strin-
gent growth requirements. Bp82 would not generate a sufficient lawn of
growth on Mueller Hinton agar for antimicrobial susceptibility testing nor
042 (B) would it grow on skim milk agar for protease assay production. Our results
Virulence Analysis of B. cereus Biovar Anthracis demonstrate that Bp82 has a growth deficiency and that B. thailandensis
S. Dupke1, T. Franz1, A. Lander1, R. Grunow1, P. L. Goossens2, C. Brezillon2, possesses characteristics which are more similar to B. pseudomallei. Conse-
S. R. Klee1; 1Robert Koch Inst., Berlin, Germany, 2Inst. Pasteur, Paris, France. quently, B. thailandensis may be a better B. pseudomallei surrogate than
During the last years, several great apes died of an anthrax-like disease in Bp82 for growth and persistence studies.
West- and Central Africa. Molecular analyses and genome sequencing re-
vealed a close relationship of the new strains to B. cereus rather than B. an-
thracis on chromosomal level, even though both characteristic virulence
044 (B)
plasmids pXO1 and pXO2 are present. Therefore the African isolates were WITHDRAWN
designated as B. cereus biovar anthracis. Virulence analysis showed that all
isolates tested are able to produce the pXO1-encoded toxin components as
well as the pXO2-encoded polyglutamic acid capsule. Also in animal experi-
ments with mice and guinea pigs, the LD50 was similar to wildtype strains of
B. anthracis. Furthermore, comparable regulation of virulence factors was
confirmed for B. cereus bv anthracis and B. anthracis using reverse tran-
scriptase quantitative PCR. Under host-mimicking culture conditions in
medium containing bicarbonate and CO2, increased expression of different
genes encoding virulence factors or regulators (e. g. capB, acpA and pagA)
could be detected. Levels of enhanced expression were comparable in B.
cereus bv anthracis and B. anthracis and led to increased production of cap-
sule and toxins. Although the B. anthracis-specific nonsense mutation in the
gene for the transcriptional regulator PlcR is absent in B. cereus bv anthracis,
the regulator is also inactive due to a frameshift mutation in the plcR gene,
resulting in non-hemolytic growth like B. anthracis. In contrast to B. anthracis,
spores of B. cereus bv anthracis displayed no intracellular outgrowth into
vegetative bacteria or replication after infection of macrophages. The reason
for this feature remains speculative. Diagnostics of these new African Bacil-
lus isolates as anthrax-causing bacteria is hindered by the fact that they dif-
fer from typical B. anthracis both in microbiological (e. g. motility, resistance
to gamma-phage) and molecular (e. g. lack of prophage regions) features.
The emergence of the isolates as well as the source of animal infection remain
unclear.

BIODEFENSE PROGRAM & ABSTRACTS 37


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

tered (incidence - 1.77 per 100 000); In addition, 10 outbreaks were de-
045 (B) scribed, affecting 36 cases in total. The most common exposure risk was con-
Efficacy Testing of Orally Administered Antibiotics against an tact with animals or animal products (82.27%). The infection had seasonal
Inhalational Bacillus anthracis Infection in BALB/c Mice distribution; a majority of cases (93.67%) occured during the summer and
G. J. Hatch, S. R. Bate, J. Fretwell, J. Vipond, S. G. P. Funnell, A. D. G. Roberts; early fall. All 79 cases were investigated (bacteriology, real time-PCR);
Hlth. Protection Agency, Salisbury, United Kingdom. 75.94% (60/79) were confirmed by Real-Time PCR and 44.3% (35/79) by bac-
Introduction: The efficacy of therapies against inhalational anthrax cannot teriology. Environmental samples were also studied to determine epidemio-
be studied in human clinical trials and must be evaluated in animal models. logical links. Laboratory investigation at NCDC was performed according to a
In a series of three studies conducted in BALB/c mice we evaluated the effi- DTRA recommeneded algorithm. Data indicate that in Georgia during 2011
cacy of post-exposure, orally administered antibiotics in preventing disease the incidence rate of anthrax in humans had significantly increased in com-
following inhalational exposure to Bacillus anthracis. Materials & Methods: parison to that in previous years. We suggest that a very hot weather condi-
Groups of female BALB/c mice were challenged with an aerosol of approxi- tion and an incomplete registration system for cattle and subsequent lack of
mately 20 LD50s of B. anthracis, Ames. From one day post-exposure, once sufficient vaccination of domestic animals in the country had been reflected
or twice daily oral gavage of 0.2ml of either the test antibiotic or carrier con- in the presented data.
trol commenced. Treatment continued for thirty days after which the animals
were observed for a further fourteen days. The test antibiotics were lev-
ofloxacin, amoxicillin, co-amoxiclav (amoxicillin plus clavulanic acid), 047 (B)
azithromycin and clarithromycin; the carrier control was sterile distilled water. Proteomic Analysis of Atypical Y. pestis Strains Isolated from
Six mice from each group were killed and their blood processed to confirm the Natural Foci in Georgia
treatment peak and trough serum levels. Throughout the studies the mice R. Solomonia1, M. Nozadze11, E. Mikeladze1, E. Zhgenti2, G. Chanturia2, M.
were observed for symptoms of disease and a clinical score assigned. Cause Kekeklidze2, R. J. Obiso3, S. Francesconi4, L. Bakanidze2; 1Life Sci. Res. Ctr., Ilia
of death was confirmed by the presence of bacteraemia. Results: A summary State Univ., Tbilisi, Georgia, 2The Natl. Ctr. for Disease Control and Publ. Hlth.,
of the protective efficacy of the test antibiotics is presented in Table 1. Tbilisi, Georgia, 3The Microbe Company, Christiansburg, VA, 4Naval Med. Res.
Table 1 Ctr., Frederick, MD.
Treatment Treatment % Median time Log-rank statistic
Yersinia pestis is the etiological agent of plague. The National Center for Dis-
dose survival to death (days) compared to control
ease Control has evaluated 40 strains of Y. pestis from two foci regions in
Georgia and from foci of neighbor countries. Genetic analysis of these strains
levofloxacin 80 mg/Kg BID 97 – p < 0.001 was obtained for these isolates of Y. pestis; the data indicates that several
amoxicillin 30 mg/Kg QD 95 – p < 0.001 Georgian isolates are atypical from Y. pestis type strains. Alterations in Y.
co-amoxiclav 30 mg/Kg QD 79 – p < 0.001 pestis virulence factor gene expression are characterized by a shift in tem-
perature and calcium ions. This property of Y. pestis is determined by low-
azithromycin 12-6 mg/Kg QD 5 1.92 p = 0.198
calcium response (LCR) elements and all Georgian isolates of Y. pestis contain
clarithromycin 30 mg/Kg QD 0 1.67 p = 0.815 this DNA sequence in spite of lacking other virulence plasmids. In this study,
control (water) 0.2 ml BID 3 1.92 – we evaluated protein expression of these atypical isolates at different phys-
iological conditions. Y. pestis was studied using 2-Dimensional (2-D) gel elec-
trophoresis and Mass-Spectrometry (MS). We selected four genetically
Conclusion: The BALB/c mouse appears to be an appropriate model for the
distinct Y. pestis isolates, (2944, 8787, 1853 and 1390), which were grown at:
evaluation of post-exposure therapies for inhalational anthrax. Orally ad-
(i) 280C; (ii) 280C +Ca2+; (iii) 370C and (iv) 370C +Ca2. Gels were stained and
ministered post-exposure prophylaxis over a 30 day period with levofloxacin,
protein spot maps and intensities were compared. Proteins that show con-
amoxicillin or co-amoxiclav conferred significant protection from inhalational
sistent, 2-fold differences in expression were excised and analyzed by Mass
anthrax whilst azithromycin and clarithromycin failed to protect against mor-
Spectrometry to determine identity. Selected isolates of Y. pestis show sev-
bidity or mortality in this murine model.
eral differentially expressed proteins including: virulence-related outer mem-
brane proteins, IV secretory pathway VirB4 components, Peptidyl-prolyl
046 (B) cis-trans isomerases, trigger factor, the F1 capsule antigen and others. At-
tempts will be made to integrate genomic and proteomic information to the
Human Anthrax Outrbreaks in Georgia in 2011 molecular biology of these isolates. These studies contribute to our under-
L. Malania1, N. Tsertsvadze1, N. Abazashvili1, M. Ramishvili1, M. Broladze1, standing of the uniqueness of Georgian and Caucus region Y. pestis strains.
S. Tsanava1, N. Avaliani1, R. J. Obiso2, P. Imnadze1; 1The Natl. Ctr. for Disease In addition, this research may influence the development of new diagnostics,
Control and Publ. Hlth., Tbilisi, Georgia, 2The Microbe Company, Christians- which can be applied to the Georgian surveillance program.
burg, VA.
Anthrax is a bacterial disease caused by a Gram positive, rod-shaped, spore-
forming bacterium; a zoonosis that affects wild and domestic mammals. An- 048 (B)
thrax is a widely spread endemic infection in Georgia, which remains a
Clinical and Epidemiologic Assessment of Brucellosis in Georgia
permanent threat to humans due to active foci. Within last five years (2006-
T. Onashvili1, E. Mamisashvili11, R. J. Obiso2, M. Nikolaishvili, M. Zakareishvili,
2010) 28-62 human cases of anthrax are registered annually. In 2011, 10
I. Beradze; M. Donduashvili, M. Kokhreidze, N. Vepkhvadze; 1Lab. of Ministry
small outbreaks and 43 sporadic cases were reported. Clinical, laboratory
of Agriculture (LMA) Tbilisi, Georgia, Tbilisi, Georgia, 2The Microbe Company,
and epidemiological aspects of all sporadic cases and outbreaks were stud-
Christiansburg, VA.
ied. Field trips were conducted in each affected area. Samples, collected from
Brucellosis, a bacterial disease caused by the genus Brucella, is a zoonosis
all potentially exposed individuals and suspected contaminated areas were
and significant cause of reproductive losses in animals. Brucellosis is widely
screened for the presence of B.anthracis. Human cases were clustered and
spread among the humans and animals in Georgia. Human infection occurs
analyzed by age, sex, location, source, laboratory investigation and case def-
from exposure to infected animals or animal products. In this study, we ana-
inition. Data were entered and analysed by the Electronic Integrated Disease
lyzed blood, serum, and milk samples from animals collected in Georgia using
Surveillance System (EIDSS). To date, 79 human anthrax cases were regis-

38 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

an algorithm of tests, including PCR and AMOS PCR to detection Brucella microflora when used as antibacterial therapeutics. The purpose of this work
species. From July 2008 through October 2011 we collected approximately was to test the efficiencies of plating of 11 bacteriophages on Y. pestis, Y.
9000 animal specimens seasonally (spring, fall) in three regions of Georgia: pseudotuberculosis, and Escherichia coli at 28 and 37°C. They included four
Kakheti, Kvemo Kartli and Imereti. To date, 32 strains (B. abortus and B. plague diagnostic phages (Pokrovskaya, φA1122, L-413C, and Y), three
melitensis) have been isolated and are being characterized genotypically. A pseudotuberculosis diagnostic phages (D’Herelle-m, PST, and R), two well-
majority of positive cases come from the Kvemo Kartli (50%) region, whereas known coliphages (T7 and P2 vir1), one new isolate from sewage (φJA1), and
25% come from Kakheti and 16% from the Imereti region. This study en- a T7 host range mutant (T7Yp). Only two phages did not form plaques on E.
hanced the capacity of the Georgian Ministry to identify and conduct sur- coli: Pokrovskaya and φJA1. The rest of the phages could plate on E. coli K-12
veillance for brucellosis in animal populations in Georgia. An accurate and albeit with lower efficiencies at 28°C than on the corresponding Yersinia
efficient surveillance system is of economic value and impacts the health of species. The EOP assays allowed the identification of phages with both high
the Georgian population. This study provided improved laboratory diagnos- diagnostic value and therapeutic potential, which included φA1122, L-413C,
tics and case definitions of Brucella infection in Georgia. Pokrovskaya, φJA1, Y, T7Yp (for plague), and d’Herelle-m (for pseudotubercu-
losis).

049 (B)
Members of the FPT Subfamily of MFS Transporters are Impor-
051 (B)
tant for Pathogenesis of Francisella tularensis and Contribute Biosynthesis of Lipoteichoic Acid in Bacillus anthracis
to Modulation of the Host Cytokine Response G. Garufi1,2, A. Hendrickx1,2, S. G. Richter1,2, K. Beeri1, J. Kern1, O.
1 2 1 1 1 1
M. Marohn , A. Santiago , K. Shirey , S. Vogel , E. Barry ; Univ. of Maryland Schneewind1,2, D. Missiakas1,2; 1The Univ. of Chicago, Chicago, IL, 2Howard
Sch. of Med., Baltimore, MD, 2Univ. of Virginia Sch. of Med., Charlottesville, Taylor Ricketts Lab., Chicago, IL.
VA. Many Gram-positive bacteria synthesize lipoteichoic acids (LTA), zwitterionic
Francisella tularensis (Ft) is the causative agent of tularemia. Due to its polymers that are covalently attached to a glycolipid anchor moiety. In
aerosolizable nature and low infectious dose, Ft poses a significant threat as Staphylococcus aureus and Listeria monocytogenes, the membrane protein
a potential bioterror agent, is classified as a category A select agent by the LtaS or its homologues are responsible for the synthesis of polyglycerol-
CDC and, therefore, is a priority for vaccine development. Survival and repli- phosphate LTA from phosphatidyl-glycerol substrate. Mutations in the ltaS
cation in macrophages and other cell types is critical to Ft pathogenesis, and gene(s) of S. aureus or L. monocytogenes cause defects in cell division that
impaired intracellular survival has been linked to reduction in virulence. The interfere with bacterial growth. Bacillus anthracis, a spore-forming Gram-pos-
Ft genome is predicted to encode 31 Major Facilitator Superfamily (MFS) itive microbe and the causative agent of anthrax, has previously been re-
transporters, and the 9 member Ft phagosomal transporter (Fpt) subfamily ported to lack teichoic acids. However, we identified four ltaS homologues,
possesses homology to virulence factors in other intracellular pathogens. here designated ltaS1-4, in the genome of B. anthracis. Using LTA-specific
Our hypothesis is that these MFS transporters will play a role in the intracel- monoclonal antibodies, LTA synthesis was observed in the envelope of B. an-
lular survival and replication of Ft and in host response to infection and may thracis vegetative forms. B. anthracis mutants lacking ltaS1, ltaS2 or ltaS3
serve as viable targets for attenuation and vaccine development. We demon- did not display defects in growth or in polyglycerol-phosphate synthesis.
strated altered intracellular replication kinetics and attenuation of virulence Deletion of ltaS4 led to the production of LTA molecules with aberrant mass.
in mice compared to WT Ft LVS in 3 of the 9 Fpt mutant strains. Importantly, B. anthracis strains lacking both ltaS1 and ltaS2 were unable to synthesize
we showed that vaccination of mice with these mutants was protective LTA and exhibited reduced viability, diminished growth, altered envelope mor-
against lethal i.p. challenge. Here we present studies that characterize the phology and aberrant cell division. Expression of some, but not all, B. an-
cytokine response to infection with Fpt mutant strains in comparison to thracis ltaS genes in S. aureus complemented the growth and LTA synthesis
parental LVS. While the cytokine responses to infection with these mutant defects of an ltaS mutant. Further, expression of some B. anthracis ltaS ho-
strains are largely unaffected in murine macrophages when compared to the mologues was sufficient to promote LTA synthesis in the Gram-negative mi-
response to LVS, there are pronounced differences in cytokine profiles in the crobe Escherichia coli. Thus, similar to S. aureus and other Gram-positive
livers of infected mice. We believe that these alterations in in vivo cytokine re- microbes, B. anthracis employs LtaS enzymes to synthesize polyglycerol-
sponse are a major contributor to the attenuation observed in these strains. phosphate LTA, which is an essential component of the envelope that pro-
Studies are currently underway to investigate further the cellular and mo- motes bacterial growth and cell division.
lecular mechanisms that underlie the observed alterations in cytokine re-
sponses. These results confirm that this subset of MFS transporters plays an
important role in the pathogenesis of Ft and suggest a strategy for attenua-
052 (B)
tion towards the development of an efficacious vaccine. Resistance of Francisella to Fosmidomycin Associated with
Mutations in the Glycerol-3-Phosphate Transporter
R. S. Mackie1, E. S. McKenney2, M. L. van Hoek1; 1George Mason Univ., Man-
050 (B) assas, VA, 2Univ. of Virginia Hlth. System, Charlottesville, VA.
Testing Diagnostic and Potential Therapeutic Values of Bacte- The methylerythritol phosphate pathway (MEP) is present and is absolutely
riophages Capable of Lysing Yersinia pestis and Yersinia required in most prokaryotes and some lower eukaryotes but absent from
pseudotuberculosis human cells, and thus is an attractive pathway for antimicrobial drug devel-
A. A. Filippov, K. V. Sergueev, Y. He, M. P. Nikolich; WRAIR, Silver Spring, MD. opment. The formation of MEP is catalyzed by 1-deoxy-D-xylulose 5-phos-
Several bacteriophages (phages) have been used for the diagnosis of plague phate reductoisomerase. The MEP pathway is critical for the formation of
and pseudotuberculosis. Some of them could also be utilized for antibacter- isoprenoids, used for cell wall synthesis and electron transport. MEP pathway
ial therapy of corresponding drug-resistant infections. There are very limited genes have been identified in many Category A and B biothreat agents, in-
data on comparative efficiency of plating (EOP) of phages capable of lysing cluding Francisella tularensis, Bacillus anthracis, and Yersinia pestis. Fos-
Yersinia pestis and Yersinia pseudotuberculosis. Such tests performed at midomycin inhibits DXR from F. tularensis subsp. holarctica LVS. This
both diagnostic (28°C) and body (37°C) temperature would help to better es- compound also inhibits the growth of Francisella tularensis NIH B38, F. novi-
timate the diagnostic value of phages and their potential impact on normal cida and F. tularensis subsp. holarctica LVS bacteria. We observed some

BIODEFENSE PROGRAM & ABSTRACTS 39


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

breakthrough colonies of F. novicida growing in the presence of fos- lected as the model bacterium to study for interaction with F. tularensis, as it
midomycin, suggesting spontaneous resistance. Sensitivity of these colonies is one of the most prevalent organisms in the soil-microbiome, a common
to a lipophilic pro-drug of FR900098 was not abated suggesting that spon- opportunistic human pathogen, and a species that F. tularensis could likely
taneous resistance is not due to mutation of DXR. We predicted that the Glyc- encounter in the soil. We have shown that Francisella and Pseudomonas can
erol-3-Phosphate transporter (GlpT) which allows entry of fosmidomycin into influence each other’s growth in vitro. P. aeruginosa produces HQNO (2-hep-
the bacteria would be defective, as we have shown that transposon insertion tyl-4-hydroxy quinoline-N-oxide) that causes a significant promotion of Fran-
mutants at the glpT locus are resistant to fosmidomycin. We performed DNA cisella tularensis novicida growth and biofilm formation of 13 and 26%,
sequencing of glpT from the fosmidomycin-resistant colonies and demon- respectively (p < 0.01). Our hypothesis is that P. aeruginosa HQNO and other
strated multiple mutations including nucleotide deletions and additions in small soluble factors may have specific effects on F. tularensis growth and
this gene. The frequency of mutation at the glpT locus in F. novicida that leads biofilm formation mediated through specific changes in Francisella gene ex-
to fosmidomycin resistance was determined to be 6.3 x 10-8. Thus we con- pression in response to this interspecies signal. Quantitative assessment of
clude that one mechanism of resistance of Francisella novicida to fos- the effect of HQNO on F. tularensis growth and biofilm formation was per-
midomycin is caused by mutations in glpT. This is the first description that we formed, including a titration curve of HQNO, where significant effects on
are aware of for spontaneous mutation in Francisella leading to fosmidomycin growth and biofilm were observed at 385 nM. Static Francisella biofilms and
resistance. biofilms under flow conditions were also examined. Further quantitative
analysis of F. tularensis biofilm was performed with COMSTAT using CLSM im-
ages of Francisella biofilms following HQNO treatment. We have shown that
053 (B) P. aeruginosa produces a small molecule that significantly affects F. tularen-
Control of Pigment Production in a White Phenotypic Variant of sis novicida growth and biofilm formation. In future work, we will assess the
Staphylococcus aureus effect of HQNO on Francisella virulence and gene expression. This study sug-
V. Antonic1, A. Stojadinovic2, M. Izadjoo1, M. R. Alavi1; 1Henry M. Jackson gests a mechanism of this previously unknown interspecies interaction be-
Fndn. for the Advancement of Military Med., Gaithersburg, MD, 2Combat tween F. tularensis and a common soil bacterium P. aeruginosa and may lead
Wound Initiative Program, Bethesda, MD. to a greater understanding of F. tularensis persistence in the natural envi-
Background: Difficult to treat Traumatic wound infections are usually caused ronment.
by multiple species of bacteria colonizing the wound bed. We isolated two
pigment variants of Staphylococcus from a wound sample, which displayed
white and yellow phenotypes. Yellow pigment is a manifestation of staphy-
055 (B)
loxanthin biosynthesis carried out by crt operon enzymes. Staphyloxanthin is A Method for Detection of Bacillus anthracis Spores in Complex
a virulence factor, which protects the microorganism against the host oxygen Matrices
free radicals and antimicrobial peptides. We used biochemical and molecu- O. Stephansson1,2, P. Ågren3,2, S. Byfors3,2, S. Ehrs1,2, M. Klint1,2, M. Lavan-
lar biological methods to characterize the white and yellow pigment variants. der3,2, C. Nilsson3,2, A. Lundin Zumpe3,2; 1Natl. Vet. Insitute, Uppsala, Sweden,
2
As quorum signaling by Pseudomonas aeruginosa is shown to affect Staphy- Swedish Joint Lab. for Food Safety and Biopreparedness, Uppsala, Sweden,
3
lococcus gene expression, we explored the potential influence of P. aerugi- Natl. Food Agency, Uppsala, Sweden.
nosa on Staphylococcal pigment production. Results: Biochemical tests by In 2004 the Swedish Laboratory for Food Safety and Biopreparedness was
Phoenix instrument and 16s Ribosomal DNA sequencing indicated that both established in collaboration between the National Food Agency (NFA) and
pigment variants were Staphylococcus aureus with similar antibiotic resist- the National Veterinary Institute (SVA). These agencies share responsibility to
ance profiles. However, staphyloxanthin production in the yellow variant was ensure a secure food chain and have a mutual interest to develop and sustain
2-fold higher than the white as measured by aceton/methanol extraction and diagnostic preparedness in case of unusual contamination of the food chain.
spectrophotometry. We examined the crt operon from the two pigment vari- In times of globalization, climate change and as a result of increased aware-
ants by sequencing. The promoter region and several biosynthetic genes that ness, the ability for reliable analysis of unusual/antagonistic contamination
have been sequenced to date are identical. We tested the effect of of the food chain is emphasized. This joint effort focuses on establishing
Pseudomonas signaling on pigment production. Interestingly, P. aeruginosa methods for sample preparation and molecular detection of BSL 3 agents in
induces pigment production in the white S. aureus variant when they are in- food, fodder, drinking water and livestock tissue samples. Work has been fo-
cubated next to each other on a plate. Conclusion: The yellow and white S. cused on developing a diagnostic capability for Bacillus anthracis, an agent
aureus variants are similar in both biochemical and antibiotic resistance pro- that has largely been considered extinct in Sweden. Hence, the reoccurrence
files; however they differ in staphyloxanthin production. Interestingly, the of anthrax at a farm in 2008 and a recent outbreak in cattle during 2011 il-
white variant is capable of pigment production in the of presence P. aerugi- lustrated the importance of upholding biopreparedness also for unusual
nosa. This observation and our sequencing data suggest that crt operon is agents. The current project concerns molecular detection of B. anthracis
functional in the white S. aureus variant but production of the pigment is con- spores in complex matrices. A protocol for germination followed by extraction
trolled by P. aeruginosa signaling. We are currently investigating the mecha- of nucleic acids and detection by real time PCR has been developed and is
nism of this control. presented here. Limits of detection (LOD) for twelve matrices was determined
and the results show that due to the highly varying properties of the sam-
ples LOD ranges from 9 to 16 000 spores/g. Further, in some cases there was
054 (B) no detection of B. anthracis, why sample preparation and/or the PCR proto-
Interspecies Interaction of Francisella tularensis and col has to be optimized to enable detection in these matrices.
Pseudomonas aeruginosa
S. N. Dean, M. L. van Hoek; George Mason Univ., Manassas, VA.
The objective of this study is to elucidate the interspecies interactions of Fran-
cisella tularensis within the polymicrobial natural environment and within the
host. These results will improve our knowledge of the persistence of this or-
ganism in its natural habitat, and may identify factors that can promote or
inhibit growth of this fastidious organism. Pseudomonas aeruginosa was se-

40 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

which strains to include in the reference panel for future animal studies. Our
056 (B) review resulted in the identification of 109 strains that met one or more of the
Comparative Genomic MGE Finding Process Streamlines Mobile parameters in the scoring matrix. Based on our scoring system, strains
Element Search K96243, MSHR305, MSHR730, 1106a, S191, and MSHR668 scored highest
S. Y. Choi1,2, N. A. Hasan1,2, J. Chun3, M. Hoq4, A. Huq5, T. A. Cebula6,2, R. R. and are suitable for inclusion in the reference panel. These strains were iso-
Colwell1,6,2; 1Univ. of Maryland, College Park, MD, 2CosmosID Inc., College lated from melioidosis patients in Thailand or Australia, have either yersinia
Park, MD, 3Sch. of Biological Sci. and Inst. of Microbiol., Seoul, Korea, Re- or B. thuringiensis-like flagella, and can be obtained from a source with a
public of, 4Univ. of Dhaka, Dhaka, Bangladesh, 5Univ. of Maryland, College known passage history.
Park, MD, 6Johns Hopkins Univ., Baltimore, MD.
Horizontal gene transfer (HGT) is a dynamic process that generates diversity
and drives microbial evolution. Moreover, these mobile genetic elements 058 (B)
(MGEs) with key features including location, size, sequence content, as well Paratransgenic and Transgenic Analysis of Cellular Interac-
as genomic architecture, are important attributes enabling pathogens in the tions between Ixodes Ticks, Mammalian Cells, and Rickettsiae:
environment to be tracked to their ultimate source. A comparative genomic Lighting the Way with Fluorescent Proteins
approach was used to predict, locate, and characterize MGEs in Vibrio T. J. Kurtti, J. D. Oliver, N. Y. Burkhardt, R. F. Felsheim, M. J. Herron, U. G.
cholerae utilizing both ORF-dependent and ORF-independent approaches. Munderloh; Univ. of Minnesota, St. Paul, MN.
With a GI-definition of five or more contiguous ORFs, 130 MGEs ranging from Ixodes ticks, most notably Ixodes scapularis and I. ricinus, harbor diverse mi-
2.9 to105 kb were identified among 46 V. cholerae genomes. While all known crobial communities. Members include the more prevalent endosymbionts
pathogenicity islands and regions associated with variable serotypes were known as the Rickettsial Endosymbiont of I. scapularis (REIS) and C. Midichlo-
identified by this method, most MGEs were novel, and BLASTP searches of ria mitochondrii in I. ricinus. In addition, these ticks serve as vectors for a
ORFs against the nucleotide sequence database yielded no extraneous hits. wide range of protozoan, bacterial and viral pathogens of humans as well as
Such a large number of MGEs indicates that although the current pandemic domestic and wild animals. These include special interest agents such as
of V. cholerae is caused by strains belonging to a single phyletic line, they Rickettsia rickettsii and Francisella tularensis. We are using paratransgenic
have diversified by lateral gene transfer from a Vibrio gene reservoir in the and transgenic approaches to examine the interactions between rickettsiae
natural environment. Notably, distribution of MGEs in non-O1/O139 V. and hosts at the cellular level. Rickettsiae are transformed using constructs
cholerae strains is much more diverse than in O1 strains. Whereas in O1 that incorporate portions of plasmids found in Rickettsia amblyommii,
strains the number of MGEs appears to plateau at approximately 40 MGEs pRAM18, pRAM23 and pRAM32, and backbones of commercially available E.
(analysis of 25 toxigenic V. cholerae O1 genomes), the number of MGEs dra- coli plasmids. These shuttle vectors are engineered to contain a multiple
matically increased to approximately 140 when non-O1/O139 strains were cloning site for facile insertion of genes to be tested, and antibiotic resist-
added. From an evolutionary standpoint, V. cholerae non-O1/O139 appears ance and fluorescent protein markers. Diverse rickettsiae indigenous to North
to be the ‘genetic harbinger’ of MGEs destined to appear in V. cholerae O1 America have been transformed using these shuttle vectors and will be use-
strains. Thus, by monitoring the ‘genetic cauldron’ of non-O1/O139 strains, ful for tracking rickettsial growth and development in ticks or tick and mam-
it becomes possible to forecast elements that endow toxigenic V. cholerae malian cell cultures. In addition, we have transformed I. scapularis cells by
O1 strains with survival advantages for new and future hostile environments. adapting a method used to transform vertebrate cells, the Sleeping Beauty
transposase system. We transformed I. scapularis cell line ISE6 and Vero cells
057 (B) to express a red-fluorescent peptide that binds to f-actin to examine rick-
ettsial motility. Live imaging of transformed REIS during colonization of tick
A Literature-Based System for Selecting Burkholderia pseudo- organs and dispersal in ticks suggests that there is co-evolutionary adapta-
mallei Strain Panels for Animal Model Testing Under the FDA tion between tick species and their symbionts which serves to restrict ex-
Animal Rule change of rickettsiae to other tick species.
K. E. Van Zandt1, A. Tuanyok2, H. C. Gelhaus, Jr.1, K. T. Brittingham1, S. A. Sar-
razine1, P. S. Keim2, R. L. Warren1; 1Battelle, Columbus, OH, 2Northern Arizona
Univ., Flagstaff, AZ. 059 (B)
Burkholderia pseudomallei is a gram-negative bacterial pathogen responsi- Comparison of Challenge Outcomes with Three Different Bacil-
ble for the disease melioidosis and is considered a biodefense threat. Current lus anthracis Isolates in New Zealand White Rabbits
treatment of melioidosis involves 2-4 antibiotics administered for up to six A. Belle1, L. Schaefer1, A. E. Boyer2, M. Gallegos-Candela2, J. R. Barr2, M. de
months and even with treatment there is a 15% relapse rate. Therefore, there la Garza3, J. Nunneley3, R. Carrion, Jr.3, K. Wycoff1; 1Planet Biotechnology Inc.,
is significant interest in developing improved medical countermeasures Hayward, CA, 2CDC, Atlanta, GA, 3Texas BioMed. Res. Inst., San Antonio, TX.
(MCMs) against melioidosis. A variety of animal models, bacterial strains, Bacillus anthracis is a gram-positive spore forming bacterium that causes in-
and experiment conditions have been used to study melioidosis, resulting in halational anthrax. While rabbits are an accepted model for studying anthrax,
disparate data and difficulty comparing studies. We have intiated the devel- every laboratory has its preferred strain/isolate of B. anthracis and values re-
opment of animal models of melioidosis suitable for licensure of MCMs under ported in the literature for time to death, bacteremia levels, and other dis-
the FDA Animal Rule which requires consistent, well characterized etiologic ease parameters vary widely. In this study, groups of four New Zealand white
agent. This is difficult in the case of B. pseudomallei because the large rabbits were challenged with an equal number of spores (2.1 x 107 CFU), by
genome of B. pseudomallei is highly diverse among various isolates. To bet- the same route (nasal instillation), with one of three B. anthracis isolates and
ter meet the FDA Animal Rule guidance and to assist in standardizing research the time to death was determined. The isolates were NR-3838 from BEI Re-
conducted across laboratories, we are developing a panel of strains that will sources (derived from Ames), Ames from USAMRIID and Vollum from USAM-
be used to achieve consistency among laboratories developing and evaluat- RIID. Bacteremia was quantified at various times post-challenge. Other key
ing melioidosis MCMs. To this end, we have reviewed published and unpub- virulence factors in the serum were quantified, including Protective Antigen
lished data on various strains of B. pseudomallei. We ranked each strain (PA), Lethal Factor (LF), Lethal Toxin (LeTx) and Capsular Antigen (PGA or
based upon a scoring matrix which had five categories: animal models, viru- Polyglutamic Acid). There were striking differences between the three iso-
lence, source, passage history, and availability of the strain to determine lates in the rabbit challenge model. Time to death with the USAMRIID Ames

BIODEFENSE PROGRAM & ABSTRACTS 41


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

isolate was 50 (± 4) hr; NR-3838 was 76 (± 28) hr, but only 2 of the 4 rabbits Spray factors for Y. pestis and F. tularensis were calculated over runs at dif-
died; Vollum was 78 (± 4) hr. There were also large differences in the meas- ferent concentrations to be 1.4E-5 and 6.9E-6, respectively. Preliminarily, the
ured virulence parameters, with Vollum>Ames (USAMRIID)>NR-3838. Taken spray factor for B. pseudomallei was determined to be 8.6E-7, although op-
together, our preliminary data suggest that the therapeutic window of op- timization studies are still in progress. The particle size distribution was
portunity is variable between the different B. anthracis isolates and that the found to be within the respirable range (~2μm) for all three organisms tested.
kinetics of inhalation anthrax in rabbits is isolate specific. Conclusion: The PARI LC plus nebulizer is an effective nebulizer for use with
Y. pestis and F. tularensis and provides good consistency and efficient trans-
mission of the bacterial agents through the exposure chamber across a broad
060 (B) range of bacterial concentrations. Preliminary results suggest this nebulizer
Genotyping of Mycobacterium tuberculosis Strains from a is also effective for use with B. pseudomallei.
Third Level Hospital in Lima, Peru
C. A. Tello, Sr., N. A. Borja, R. Garcia de la Guarda; San Marcos Univ., Lima,
Peru.
062 (D)
Objective: To characterize the genotypes of Mycobacterium tuberculosis by A PCR Platform Comparison Study to Support Sustainability in
two PCR based methods and begin an epidemiology study. Methods: Twenty Resource-Limited Settings
one patients diagnosed with tuberculosis (TB) between June and October A. Ongarbaev1, A. Khodiev1, A. Smagin2, V. Garib2, G. Abdukhalilova2, G.
was included in the study. The genotypes of M.tuberculosis isolates were es- Ibadova2, R. Obiso3, S. Francesconi4, A. Baccetty5, J. Callahan6, D. Williams7;
1
tablished by DRE PCR (Friedman et. al., 1995) and PCR based Genotyping Technology Management Company, Tashkent, Uzbekistan, 2Tashkent Inst.
(Kotlowsky et. al., 2004). The susceptibility to isoniazid, rifampicin, and strep- of Postgraduate Med. Ed., Tashkent, Uzbekistan, 3The Microbe Company,
tomycin was studied. Were studied the risk factors for active transmission of Christiansburg, VA, 4Naval Med. Res. Ctr., Silver Spring, MD, 5Defense Threat
TB. Results: A total of 21 M. tuberculosis isolates from samples of new and Reduction Agency, Ft. Belvoir, VA, 6Biotechnology and Diagnostic Solutions
retreated TB cases were collected at Guillermo Almenara National Hospital. Animal Hlth. Life Technologies, Austin, TX, 7Threat Reduction Support Ctr.,
We registered a 66.7% of primary resistance and 33.3% of secondary resist- Alexandria, VA.
ance. The 9.5 % of the patients were VIH+. The 76.2 % were in the range of Background: The US Defense Threat Reduction Agency through its Biological
the productive age. When considered the patients with MDR strains the 80% Threat Reduction Program, equips diagnostic laboratories with real-time PCR
reported previous TB. The epidemiology and demographic data were col- platforms for the surveillance of especially dangerous pathogens within part-
lected by clinical records. Upon DRE and PCR-based genotyping analysis of ner countries. To this end, the existing PCR platform (Roche LightCycler 2.0)
21 M. tuberculosis strains, we reported the presence of 18 and 15 different provided to Uzbekistan is being replaced with a locally accessible and hence
fingerprints respectively, the number of bands for each fingerprint ranged more sustainable system (Qiagen Rotor-Gene Q). Prior to this change, we as-
from 8 to 1 band(s). In the consensus tree we established 3 clusters. The MDR sessed the potential impact of switching PCR platforms. Assays intended for
isolates were non-included in a specific cluster. Conclusions: Lima has a high the LightCycler were tested on the Rotor-Gene. We evaluated and compared
incidence rate of TB MDR. Although many clusters don’t have an epidemiol- sensitivity (Se) and PCR efficiency (E) between the two platforms to ensure
ogy link, the patients of these groups registered a stance in the same hospi- equivalence. Methods: Eight commercial and “in-house” PCR assays for vet-
tal area. The isolates present a high variability of genotypes and a low level erinary diseases were run in parallel on the two PCR platforms. A ten-fold di-
of clonality, possibly due to a long stay in time of different strains in Peru that lution series of RNA- or DNA-positive controls were prepared for real-time
could be creating different stages of microevolution. PCR. Each control and control dilution was treated as an unknown sample
and tested four times on each platform. A standard curve was constructed
for each test. Real-time PCR efficiency was estimated using the formula:
061 (B) E=10^[-1/ΔC] -1. Results: The PCR assays tested retained sensitivity and ef-
Evaluation of a Commercially Available Disposable Nebulizer ficiency on the Rotor-Gene platform within the established limits: Se - 100%
for Use in Nose-Only Pneumonic Plague, Tularensis, and Me- (relative to the LightCycler); and 90%<E<110%. Conclusion: The equiva-
lioidosis Murine Models lence observed between the two PCR platforms indicates that replacing the
LightCycler will not reduce performance capability in Uzbekistan. We suggest
W. C. Lin, K. Siefkas, S-C. Hu, D. Sullivan, B. Gingras, L. Holland; IIT Res. Inst.,
the transfer of assays between different platforms supported by a relevant
Chicago, IL.
comparison study to ensure sustainability.
Objectives: Our objective was to evaluate the PARI LC Plus nebulizer, for the
development of murine models of infection for pneumonic plague, tularemia,
and melioidosis using IITRI’s nose only bioaerosolization system to evaluate 063 (D)
efficacy of experimental therapeutics and vaccines. The causative agents for
these illnesses used for testing the PARI LC plus nebulizer were the highly Development of User-Defined Assays for the Genexpert Plat-
contagious Gram negative organisms Yersinia pestis CO92, Francisella tu- form
larensis SCHU S4, and Burkholderia pseudomallei ATCC 23342, which are E. Felder, R. Wölfel; Bundeswehr Inst. of Microbiol., Munich, Germany.
HHS/USDA Select Agents and NIAID Category A and B Priority Pathogens. One of the major tasks of the Department for Medical Bio-Reconnaissance
Methods: 100mL cultures of Y. pestis, F. tularensis, and B. pseudomallei were and Verification at the Bundeswehr Institute of Microbiology in Munich is
grown in Heart Infusion Broth with 1% xylose at 28°C, Brain Heart Infusion rapid and reliable identification of biological agents and other dangerous
Broth with IsoVitaleX at 37°C in 5% CO2, or Trypticase Soy Broth at 37°C, re- pathogens even under restrictive field conditions comprising varying climate
spectively. The bioaerosol system utilized a 64-port Cannon-style nose only and limited cooling capacity at missions abroad. The GeneXpert system from
exposure chamber and six PARI LC Plus nebulizers operating at 20psi for Cepheid, available in Germany since a few years, combines sample process-
10minutes. Aerosol sizing was measured with an Anderson 6-stage impactor ing and real-time PCR detection from e.g. human samples in one cartridge,
while aerosol titer was measured with dual all-glass impingers. Determina- saving time consuming and labour intensive working steps from specifically
tion of spray factor and assessment of aerosol consistency were accom- trained laboratory personal. At this time point kits for the detection of bio-
plished through independent aerosolization of the three organisms with logical agents in humans are not commercially available. Nevertheless
dilutions at concentrations that ranged between 1E3 to 1E8 CFU/mL. Results: Cepheid offers an open platform for in-house development of cartridges from

42 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

third parties. The open GeneXpert platform was used at the Institute of Mi- ger to the human population. As a preparedness of such incidents, a rapid de-
crobiology to develop cartridges for the detection of Yersinia pestis, Fran- tection system for identification of biothreat agents is needed. The S.O.N.D.E.
cisella tularensis, Brucella abortus and Burkholderia mallei from bacterial project intends to develop a suitcase size platform for the detection of cate-
cultures as well as Crimean Congo hemorrhagic fever virus from viral sam- gory A biothreat pathogens by nucleic acid and antigen detection. One sub-
ples. PCR chemistry for real time PCR detection from bacterial (qPCR) and project is dealing with the development of molecular methods such as
viral (RT-qPCR) samples was especially compiled for freeze drying. Complete enrichment of pathogens and nucleic acid isolation. These assays are de-
PCR mastermix pellets which can be placed into the cartridges in one piece signed to be used in an integrated bio-analytical system being developed by
could be generated for bacterial PCR detection. Software programs for sam- our S.O.N.D.E. project partners for rapid detection of a number of pathogens
ple preparation and PCR of each biological agent were separately written and and toxins. To concentrate enveloped and non-enveloped viruses from large
linked with the GeneXpert cycler. Using those ready-to-use software pro- volume biological samples, an enrichment method based on polymer was
grams together with our in-house freeze dried real time PCR mastermixes and developed. Viruses captured by polymer were successfully analyzed for nu-
the automatic sample processing from the GeneXpert system, we could sig- cleic acid detection, virus infectivity assay and electron microscopy. Also the
nificantly reduce hands-on time for the end user. The freeze-drying method nucleic acid purification using paramagnetic beads was successfully devel-
for the generation of complete real-time PCR mastermixes as well as for a oped and tested on different model pathogens (gram+ and gram- bacteria,
bacterial sample processing control was well established and provides a RNA and DNA viruses). Purified nucleic acid was detected by real-time PCR
basis for the development of more assays for the detection of bacterial and an isothermal amplification method for different pathogens in spiked
agents. In summary, the GeneXpert system with its in-house designed car- samples and the limit of detections was 10-100 genome copies/ml. Cen-
tridges is a suitable platform for the detection of biological agents and could trifugal microfluidic platform and portable device have been developed by
be used even in a low-resource laboratory environment. our project partners for the detection of biothreat pathogens in automated
“sample to answer”-platform. Developed methods like nucleic acid isolation
and isothermal amplification were integrated into this platform. Furthermore,
064 (D) this platform is going to be evaluated and compared with conventional lab-
Production and Qualification of Unrestricted Reagents to Sup- oratory methods as well as commercial available rapid diagnostic platforms.
port Anthrax Assays
K. H. Clement, T. L. Rudge, Jr., N. T. Huber, K. A. Newman, K. A. Sankovich, R.
H. Migliozzi, C. L. Sabourin; Battelle, Columbus, OH.
066 (D)
Thousands of clinical and non-clinical samples are tested each year in various Indirect Phage-Based Detection of Yersinia pestis: Field and
in vitro assays to assess responses to anthrax vaccines and therapeutics. Clinical Applications
Under a National Institute of Allergy and Infectious Diseases (NIAID) contract, K. V. Sergueev, M. P. Nikolich, A. A. Filippov; WRAIR, Silver Spring, MD.
we validated (for several specific intended uses) lethal Toxin Neutralization Lysis with specific bacteriophages is often used for the identification of
Assays (TNA), the anti-Protective Antigen Immunoglobulin G (anti-PA IgG) En- Yersinia pestis and plague diagnosis, but the assay usually includes the iso-
zyme Linked Immunosorbent Assays (ELISA) and PA ELISAs for multiple lation of pure Y. pestis culture and takes about three days. We have recently
species. These assays were validated using reagents provided by NIAID’s developed an alternative, rapid and sensitive test of indirect Y. pestis detec-
reagent repository, BEI Resources, which cannot be used for commercial pur- tion based on quantitative real-time PCR (qPCR) monitoring of the amplifica-
poses. Therefore, new reagents were recently produced, characterized, and tion of reporter phages φA1122 and L-413C (Sergueev et al., 2010; PLoS ONE
then qualified using BEI Resources’ reagents as the reference standard. Crit- 5(6): e11337). In this work, we tested potential field application of this
ical reagents including cell banks, reference standards and quality controls method. Using JBAIDS platform, a portable field-deployable instrument
were produced in large, stable lots and have been well-characterized to make (Idaho Technology Inc., Salt Lake City, UT), the phage detection limit observed
certain that the assays using these reagents remain in control for future use. (and thus the limit of Y. pestis indirect detection) was identical to that ob-
To date, rare reagents have been produced and qualified for various species tained using a full-sized LightCycler 2.0 real-time PCR instrument (Roche Ap-
for the TNA and ELISA assays. Another critical component to ensuring that plied Science, Indianapolis, IN) and was equivalent to one live bacterial cell
these assays remain in control longitudinally is to monitor reagent stability in four hours. We also explored potential clinical and epizootiological appli-
and functionality. The reagents are tracked in a custom-made Sample Track- cations of this indirect detection method using simulated bovine serum and
ing Database, which inventories all aliquots using unique bar codes and is mouse tissue (spleen, liver) samples. Omitting the step of sample preparation
linked to a specialized BioAssay Database for tracking assay results. These and DNA concentration, satisfactory levels of sensitivity were observed when
databases cross-talk to follow reagent location, freeze/thaw cycles, and to using diluted spleen suspensions. The most promising results were obtained
trend reagent performance in the assays to monitor stability and quality. In with diluted and whole serum. These results indicate that the method of in-
summary, these new, unrestricted reagents have been successfully bridged direct qPCR-mediated phage-based detection of Y. pestis can be used with
to BEI Resources reagents, and show excellent performance in the anthrax as- sample types that would be tested under field or clinical conditions.
says for future testing of clinical and non-clinical samples. These reagents
are available for unrestricted use at Battelle.
067 (D)
Quantification of Anthrax Lethal Factor by Mass Spectrometry:
065 (D) Application to Clinical Anthrax
Development of Rapid Diagnostic Platform for Detection of Cat- A. E. Boyer1, A. R. Hoffmaster1, C. P. Quinn1, M. Gallegos-Candela1, R. C. Lins2,
egory A Biothreat Pathogens in the Field R. A. Stoddard1, E. Steward-Clark3, P. Maniatis1, C. Marston1, L. X. Cronin1, D.
P. Patel1, M. Weidmann2, K. Achazi1, S. Linke1, O. Strohmeier3, D. Mark4, T. Aranio1, H. Li1, V. Semenova3, K. Isbell2, S. Shadomy1, M. Lehman1, D. Blaney1,
van Oordt4, J. Drexler5, M. Eberhard5, F. von Stetten4, M. Niedrig1; 1Robert N. Pesik1, T. L. Smith1, J. R. Barr1; 1CDC, Atlanta, GA, 2Battelle Analytical Serv-
Koch Inst., Berlin, Germany, 2Univ. Med. Ctr., Göttingen, Germany, 3Dept. of ices, Atlanta, GA, 3CDC, Chamblee, GA.
Microsystems Engineering (IMTEK), Freiburg, Germany, 4HSG-IMIT, Freiburg, The toxins of B. anthracis are major virulence factors for anthrax and con-
Germany, 5QIAGEN Lake Constance GmbH, Stockach, Germany. tribute to rapid disease progression by disabling host immune defenses. We
Increased threat of infectious diseases and bioterrorist incidents pose dan- have developed and validated mass spectrometry (MS) based methods that

BIODEFENSE PROGRAM & ABSTRACTS 43


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

can detect and quantify anthrax lethal factor (LF) to measure toxemia during
treatment and provide early diagnosis of anthrax. The MS based method for
069 (D)
LF includes three steps that contribute to sensitivity and specificity: 1) LF spe- Negative Capture for Selective Enrichment of Pathogen-Derived
cific mAb purification, 2) LF specific enzymatic proteolysis of a selected sub- Nucleic Acids in Clinical Samples
strate, and 3) detection of the toxin-specific hydrolysis products by Z. Bent, S. Langevin, V. VanderNoot, P. Lane, D. Curtis, M. Tran-Gyamfi, S.
MALDI/TOF/MS yielding a detection limit of 5 pg/mL. LF levels measured in Branda, T. Lane; Sandia Natl. Labs, Livermore, CA.
rhesus macaques and NZW rabbits have shown an association with survival Second generation sequencing (SGS) has enormous potential for use in de-
and a point of no return. The LF-MS method was applied to human inhala- tecting and characterizing novel, emerging, and engineered pathogens in clin-
tion, cutaneous, gastrointestinal and injection anthrax cases to confirm its ical samples with no a priori knowledge of the pathogen required. However,
ability to detect and quantify toxins in these forms of anthrax and during an- brute force sequencing of clinical samples has limited utility because the
timicrobial and anthrax immune globulin (AIG) treatment. For three inhala- overwhelming majority of sequences map to the host and not the pathogen
tion anthrax cases, anti-PA levels measured by ELISA were consistent genome. We have developed a technique for affinity-mediated depletion of
following AIG delivery (133.49±0.135 μg/ml). LF levels appeared to correlate host-derived nucleic acids (NA) for efficient SGS analysis of pathogen-de-
with the route of exposure and disease form. The LF levels were highest in two rived NA in clinical samples. In this technique, which we call “negative cap-
inhalation anthrax cases (327.59 and 57.985 ng/ml), followed by the geo- ture” the NA from a clinical sample are hybridized with tagged probe
metric mean concentration±standard error for 12 injection anthrax cases sequences that are complimentary to the host. NA that bind to the probes
(13.83±0.49 ng/ml), three gastrointestinal cases (0.525±0.692 ng/ml), and can then be removed from the solution using the probe’s tag, thereby leav-
83 cutaneous cases (0.117±0. 221 ng/ml). LF levels during treatment of a ing behind a population that is enriched for pathogen sequences. Negative
human inhalation anthrax case in 2011 declined gradually with antimicrobial capture can be carried out manually using standard laboratory equipment, or
and rapidly with AIG treatment, similar to that reported previously (Walsh et with less hands-on labor using a meso-fluidic chromatography device that
al, 2007). LF quantification by MS was shown to be effective for toxin detec- we have developed. In each case, negative capture increases the proportion
tion in all forms of anthrax and for monitoring effectiveness of anti-microbial of SGS reads mapping to the pathogen genome, as compared to that yielded
and antitoxin treatments. by brute-force SGS of the original sample. Importantly, the additional “hits”
on pathogen are well distributed across the genome. Thus, negative capture
serves to target SGS to pathogen NA, and thereby improves sequence cov-
068 (D) erage of the pathogen’s genome. This added information will prove to be very
Applying Elastic Light Scatter Technology (BARDOT) to Inte- valuable in characterizing the pathogen and distinguishing it from close rel-
grate Organism Classification atives. Further, due to its flexible nature, negative capture can easily be ap-
H. Motlagh1, A. Giordano1, C. Waters1, E. Bae2, J. G. Thomas1, J. Robinson2; plied to pathogen sequencing from clinical samples of virtually any host.
1
West Virginia Univ., Morgantown, WV, 2Purdue Univ., West Lafayette, IN.
Introduction: BARDOT (BActerial Rapid Detection using Optical scatter Tech-
nology) distinguishes Forward Scatter Phenotype (FSP), showing altered bac-
070 (E)
teria not identified by traditional methods. We expanded our international Surveillance on Plague Natural Foci in Georgia
library for “pattern matching,” assessing the system’s ability to triage com- L. G. Bakanidze, P. G. Imnadze, S. A. Tsanava, N. S. Tsertsvadze; Natl. Ctr.
ponents of Bioterrorism Surveillance: Phenotype/Bioburden, Mechanism of Disease Control and Publ. Hlth. of Georgia, Tbilisi, Georgia.
Resistance, Institutional Surveillance, and Origin. Materials/Methods: BAR- Georgia is located on the crossroad of Europe and Asia, it was one of the
DOT system is comprised of interactive components: red laser reader, com- chains of the Great Silk Road. It always was very vivid transition point, thus
puter, and software. The FSP detected by the laser diode module, integrates it was not protected from spread of different epidemics, and, among them,
into ELS (Elastic Light Scatter), a mathematical function of culture colony re- plague. Plague in Georgian manuscripts first was mentioned in XI century. In
fractive index, texture, size and shape/symmetry. Comparison of ELS was cal- XV century, later in another Georgian manuscript - “Book of Medical Treat-
culated on Circulatory Symmetric Features and Texture patterns, extracting ment-Ustsoro Karabadini” (“Peerless Karabadini”). Only single cases of
107 features from one image. Dilutions of Organisms yielding 25 to 50 iso- plague were mentioned in XVI - XVII centuries, official registration of plague
lated colonies were grown on Mueller Hinton, analyzed at 12, 24 hours. cases started in the XIX century. Several plague epidemics were registered in
Global clinical isolates obtained from South Africa, focused on MRSA, MSSA, Georgia during XIX century, they mainly started in the south Georgia, and
and ESBL producing E. coli and K. pneumonia. Results: Laser scatter images later spread on east, north, west and in Tbilisi. The Transcaucasian Anti-
via FSP were clustered based on ELS for 75 WVUH clinical isolates and 40 Plague Center was founded in Tbilisi In 1933. Later existence of two natural
global isolates enlarging our “library” for “pattern-matching.” Multiple-Drug foci of plague on the territory of Georgia (plain-foothills, high mountainous)
Resistance Organisms had unique, identifiable patterns corroborated by was established. Georgian Anti Plague Station for decades carried out active
Cross Matrix Validation. MRSA (98.6%) could be segregated from S. epider- surveillance on natural foci. Several epizootics were described in both foci.
mis (99.1%), ESBL E. coli (88.4%) segregated from non-ESBL producing E. Totally 122 strains of Yersinia pestis were isolated in Georgia, nowadays 46
coli (99.6%) and ESBL positive K. pneumonia (97.4%) from ESBL-negative strains are kept at NCDC Microbial Library. After collapse of Soviet Union
(97.3%). Globally, matrix analysis showed segregation of WVUH MRSA under difficult economic situation in independent Georgia active surveillance
(90.3%) and MSSA (69.8%) from South Africa MRSA (95.7%) in MSSA on plague natural foci was quitted, and only within the scope of US DoD De-
(93.2%). Based on Scatter Plot Matrix, we extracted ELS “pattern matching” fense Threat Reduction Agency (DTRA)-funded CBR projects reviving it still
to Four-tiered analysis: Potential Bioterrorism Phenotype and Bioburden became possible. In 2005-2009 the CBR project GG-1 “Ecology, Genetic Clus-
(CFU’s/ml), Mechanism of Resistance, Phenotype Location (Institutional Sur- tering and Virulence of Yersinia pestis Strains Isolated from Natural Foci of
veillance), and Infection Control. Conclusion: These give validity to a four Plague in Georgia” enabled us to start surveillance on plague foci, and it was
component Discussion Tree obtained simultaneously for clinical action, lo- continued in next project GG-18 “Active Surveillance on Especially Dangerous
cally and nationally. Pathogens in the Southern Caucuses Region.” Though now new isolates were
obtained during last years, and the plague foci in Georgia are so close to pop-
ulated areas, that they must be under permanent control, in order to be able
to rapidly response to emergencies. Besides, it gives us possibility to study

44 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

Y. pestis isolates from NCDC collection with modern sophisticated methods biological threat in a very precise manner by activating only the modules that
of investigation. are useful to cope with a given situation in the most possible appropriate
way. This modular approach at the same time creates the most economic way
of response in a crisis or as a preventive measure.
071 (E)
Performance of Rapid Viability PCR Method for Detection of
Bacillus anthracis Ames Spores from Sponge-Stick, and Vac-
073 (E)
uum Sock and Filter Sample Types The Microfluidic Bioagent Autonomous Networked Detector (M-
S. R. Shah1, S. Letant2, G. Murphy2, T. Alfaro2, S. Kane2; 1EPA, Washington, BAND): An Update
DC, 2LLNL, Livermore, CA. M. Sanchez, L. Probst, E. Blazevic, B. Nakao, M. A. Northurp; Microfluidic
The U.S. Environmental Protection Agency (EPA) in collaboration with the Systems, Fremont, CA.
Lawrence Livermore National Laboratory (LLNL) has been developing rapid vi- We will update the status of the M-BAND: a fully automated air-borne bio-
ability testing methods for detection of biothreat agents in environmental threat detection system for biosurveillance applications and its internal assay
samples. Following an intentional or accidental anthrax event, hundreds to capabilities. The system, including a multiplexed, nucleic-acid-based detec-
thousands of environmental samples of diverse types could need to be rapidly tion assay, was designed and built with DHS Science and Technology funding,
processed and analyzed in order to both characterize the extent of contami- and shipped for field testing by Microfluidic Systems Inc (MFSI), a subsidiary
nation and determine the efficacy of remediation activities. The Rapid Viabil- of PositiveID Corporation (PSID). Field testing was performed by third par-
ity Polymerase Chain Reaction (RV-PCR) method which integrates high ties in the US and Europe. The system and assay identifies pathogenic strains
throughput sample processing with real-time PCR, has been developed to de- of Bacillus anthracis, Yersinia pestis, Francisella tularensis, Burkholderia
tect Bacillus anthracis Ames spore levels of 10 CFU/sample in wipe, air filter, mallei, Burkholderia pseudomalleiand Variola major virus. In order to assess
and water samples. The method was recently evaluated and optimized for ap- the assay’s ability to detect unknown samples, our team also challenged it
plication to other sample types including vacuum socks and filters, as well as against a large and diverse series of blind samples provided by the Depart-
sponge-sticks. The results indicated that the spore recoveries from these sam- ment of Homeland Security (DHS). These samples included natural occurring
ple types were lower than those from wipe samples. Upon further optimiza- isolated strains, near-neighbor isolates, and environmental samples. Our re-
tion, the RV-PCR method consistently demonstrated a limit of detection at the sults indicate that the multiplex assay was specific and produced no false
level of 10 CFU/sample in the presence of 250 mg of Arizona test dust with positives when challenged with in house gDNA collections and DHS provided
these samples, following an algorithm based on positive results with PCR as- panels. The use of a multiplex assay format saves considerable time and cost,
says targeting one gene each on the B. anthracis pXO1 and pXO2 plasmids. while providing critical sensitivity and specificity. Herein, we present a viable
Additional challenges including high levels of dead B. anthracis Ames spores, analytical tool for the rapid identification of six Centers for Disease Control
and high levels of combined live Bacillus atrophaeus subspecies globigii and Prevention category A and B biothreat organisms, either in an automated
spores and Pseudomonas aeruginosa cells were also tested with vacuum sam- field-based system, or in a laboratory setting.
ples. This work was performed under the auspices of the U.S. Department of
Energy by LLNL under Contract DE-AC52-07NA27344. The National Homeland
Security Research Center of the U.S. EPA funded this research. 074 (E)
Investigation on Drinking Water Quality of the Bogra Sadar Up-
azila: Findings from a Rapid Assessment Lesson in the Bogra
072 (E) District of Bangladesh
An Operational Special Unit to Amend Biological Security M. Mollik1,2; 1Peoples Integrated Alliance, Dhaka, Bangladesh, 2PreSci. Trust
M. H. Richter, M-H. Lee, C. Herzog; Robert Koch Inst., Berlin, Germany. Funds, Phoenixville, PA.
As part of a national program to timely and efficiently respond to severe and Bangladesh is a low-lying, riparian country located in South Asia with a
extraordinary biological events a special operational unit has been estab- largely marshy jungle coastline of 710 kilometers on the northern littoral of
lished at the Center of Biological Security at the Robert Koch-Institute located the Bay of Bengal. Bangladesh’s alluvial soil is highly fertile but vulnerable
in Berlin, Germany. Among the events that can lead to an activation of the to flood and drought. Most of the people in Bangladesh are using ground
unit are highly unusual disease outbreaks, bio-accidents, or bio-terroristic water as a source of safe drinking water. The present investigation assesses
activities. The unit is also involved in preventive counter measures as in the the chemical and bacteriological quality of drinking water in several differ-
run up to large public events or state visits. Its unique modular concept al- ent locations of the Bogra Sadar upazila (sub-district) in Bangladesh. The
lows timely and most appropriate adjustments to mitigate severe biological samples were collected from supply water and tube-wells water, and ana-
threats or events. Such actions can span from theoretical support of federal lyzed for Arsenic (As), Chloride (Cl), Calcium (Ca), Iron (Fe), Lead (Pb), Mag-
units by phone up to active engagement at the site of the event, sample ac- nesium (Mg), pH, Phosphate (PO4), total coliform, total hardness, and total
quisition and on site diagnostics. The unit is therefore assembled of experi- viable bacteria. The Calcium (Ca), Magnesium (Mg), Phosphate (PO4), and
enced scientists in the field who have extensive know-how in handling and total hardness concentrations were found to be within the permissible limits.
collecting highly pathogenic agents in laboratory and field settings. A pivotal In contrast, 13% of the water samples contained Arsenic (As), 8% of the sam-
part is the integration of laboratories with reference character and close co- ples contained Chloride (Cl), 18% of the water samples contained Iron (Fe),
operation with forensic and crime fighting units. Developing sampling strate- and 5% of the water samples contained Lead (Pb) more than the permissible
gies that can increase the likelihood of acquiring a positive sample in limits. In addition, total coliform bacteria were found to be present in almost
contaminated areas are also part of the scientific project to improve the unit’s all samples of water ranging from 60%-100%. Total viable bacteria were
performance further. Also, developing and implementing of field diagnostics found in 69% samples which exceeded the permissible limits recommended
is a central part within the unit’s framework. Moreover, frequent training and by the World Health Organization. This investigation indicates that the water
consecutive skill, fit, and health tests of the unit leaders and staff assures used for drinking in many locations of the Bogra Sadar upazila (sub-district)
physical and psychological preparedness of the unit. Momentarily more top- is unsafe for human consumption due to both bacterial and chemical con-
ics are subsequently added to complement the unit’s modular structure. tamination. Tube-well water of this sub-district should be treated before using
Upon completion of its modular character, the unit will be able to react to a for drinking purposes.

BIODEFENSE PROGRAM & ABSTRACTS 45


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

075 (E) 077 (F)


Multi-User Validation of a Real-Time PCR System for Detecting Ethical & Social Issues Related to Synthetic Biology: What We
Bacillus anthracis Collected by Air Sampling Devices in a Liq- are Doing as Academicians?
uid Matrix S. T. Hakim, S. M. H. Tayyab, S. G. Nadeem; Jinnah Univ. for Women, Karachi,
U. K. Spaulding1, K. M. Clemens1, T. Hadfield2, V. Ryan2, S. Brunelle3, I. M. Pakistan.
Ota1; 1Idaho Technology, Inc., Salt Lake City, UT, 2MRIGlobal, Inc., Palm Bay, Synthetic Biology (SB) is the design and construction of new biological parts,
FL, 3AOAC International, Gaithersburg, MD. devices, and systems, and the re-design of existing, natural biological sys-
The RAZOR™ EX Anthrax Air Detection System, a real-time PCR system de- tems for useful purposes. Finding the right balance between formal statutory
veloped by Idaho Technology, Inc. (ITI), has undergone testing in accordance regulation and self-regulation of the scientific community remains a con-
with the harmonized Performance TestedSM Method (PTM) - Official Methods tentious issue and will occur only after the risks of synthetic biology are more
of Analysis (OMA) validation process established by AOAC INTERNATIONAL widely understood and debated. A questionnaire based cross sectional sur-
(AOAC). This system was developed for the US Department of Homeland Se- vey. Total 25 young researchers of post graduate level in Karachi were sur-
curity Bioinformatics and Assay Development 2 program which is directed at veyed at two Universities of the city. Main outcome measures was
establishing standards and third-party validation protocols to evaluate bio- perceptions on the basic principles of Synthetic Biology. All participants were
threat-detection tools. The RAZOR Anthrax System detects and identifies vir- females with mean age of 26+ 2 years, and enrolled for MS/ M.Phil. and Ph.D
ulent Bacillus anthracis spores present in samples from air collection devices dissertation. They responded that they were not aware of social and ethical
via the detection of three targets: 1) the pXO1 plasmid, 2) the pXO2 plasmid, issues related to synthetic biology before coming into the research field; even
and 3) B. anthracis chromosomal DNA. An “Anthrax Detected” result is ob- they never heard this term in this scenario before. A significant number of re-
tained when all three targets are detected. The validation process included spondents agreed that there is a need of creating awareness about benefits
evaluation by ITI, who is the method developer (MD), by MRIGlobal, an in- and risks related to synthetic biology. It was also recommended that we have
dependent laboratory (IL) and by twelve independent collaborators. Based on to educate the community of students and teachers, especially those who
the results of the MD and IL validations, a PTM certificate was conferred in Oc- are involved in research to take guidance, and approval from their institu-
tober, 2011. The work presented here describes the collaborative testing per- tional ethical review boards (if exist!) before start of any research study. Few
formed according to the Biothreat Agent Method guidelines established by more issues were also analyzed including use of laboratory animals, using
AOAC. Twelve independent collaborators, four at each of the three partici- clinical specimens without informing patients, sickness certification, human
pating MRIGlobal laboratories, participated in the study. A simulated liquid cloning, and disclosure of information to different parties without prior per-
air collection matrix was tested, spiked with B. anthracis at 2000 spores/mL mission of the subject and publication of sensitive research. Conclusion: A
or Bacillus cereus, a near neighbor, at 20,000 spores/mL. Each collaborator code of ethics and standards should emerge for biological engineering as it
tested a total of 24 randomized blind-coded samples, including 12 B. an- has done for other engineering disciplines in both research and academic in-
thracis-spiked samples and 12 B. cereus-spiked samples. Currently, the col- stitutions of the country and all the concerned should forced to follow the
laborative testing results are being reviewed. We expect that the RAZOR rules properly.
Anthrax System will meet the requirements for OMA status as its perform-
ance has been robust through all previous testing.
078 (F)
Identification and Molecular Characterization of Novel Group C
076 (F) Orthobunyaviruses Isolated from Mosquitoes Captured in Peru
A Hemorrhagic Fever Virus Sequence Database and Analysis L. L. Lofts, M. J. Turell, J. R. Kugelman, G. I. Koroleva, G. F. Palacios, C. A.
Platform Whitehouse; USAMRIID, Frederick, MD.
C. Kuiken; Los Alamos Natl. Lab., Los Alamos, NM. Members of the genus Orthobunyavirus (family Bunyaviridae) are seg-
Hemorrhagic fever viruses are a diverse set of over 80 viral species from nine mented, negative-sense, single-stranded RNA viruses that are responsible
different genera and five viral families: arena-, bunya-, flavi-, filo-, and to- for mild to severe disease in humans. As part of a long-term field study of ar-
gaviridae. All these viruses are highly variable and evolve rapidly, making bovirus ecology in the Amazon basin of Peru, more than 160 viral isolates
them elusive targets for the immune system and for vaccine and drug design. were identified in mosquitoes captured near Iquitos, Peru. Preliminary anti-
Presently about 50,000 HFV sequences have been published. A central web- gen detection analysis classified some of these Peruvian viral isolates as ei-
site that provides annotated sequences and analysis tools will be helpful to ther Group C or Bunyamwera group orthobunyaviruses. Entire deep
HCV scientists worldwide. The HFV sequence database collects and sequence sequence genome alignments confirmed the classification of the bun-
data, performs automated quality control and annotation, and provides them yamwera viruses; however, based on the S segment, the Peruvian viral iso-
to the public via a website that contains a user-friendly search interface and lates classified as Group C orthobunyaviruses failed to group with the Group
a large number of sequence analysis tools, following the model of the highly C viruses isolated from mosquitoes captured in Brazil. To further character-
regarded and widely used Los Alamos HIV database. Offering similar capa- ize the Group C orthobunyaviruses, deep sequence genomes were deter-
bilities for viruses from so many different taxonomical classes presents ad- mined for the Brazilian Group C mosquito viral isolates. The results provide
ditional challenges. Much of the database’s infrastructure depends on insights for the intra- and inter-reassortment among the orthobunyavirus
computer code that can place any sequence in the proper location relative to RNA segments (S, M, and L).
a central reference sequence. Because of the diversity of these viruses, a ref-
erence sequence needs to be chosen for every species. The NCBI RefSeq
database is used for this purpose; for sequences that are not assigned to a
genus or species, a reference sequence is chosen using a similarity search.
The sequences are internally aligned to a genus-level Hidden Markov align-
ment model using HMMer; the location of the gaps is also stored, and upon
retrieval gaps are inserted where necessary to maintain the alignment. The
HFV website can be accessed via http://hfv.lanl.gov.

46 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

079 (F) 081 (G)


Development of a High-Throughput Bioinformatics Typing Sys- Inhibition of Lipoteichoic Acid Synthesis in Staphylococcus au-
tem for the Phylogenomic Analysis of Infectious Category A reus and Bacillus anthracis
and B Agents S. Richter1,2, A. P. A. Hendrickx2, O. Schneewind2,1, D. Missiakas2,1; 1Howard
M. Eppinger, S. Agrawal, S. Daugherty, K. Galens, A. Maroo, L. Tallon, H. Tet- Taylor Ricketts Lab., Argonne, IL, 2Univ. of Chicago, Chicago, IL.
telin, C. Fraser-Liggett, J. Ravel; Inst. for Genome Sci. (IGS), Univ. of Mary- Staphylococci and many other Gram-positive bacteria incorporate zwitteri-
land, Sch. of Med., Baltimore, MD. onic lipoteichoic acid (LTA) into the cell wall. The polymer consists of a back-
Background: Infectious diseases outbreaks impose a significant public health bone of repeating glycerol-phosphate units decorated with D-alanine and a
threat and the understanding of the genomic plasticity of the underlying bac- glycolipid anchor. Membrane protein lipoteichoic acid synthase (LtaS) cat-
terial agents is crucial to the epidemiology, surveillance, diagnostic and foren- alyzes the biosynthesis of the polymer by transferring the polar head group
sic communities. We used the human pathogens Y. pestis and E. coli O157:H7 of phosphatidylglycerol to the growing chain of polyglycerophosphate. Mu-
as model systems both of which present with severe and potentially life- tations in S. aureus ltaS cause severe cell division defects and trigger cell
threatening complications in infected patients. Genomes were screened for death at 37°C. A two-step screening protocol was designed to identify small
mutations and structural variants to assess the genome polymorphisms and molecule compounds that inhibit growth of staphylococci by disrupting LTA
outbreak dynamics. Methods: Next generation whole-genome sequencing biosynthesis. High-throughput screening for growth inhibition was conducted
followed by phylogenomic polymorphism discovery analyses in the con- with chemical libraries. Strong hit compounds were further tested for their ef-
served genomic backbone as well as the dissected mobilome allowed track- fect on LTA biosynthesis at sub-lethal concentrations. One compound was
ing the pathogenome evolution. To investigate the phenotypic variations on identified as LTA inhibitor for staphylococci. The inhibitory activity was con-
the level of individual polymorphisms, we have developed a high-through- firmed with a heterologous system that utilizes S. aureus LtaS for synthesiz-
put bioinformatics pipeline for SNP discovery and validation taking into ac- ing LTA in E. coli. We also demonstrate that the compound is active against
count the coverage and quality of underlying sequence reads. The SNP Bacillus anthracis and Enterococcus faecalis.
pipeline is part of Ergatis, a web-based utility that is used to create, run, and
monitor reusable computational analysis pipelines. Results: Phylogenomic
analyses revealed numerous novel SNPs previously not available for the typ- 082 (G)
ing of these pathogen and other structural biomarkers many of which seem Discovery of Targeted and Broad Spectrum Antiviral Counter-
to be intimately associated with their niche-adaptation, pathogenesis and measures for Alphaviruses
outbreak assignment. Conclusions: The detected polymorphisms in strains D. Chung1, B. Beitzel2, J. Arterburn3, J. O. Tret1, J. Smith2, C. Schmaljohn2, G.
acquired during routine laboratory cultivation and within strains derived from Gene Olinger2, C. Scully2, C. B. Jonsson1; 1Univ. of Louisville, Louisville, KY,
a single outbreak suggest the need to study of outbreak populations rather 2
U.S. Army Med. Res. Inst. of Infectious Diseases, Fort Detrick, MD, 3New
than rely on single outbreak-type strains. The established high-resolution Mexico State Univ., Las Cruces, NM.
phylogenomic frameworks achieved refined a phylogenetic accuracy and res- Venezuelan (VEEV), eastern (EEEV), and western (WEEV) equine encephali-
olution and aids current molecular assays used in public health laboratories tis viruses are positive strand RNA viruses of the genus Alphavirus, family To-
in order to better prepare for future outbreaks and better risk assessment in gaviridae, that are endemic throughout the Americas. Naturally transmitted
response to novel emerging virulence phenotypes. by mosquitos from bird and rodent hosts, alphaviruses cause severe disease
and life-threatening encephalitis in humans and equines. Alphaviruses rep-
resent significant potential biological defense threats and are classified as
080 (G) Category B priority biodefense agents. There are limited therapeutics avail-
Humanization of Anti-Ricin Neutralizing Monoclonal Antibody able to treat diseases that they cause. Our primary efforts focus on identify-
W. Hu, J. Yin, L. Negrych, D. Chau, J. Cherwonogrodzky; Defence R & D, Med- ing small molecules targeting the alphaviruses by leveraging high throughput
icine Hat, Canada. and virtual screening. In our current pilot screening for VEEV, 384-well cell-
Ricin is regarded as a high terrorist risk for the public due to high toxicity based assays using a luciferase-reporter for V3526, and a CPE-based assay
and ease of production. Unfortunately, there is currently no therapeutic or for TC83 and/or VEEV 3000 were used to screen against the Prestwick Library
vaccine available against ricin. The murine monoclonal antibody D9 can and a targeted library of 658 active compounds identified as actives in a
strongly neutralize ricin and is a good candidate for the development of a screen for bluetongue virus. Tranilast was identified as an active compound
humanized antibody. Humanization of D9 variable domains was achieved by and prioritized for further characterization of structure-activity. A set of 14
a complementarity-determining region grafting approach. The humanized compounds based on an analysis of the functional groups and overall struc-
D9 variable domains were further grafted onto human heavy and light chain tural correlations to tranilast were made, but no improvement in potency was
constant domains to assemble the whole antibody gene. Meanwhile, a foot- observed. The drug diclofenac was also identified with a potent antiviral ac-
and-mouth-disease virus-derived 2A self-processing sequence was intro- tivity (EC50= 37 μM, SI50 >3). While the EC50 is high, diclofenac is a nons-
duced between heavy and light chain DNA sequences to express a teroidal anti-inflammatory drug (NSAID), and although its mechanism of
full-length antibody from a single open reading frame driven by a single action is not confirmed, it has been shown to block voltage-dependent
promoter in an adenoviral vector. After expression in mammalian cells and sodium channels and acid sensing channels along with its activity as a COX
purification, the humanized antibody was demonstrated to have equimolar inhibitor. Current plans include testing this compound in a mouse model chal-
expression of full-length antibody heavy and light chains and to retain anti- lenged with VEEV. In summary, we have screened more than 350,000 com-
gen-binding specificity and neutralizing capacity comparable to its parent pounds experimentally and over 1,300,000 compounds virtually, and are
D9. This humanized D9 has potential use for prophylactic or therapeutic continuing to analyze the efficacy of those showing promise.
purposes against ricin poisoning.

BIODEFENSE PROGRAM & ABSTRACTS 47


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

083 (G) 085 (H)


Generation of Biologically Active Nano-Aerosol by an Electro- Immunogenicity and Efficacy of a Recombinant Plague Vaccine
spray-Neutralization Method (rF1V) in Swiss Webster Mice
V. N. Morozov1, I. L. Kanev1, N. K. Balabaev1, A. V. Glyakina1, M. L. van Hoek2; P. Fellows1, J. Price1, W. Lin2, A. Rom2, S. Marshall3, L. Holland2, L. Wolfraim1;
1
RAS, Moscow, Russian Federation, 2George Mason Univ., Manassas, VA. 1
Dynport Vaccine Co., LLC, Frederick, MD, 2IIT Res. Inst., Chicago, IL, 3BioStat
Biological nano-aerosols promise tremendous opportunities in therapy of Solutions, Inc., Mount Airy, MD.
pneumonic lung infections (eg tularemia), lung diseases and in vaccination. Objective: The objective of this study was to determine minimum protective
However, generation of nano-aerosolized protein (e.g. antibodies or vaccines), titers in Swiss Webster mice vaccinated with recombinant F1V (rF1V) and sub-
DNA, antibiotics and other drugs meets serious problems caused by their sequently challenged by aerosol exposure to Yersinia pestis CO92 to support
fragility. We describe a simple method for manufacturing biological nano- vaccine licensure strategy under the Animal Rule. Methods: Groups of Swiss
aerosols under mild conditions. It is based on electro-hydrodynamic atomiza- Webster mice were vaccinated by intramuscular injection on Days 0, 14, and
tion of drug solutions followed by a gas-phase neutralization of a cloud of thus 28 with dosages of the rF1V vaccine ranging from 0.0032 to 16 μg in a 100 µL
generated highly charged nano-clusters and ions with a cloud of small oppo- volume. Control mice received saline using the same schedule and route of
sitely charged ethanol ions also generated by electrospraying. We demon- administration. Serum anti-rV, anti-rF1 and anti-rF1V titers were determined
strated that enzymes aerosolized by this mild technique retained their specific on Day 43 by Bridge ELISA. Mice were challenged on Day 44 by aerosol ex-
activity, which testifies that protein molecules are not notably damaged during posure to a lethal dosage of Y. pestis CO92. Results: A dose response curve
the process, despite the two highly energetic processes involved: electro- was obtained with respect to Bridge ELISA titers and probability of survival
spraying and gas-phase neutralization. To further evaluate possible damaging as well as vaccine dosage and probability of survival. Logistic regression mod-
factors in the technology we simulated process of gas-phase neutralization of eling shows a statistically significant relationship between increased levels
dry and hydrated multi-charged protein ions with small counter-ions (OH- and of anti-rV, anti-rF1 and anti-rF1V Bridge ELISA titers and increased probabil-
NO3-, both dry and hydrated). Our computer simulations have revealed that ity of survival (p<0.0001) with anti-rF1V titers the most highly associated
the hydration layer covering the surface of protein ion remarkably protects pro- with protection. Following challenge, the highest dosage of the vaccine
tein structure by channeling the impact energy to the water molecules. Thus, tested, 16 µg, elicited 87% protection. The 0.08 µg dosage of the rF1V vac-
while the impact of a small dehydrated single-charged counter-ion with a de- cine was the lowest dosage providing protection (25%) following challenge.
hydrated lysozyme macroion with 8 uncompensated charges resulted in a short All control mice died following challenge. There was also a statistically sig-
local heating of 20-25 protein atoms by more than 700 K, only 5 atoms were nificant relationship between vaccine dosage and probability of survival
heated in a similar impact of the same ion with a hydrated lysozyme macroion. (p<0.0001). The protective titer and protective vaccine dosage at 50% sur-
These studies suggest that proteins retain their structure and function upon vival were estimated at 225 U/mL of anti-rF1V and 2.32 µg of rF1V vaccine,
nano-aerosol generation and may be used in therapy or as inhaled vaccines. respectively. Conclusions: The rF1V vaccine, when administered in a three-
dose schedule, provides protection in a dose-related manner to Swiss Web-
ster mice following aerosol exposure to Y. pestis. Minimum protective titers
084 (H) were established and are an important component in bridging protective cor-
H5N1 Avian Influenza Specific Preventive Maintenance in the relates between species.
Russia
D. Surnev, Yu. Kuznetsov, T. Okovytaya, V. Yelnikov; JSK Pokrov Biological
Plant, Pokrov, Russian Federation.
086 (H)
The analysis of epizootic situation indicates a real threat of a wide spread of Ribosomal Protein TgP0 in the Invasion of Host Cells by Toxo-
avian influenza on the territories of different states. The main carriers of avian plasma gondii Tachyzoites
virus are wild migratory ducks. Avian influenza virus is reproduced in wild S. Rajagopal, S. Sharma; Tata Inst. of Fundamental Res., Mumbai, India.
ducks’s intestines and is excreted with excrements. As a result, the infection Apicomplexan parasites such as Plasmodium, Toxoplasma, Theileria,
spreads in water reservoirs, where from it can be contracted through domes- Neospora and Eimeria are highly successful parasites with a wide range of host
tic ducks and geese to other poultry. With poultry the avian influenza virus can specificities. A key event in the establishment of an infection is host cell inva-
progress as latent infection causing only decrease in productivity, as well as sion. Proteins on the parasite surface mediate interactions to enable host cell
devastating epizootic, accompanied by high death rate (up to 100 %). In the recognition and consequently, invasion. Earlier work from our lab, aimed at
Russian Federation the infection of avian influenza has not been registered for identifying immunoprotective antigens of the malarial parasite, established
a long time. Its outbreak occurred for the first time in Novosibirsk region in the ribosomal phosphoprotein PfP0 as a potential protective antigen. The can-
July, 2005. The results of molecular-biological studies have shown the agent didature of the ribosomal P0 protein as a protective protein has been subse-
belongs to high-virulent subtype H5N1. The disease was accompanied with quently validated by passive immunization and in vitro growth inhibition
high death rate, up to 100% in poultry. At the end of 2005 on the instructions studies. Antibodies to P0 blocked invasion of both Plasmodium falciparum as
of the Ministry of Agriculture of the Russian Federation the Russian scientists well as Toxoplasma gondii, and P0 protein was found to localize to the surface
developed the technology of vaccine production of H5N1subtype avian in- of the merozoites and tachyzoites, respectively. The ribosomal P0 protein lacks
fluenza vaccine. The preparations made on the basis of the virus strains, ho- any classical secretory signals for localization to the surface or secretory or-
mologous to epizootic possess the highest protective potential. Thus, a virus ganelles of the parasite. To identify such signals, we have generated truncated,
isolate isolated during the outbreak in Novosibirsk region as a vaccine mate- epitope-tagged constructs for transient transfection of Toxoplasma gondii. Ad-
rial was implemented. After successful carrying out of laboratory and field ditionally, post-translational modifications of the protein might also act as trig-
tests, inactivated emulsified vaccine against H5N1avian influenza made on the gers for differential localization during different stages of growth. We are
basis of strain homologous to the one spread in the Russian Federation, has characterizing post-translational modifications of parasite P0 by 2D gel elec-
been introduced into veterinary practice. Since 2006 avian influenza vaccine trophoresis and Mass Spectrometry. Specific antibodies to Toxoplasma gondii
vaccine against H5N1avian influenza has been widely and successfully used in P0 have been raised and these are being used in suitable invasion inhibition as-
the Russian Federation for specific preventive maintenance of the avian in- says to establish the point where P0 might mediate its role in the timeline of
fluenza in private farming and peasant household. invasive events. The results of these experiments will be discussed.

48 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

087 (H) 089 (H)


Advancing Clinical Grade RV10-2 Plague Vaccine Toward Licen- A Clinical Phase II Study Confirming the Safety and Immuno-
sure genicity of One or Two Doses of IMVAMUNE (MVA-BN) Small-
L. E. F. Quenee, N. A. Ciletti, D. Elli, T. M. Hermanas, O. Schneewind; Univ. of pox Vaccine in Vaccinia-Experienced Elderly Subjects
Chicago, Argonne, IL. R. N. Greenberg for the POX-MVA-024 Sites1, P. Chaplin2; 1Univ. of Kentucky
Yersinia pestis causes plague, a zoonotic disease with high mortality rates Sch. of Med., Lexington, KY, 2Bavarian Nordic GmbH, Martinsried, Germany.
that can be transmitted by fleabite or aerosol. A US Food and Drug Adminis- Background: IMVAMUNE® (MVA-BN®) is a live, highly attenuated Modified
tration (FDA)-licensed plague vaccine is currently not available. Vaccine de- Vaccinia Ankara virus vaccine formulated at a dose of 1 x 108 TCID50 / 0.5 ml.
velopers have focused on two subunits of Y. pestis: LcrV, a protein at the tip IMVAMUNE® does not replicate in human cells and is in clinical development
of type III secretion needles, and F1, the fraction 1 pilus antigen. The rV10-2 as a 3rd generation smallpox vaccine. Methods: A Phase II study was con-
vaccine is a non-toxigenic variant of LcrV lacking residues 271-300. We de- ducted to evaluate safety and immunogenicity (using both ELISA and PRNT)
veloped Current Good Manufacturing Practice (cGMP) protocols for rV10-2 of one and two doses of IMVAMUNE® smallpox vaccine in 56-80 year old vac-
and demonstrated its efficacy against lethal pneumonic plague challenge cinia-experienced subjects (n= 120). Subjects received either two injections
with wild-type or F1 mutant Y. pestis in mice, rats, guinea pigs, cynomolgus of 0.5 ml IMVAMUNE® or one injection of 0.5 ml Placebo and one injection
macaques and African Green monkeys. To further support FDA Investigational of IMVAMUNE® four weeks apart. Results: Vaccinations were well tolerated
New Drug (IND) authorization review and clinical testing for rV10-2, the vac- by all subjects. No serious adverse events related to IMVAMUNE® and no
cine was subjected to GLP long term stability testing. Clinical grade vaccine cases of myopericarditis were reported. The overall incidence of unsolicited
vials were stored under controlled temperature conditions and analyzed for AEs was similar in both groups. A second dose did not increase reactogenic-
stability over the course of 24 months. A GLP toxicology study in New Zealand ity. ELISA as well as PRNT results were comparable for the two groups
white rabbits was performed to determine preclinical safety in preparation
IMVAMUNE ELISA PRNT
for a future Phase I clinical trial.
Group N/Seroconversion Rate GMT/Seroconversion Rate GMT
(individual peak titers)
088 (H) 2 doses 61/90.2% 992.4/95.1% 257.6
Tularemia Vaccine Trials in Nonhuman Primates 1 dose 58/84.5% 645.2/77.6% 139.6
R. Tempel1, R. Shirley2, A. Legasse1, F. Heffron1, M. Axthelm1; 1Oregon Hlth. Response rates and seroconversion rates measured by PRNT increased after
& Sci. Univ., Portland, OR, 2Virogenomics, Inc., Portland, OR. a second dose. A second dose increases GMTs in the ELISA and to a higher ex-
Francisella tularensis is the gram-negative bacterial pathogen that causes tu- tent in the PRNT. Conclusions: One or two doses of IMVAMUNE® were safe
laremia, a debilitating and potentially fatal disease. Inhalation of as few as and immunogenic in the 56-80 years old vaccinia-experienced population.
ten bacteria can produce a severe pneumonic illness in humans. Despite its Safety, reactogenicity and immune responses are similar to that seen in the
high infectivity, classification as a category A select agent, and use as a bio- younger (18-55 year old) healthy population as investigated in other trials.
logical weapon, there is currently no FDA-approved vaccine against tularemia. The results indicate that in an emergency situation it is sufficient to vacci-
Our previous studies identified several Francisella genes that, when mutated, nate this population only once. Supported by Contract No. DMID-N01-AI-
give rise to attenuated strains in cell culture and mouse models of infection. 40072.
Deletion derivatives of two of these strains, ΔdsbB and ΔpdpB, in Francisella
Schu S4 provided protection against wild-type challenge in mice, indicating
the potential of these genes to be included in a live attenuated vaccine. Here, 090 (H)
we describe testing these strains in the nonhuman primate (NHP), rhesus
macaque. Groups of three NHPs were inoculated subcutaneously with ΔdsbB LC16m8, an Attenuated Smallpox Vaccine Will Be a Useful
(108 CFU) or ΔpdpB (109 CFU). Four weeks after the primary inoculation, the Countermeasure against Bio-Terrorism with Smallpox
NHPs were boosted with 1010 CFU of the respective attenuated strain. Thirty- H. Yokote1, Y. Shinmura1, C. Uemura1, T. Kanehara1, S. Maruno1, H. Matsui1,
three days post-boost, 104 CFU of wild-type Schu S4 were inoculated into the K. Ohkuma1, K. Yokoi1, A. Funatsu1, S. Gordon2, G. Franchini2, S. Hashizume3;
1
lung. Two NHPs that received only vehicle during the primary and boost in- Kaketsuken, Kumamoto, Japan, 2Natl. Cancer Inst., Bethesda, MD, 3Univ. of
oculations were concurrently challenged with 104 CFU of wild-type Schu S4. Chiba, Chiba, Japan.
The challenge dose was chosen based on our results that intrabronchial in- Background: During the current situation where numbers of patients with
oculation of 104 CFU resulted in tularemia-related death nine and 18 days some sort of immune impairment such as eczema, HIV-positive, etc. are in-
after infection. In contrast, naïve NHPs infected with 100 CFU survived until creasing, safer smallpox vaccines than the conventional ones are being de-
the end of the study (day 23). During the study, blood, lymph node, bone sired for a broad range of people including such immune impairment patients
marrow, and bronchoalveolar lavage samples were obtained periodically to in case of bioterror attacks with smallpox. LC16m8 is an attenuated licensed
determine the bacterial load, and O2 saturation, CBC panels, enzymatic func- smallpox vaccine and has been stockpiled in Japan. LC16m8 has demon-
tion, and protein levels were monitored to assess the overall health of the strated a good protective efficacy in healthy animal models and has given so
NHP. We will report these results, along with the gross pathology of the or- far to approximately 100,000 healthy people without any severe adverse
gans after the NHP has succumbed to disease or upon necropsy at the end events. We have further conducted a research into how LC16m8 behaves
of the study (28 days post-challenge). While there have been several studies under the immune impairment using immunocompromised animal models.
describing protective strains of Francisella in mice, these are the first such Methods: To investigate the immunologic basis of containment of vaccinia in
studies in a NHP model of infection. the vaccination site or dissemination from the skin lesions to distal sites,
macaques were made depleted systemically of T- or B-cells and vaccinated
with either LC16m8 or Dryvax. To evaluate the protective efficacy in the im-
munocompromised hosts, LC16m8 was vaccinated to MHC class I-KO and
MHC class II-KO mice. The mice were challenged with a lethal dose of vac-
cinia WR at 2 or 3 weeks post vaccination and observed for 14 days. Results:
B-cell depletion did not affect the size of skin lesions induced by either vac-

BIODEFENSE PROGRAM & ABSTRACTS 49


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

cine. However, while depletion of both CD4+ and CD8+ T-cells made no ad- on Days 0 and 30. Controls were given phosphate buffered saline only (PBS)
verse effects and systemic dissemination on LC16m8-vaccinated animals, it on the same vaccination regimen. All animals were aerosol challenged with
caused disseminated vaccinia in the spleen or cerebrum of macaques im- a target of 50 LD50 Y. pestis CO92 70 days after initial vaccination. Results:
munized with Dryvax. No viral DNA was detected in the other major organs Controls succumbed to Y. pestis within 9 days post exposure, as did all ani-
(e.g. heart, genitals) of vaccinated animals. A single dose of LC16m8 protects mals in the 0.5 μg group (by 5 days post exposure). 70% of animals in the 8
both MHC class I-KO and MHC class II-KO mice from a lethal WR challenge. μg group survived, whereas 20% of animals in groups 2 μg and 0.125 μg sur-
Conclusions: The study showed that LC16m8 is well-tolerated and effective vived. Y. pestis was cultured (controls and vaccinated) in the blood, lungs,
in the immunocompromised hosts. It is suggested that LC16m8 can be a po- spleens, livers, brains, and lymph nodes from those animals that succumbed
tent candidate smallpox vaccine for immune impairment patients. to infection. Vaccinated animals that survived Y. pestis challenge had no de-
tectable tissue burden. Pathology showed minimal lung alterations in three
of four CM that survived challenge, indicating strong protection. The other
091 (H) animal had localized residual fibrosis indicating vaccination was effective in
Development of a Guinea Pig Passive Transfer Efficacy Model limiting infection. Survival correlated with a higher dose of vaccine as well
Using Immunoglobulin Derived from Clinical Trial Volunteers as higher serum IgG anti-F1 and -V levels measured by Protein G ELISA and
Vaccinated with Recombinant Botulinum Vaccine A/B maintained lymphocyte and monocyte counts. Conclusions: These data sug-
gest that an optimal dosage of vaccine is required to induce optimal serum
S. Martin1, W. Swiderski1, K. Metcalfe1, N. Niemuth2, M. Vassar2, J. Shearer1;
1 IgG levels of anti-F1 and V for protection against Y. pestis challenge.
DynPort Vaccine Company, Frederick, MD, 2Battelle BioMed. Res. Ctr.,
Columbus, OH.
Purpose: This study was designed to characterize a guinea pig model for eval- 093 (H)
uation of passive protection against inhalational exposure to botulinum neu-
rotoxin complex (BoNT) serotypes A1 (BoNT/A1) and B1 (BoNT/B1). Generation of Protective Immunity against Fatal Rickettsial
Methods: Naïve guinea pigs were exposed to a range of dosages of Disease
aerosolized BoNT/A1 or BoNT/B1 to determine the dosage lethal to 50% of S. P. Riley1,2, M. M. Cardwell1,2, R. Hillman1,2, L. Quenee1,2, J. J. Martinez1,2;
1
animals (LD50) and to evaluate the progression of disease. Mortality, median Univ. of Chicago, Chicago, IL, 2The Howard Taylor Ricketts Lab., Lemont, IL.
time-to-death (MTTD), onset and duration of clinical signs of illness associ- Pathogenic rickettsiae are the causative agents of Rocky Mountain Spotted
ated with challenge for both BoNT serotypes were evaluated. For passive Fever (RMSF), Mediterranean Spotted Fever (MSF), typhus and other human
transfer, immunoglobulin was purified from the sera of rBV A/B-vaccinated diseases with high morbidity and mortality. Previous reports have demon-
clinical volunteers and the challenge dosage required to produce <80% sur- strated that survivors of rickettsial disease are immune to subsequent in-
vival for each BoNT serotype was determined using a stage-wise approach. fections; however, the molecular basis of this immunity was not well defined.
At 24 hours after passive transfer, guinea pigs were aerosol challenged with We have recently demonstrated that purified recombinant outer membrane
BoNT/A1 or BoNT/B1 and observed for clinical signs of botulism and mor- protein B (rOmpB) is sufficient to elicit protective humoral immune re-
tality for 14 days. Results: The guinea pig aerosol LD50 for BoNT/Al was es- sponses against lethal disease in a murine model of MSF mediated by Rick-
timated to be 158 mouse intraperitoneal LD50 (MIPLD50) and estimated to be ettsia conorii. This study also revealed that protective immunity was
<221 MIPLD50 for BoNT/B1. Increases in mortality and decreases in MTTD correlated with the ability of generated antibodies to facilitate complement-
following exposure to either BoNT were inversely related to dosage. Changes mediated elimination of rickettsiae from blood and plasma. The evolution-
in breathing was the first sign of disease observed following challenge. The ary and amino acid conservation of rOmpB among closely and distantly
MTTD following onset of the first clinical sign was dosage related for both related rickettsial species suggests that this protein may be sufficient to
BoNT. Passively immunized guinea pigs were protected against inhaled serve as a protective antigen in other rickettsial disease models. To test this
dosages 700 times the guinea pig aerosol LD50 of BoNT/A1 and BoNT/B1. hypothesis, we initially developed a C3H/HeN murine model of fatal Rocky
Conclusions: Guinea pigs passively immunized with human antibodies were Mountain Spotted Fever (RMSF) mediated by the CDC-classified Select
protected against aerosol challenge levels several orders of magnitude above Agent, Rickettsia rickettsii, and have demonstrated that the progression to
the aerosol LD50 for each BoNT serotype. The guinea pig model is suitable disease and pathologic injuries to the spleen, liver, heart and lungs closely
for evaluation of the effectiveness of antibodies induced by rBV A/B vacci- mimic those observed in fatal R. conorii infections. We are currently utilizing
nation. this model to test the efficacy of recombinant purified R. rickettsii rOmpB
and other conserved rickettsial antigens in the generation of protective im-
munity against fatal disease. We will also determine whether complement-
092 (H) mediated killing also plays a crucial role in protection against fatal R.
Increased Dose of Recombinant F1/V Plague Vaccine Results in rickettsii infections. With the threat of the potential use of rickettsial species
Increased Survival in Plague-Challenged Cynomologus as agents for bioterrorism, understanding the complex interplay between
pathogen and host is crucial for the development of novel antimicrobials
Macaques
and more efficacious therapies against fatal diseases.
K. A. Overheim, S. Lemoine, K. Gallegos, F. Romero, I. Fisher, A. Monier, C.
Schneider, M. W. Valderas, J. Wilder, R. Russell, R. L. Sherwood, R. LeClaire;
Lovelace Respiratory Res. Inst., Albuquerque, NM.
Background: Yersinia pestis is the causative agent of plague and can mani-
fest as pneumonic or bubonic forms. Previously we tested a recombinant
plague protein F1/V vaccine in Cynomolgus macaques (CM) at 10 μg of vac-
cine vaccinated on Day 0 and 30. All vaccinated CM survived a challenge with
50 LD50 of Y. pestis CO92. In this study we chose to determine if reducing the
dose of the recombinant protein F1/V vaccine would provide adequate pro-
tection in CM, as well as to further determine the correlates of protection.
Methods: Groups of NHP (6 animals per group) were vaccinated intramus-
cularly with varying doses of the recombinant vaccine (0.125, 0.5, 2, or 8 μg)

50 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

clusion: PMNs are not effective killers of BA spores but they efficiently kill
094 (I) vegetative bacilli. While oxidative mechanisms do not appear to play an im-
Determination of Efficacious Praziquantel Dose in Different portant role in killing BA, PMN autophagy may be an important mechanism
Mouse Strains: BALB/C and Swiss Mice for Treatment of Schis- to eliminate BA.
tosoma mansoni
M. P. M. N. NA1, D. Y. S. N. yole2; 1Mombasa Polytechnique Univ. Coll., Nairobi,
Kenya, 2Kenya Polytechnique Univ. Coll., Nairobi, Kenya.
096 (I)
Background: The drug Praziquantel has been in use for treatment of schis- Burkholderia pseudomallei Induces IL-1beta and IL-6 Secretion
tosomiasis however the effective dose that can clear the worms completely via Activation of MAPK and STAT3 in Differentiated THP-1 Cells
has not been determined. Mouse models have been used in this study to de- M-H. Cho1, Y-W. Shin1,2, J-H. Chun1, K-J. Hong1, G-e. Rhie1; 1Korea CDC, Chung-
termine effective Praziquantel dosage.This work therefore sought to deter- buk, Korea, Republic of, 2Yonsei Univ., Seoul, Korea, Republic of.
mine the effective dose of praziquantel in different mouse strains, BALB/c Burkholderia pseudomallei is a bacterial pathogen that causes a broad spec-
and Swiss mice. Methods: Six mice were divided into four groups namely trum of clinical symptoms collectively known as melioidosis. This study aims
PZQ1350, PZQ900, PZQ450 and infected control. Mice were infected at week to investigate the underlying mechanism of inflammatory responses elicited
0 and at week 4 all the experimental groups (ie PZQ1350, PZQ 900 and by B. pseudomallei in the human monocytic cell line THP-1, which was dif-
PZQ450 mg/kg bw (Farah et al, 2000), respectively) were treated using vary- ferentiated into macrophages with phorbol 12-myristate 13-acetate (PMA). B.
ing praziquantel dosages. At week 6 all mice were perfused to recover adult pseudomallei infection on differentiated THP-1 cells led to the specific ex-
worms. Gross pathology and histopathology of the liver tissue were ob- tracellular release of interleukin (IL)-1β and IL-6 in a dose-dependent manner
served. Results: More worms were recovered in infected control and there as shown by ELISA. Also, we confirmed that B. pseudomallei-induced IL-1β
was less reduction of worm burden was observed in this group p<0.001. and IL-6 production occurs via activation of signal transducer and activator of
Gross pathology was severe and more granulomas were also recorded in this transcription-3 (STAT3) by Western blot analysis. Moreover, a STAT3 inhibitor
group. In the experimental groups PZQ1350 had few numbers of worms re- (static) inhibited B. pseudomallei mediated IL-1β and IL-6 production. p38,
covered and a high worm reduction P<0.001. Also no granulomas were ERK1/2 and JNK phosphorylation, modulate IL-1β and IL-6 secretion follow-
recorded in this group due to the effect of drug and humoral responses in ing infection of B. pseudomallei on THP-1 cells. Also, B. pseudomallei induced
clearing the worms. This was followed by PZQ900 but PZQ450 which is the IL-1β and IL-6 secretion was reduced in the presence of the inhibitors of p38-
standard dosage used indicated more worm recovery and less reduction in MAPK (SB203580), ERK1/2 (PD98059) and JNK (SP600125). These findings
worm burden. Conclusion: The findings imply that the higher the dosage the provide the key role of STAT3 and MAPK kinase pathways in IL-1β and IL-6
more the protection against Schistosoma mansoni infection; PZQ1350 indi- production in response to inflammation induced by B. pseudomallei.
cated better responses both in worm recovery, worm reduction, and pathol-
ogy compared to other groups. The findings also indicate that Swiss mouse
was a better permissive host than BALB/c as it allowed more parasites to ma- 097 (I)
ture instead of destroying them.Further experiments could be done,with de- Increased Protection of African Green Monkeys Vaccinated with
termination of humoral and cellular responses.
Increasing Doses of a Recombinant F1/V Plague Vaccine
K. A. Overheim1, S. Lemoine1, K. Gallegos1, K. Brooks1, I. Fisher1, A. Monier1,
095 (I) C. Schneider1, M. W. Valderas1, J. Wilder1, R. Russell1, R. L. Sherwood1, R.
LeClaire2; 1Lovelace Respiratory Res. Inst., Los Lunas, NM, 2Lovelace Respi-
A Novel Mechanism by which Human Neutrophils Kill Bacillus ratory Res. Inst., Albuquerque, NM.
anthracis Background: Yersinia pestis is the causative agent of plague and can mani-
G. Ramachandran1, L. Baillie2, P. Tsai3, G. Rosen3, A. Cross1; 1Univ. of Maryland fest as pneumonic or bubonic forms. Previously we tested a recombinant pro-
Med. Sch., Baltimore, MD, 2Welsh Sch. of Pharmacy, Cardiff, United Kingdom, tein vaccine (F1/V in alum) in African green monkeys (AGM) using 10 μg of
3
Dept. of Pharmaceutical Sci., Baltimore, MD. vaccine administered intramuscularly (i.m.) on Days 0 and 30. All vaccinated
Background: Bacillus anthracis (BA), the causative agent of anthrax, is ac- AGMs, with the exception of one animal, succumbed to challenge with 50
quired by mammalian hosts from the environment as spores that are able to LD50 of Y. pestis CO92. Therefore, we decided to determine a dose of vaccine
survive harsh conditions. BA spores must germinate inside host cells to veg- that would confer protection to AGM recombinant protein F1/V vaccine, as
etative bacilli before they can express the virulence factors that enable them well as determine the correlates of protection. Methods: Groups of NHP (6 an-
to evade host defense and disseminate. While the role of macrophages and imals per group) were vaccinated i.m. with varying doses of the recombinant
dendritic cells in this initial interaction is well established, the role of poly- vaccine (100 μg, 25 μg, 6 μg, 1.5 μg) on Day 0, 28, and Day 56. Controls were
morphonuclear leukocytes (PMNs) is not well defined. We now test if BA 34F2 given phosphate buffered saline only (PBS) on the same vaccination regi-
Sterne spores and/or vegetative bacilli are killed by human PMNs in vitro and men. All animals were aerosol challenged with 25 LD50 Y. pestis CO92 70 days
also determine the mechanism of killing. Results: At an MOI of 1:1 PMNs are after initial vaccination. Results: All but one control succumbed to Y. pestis
unable to kill 34F2 Sterne or the germination deficient ΔgerH spores. How- infection within 6 days post exposure. The low dose vaccinated group (1.5
ever at the same MOI, PMNs killed over 80 % of vegetative bacilli within 20 μg) all succumbed by Day 5 post exposure. 70%, 50% and 33% of animals in
min even in the absence of opsonins. In the absence of PMNs BA bacilli are the 100 μg, 20 μg, and 6 μg groups survived to 21 days post exposure. Y.
extremely susceptible to killing by superoxide generated by epigallocatechin pestis was cultured from control AGM and vaccinated AGM that succumbed
gallate. Human PMNs generate low levels of superoxide (or nitric oxide) only to disease from their blood, lungs, spleens, livers, brains, and lymph nodes.
in the presence of BA at a MOI of ≥ 1: 100. Lysates of human PMNs (106/ml) Increased doses of vaccine roughly correlated with increased serum anti-F1
were unable to kill 106 BA, but very high doses (10-100 ug/ml) of antimicro- and anti- rV antibody titers. Conclusions: These data suggest that an opti-
bial peptide, α-defensin, were required to kill BA bacilli. By using an inhibitor mal serum IgG level to r-V and r-F1 are needed in AGM to confer protection
of autophagy, 3MA, we identified autophagy as a mechanism by which PMNs against Y. pestis challenge.
killed BA. In the presence of 3MA, there was over 50 % reduction in killing of
BA by PMNs. Moreover, formation of LC3 puncta, an autophagy marker, dra-
matically increased in the presence of BA within 20 min of incubation. Con-

BIODEFENSE PROGRAM & ABSTRACTS 51


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

placed at nine locations in the lab, and subjected to formaldehyde of 1.7 g/m3
098 (I) at >80% humidity at 22±2°C for 16h. Extraction media was EMEM with 10%
Ebola Virus Secreted Glycoprotein Can Function as a Decoy for FBS. Titrations were by ten-fold dilutions and eight replicates per dilution,
Antibodies using PK15 cells and immunoperoxidase staining, and controls and reduc-
G. S. Mohan, Y. Chinglai, L. Ye, R. Compans; Emory Univ., Atlanta, GA. tion factors according to (1). No PPV could be recovered after fumigation,
Through an RNA editing mechanism, Ebola Virus makes two isoforms of its sur- with a residual titre of ≤-0.27 log10 TCID50 disc-1 according to (2). A PPV re-
face glycoprotein (GP). The membrane-bound isoform (mGP) is the primary duction factor of ≥7.0±0.37 proved the fumigation process efficient, and the
viral mediator of host cell attachment and entry. However, ~80% of GP tran- controls showed that the assay was reliable (Table 1)
scripts encode a nonstructural secreted GP (sGP) whose function has been Table 1. Titres in TCID50a log10 ±SDbdisc-1 (recovery control) or 50 μl-1 (stop/in-
widely debated. Here, we demonstrate that sGP can serve as a decoy for anti- terference controls)
EBOV antibodies. In mice immunized with the wild-type GP construct, sGP was
Controls Extracted EMEM 10% FBS EMEM Reduction
able to compete with mGP for antibody binding. Surprisingly, in mice immu-
steel disc
nized with mutant constructs expressing solely mGP, sGP was unable to com-
pete with mGP, even at 16-fold molar excess. In these mice, sGP also had no Recovery controlc 6.7±0.37 n.a. n.a. n.a.
effect on antibody-dependent pseudovirus neutralization, while sGP inhibited Stop control 1.5±0.45 1.4±0.25 n.a. -0.12±0.51
neutralization in wild-type GP-immunized mice. Together, these data suggest
Interference control 3.1±0.44 n.a. 3.1±0.36 0±0.57
that during natural infection, an excess of sGP focuses the host immune re-
sponse against epitopes shared with mGP, allowing sGP to absorb anti-EBOV Cytotoxicity control No cytotoxicity n.a. No cytotoxicity n.a.
antibodies. Interestingly, serum from mice primed against sGP and then n.a. = not applicable; atissue culture infectious dose; bstandard deviation;
boosted with mGP still cross-reacts with sGP, suggesting that there may be a c
after 16h at 22±2°C
component of original antigenic sin at play. This is particularly germane to Ebola This work was part of a joint project between the Swedish Defence Research
vaccinology, given the implication that vaccination of previously-exposed indi- Agency and the National Veterinary Institute, concerning various decontam-
viduals may simply boost an already non-protective immune response. ination strategies of high risk pathogens in the community, financed by the
Swedish Civil Contingencies Agency. Future experiments will include high
099 (J) pathogenic avian influenza and spores of B. antracis. References: (1) Guide-
line of “Deutsche Vereinigung zur Bekämpfung der Viruskrankheiten e.V.“
Bioactivity of an Indigenous Plant Used in the Treatment of and Robert Koch Institute for testing the virucidal efficacy of chemical disin-
Malaria against Anopheles Mosquito Larvae in South West fectants in the human medical area” (2) Blumel, J., I. Schmidt, H. Willkom-
Nigeria men, and J. Lower. 2002. Inactivation of parvovirus B19 during pasteurization
F. O. Omoya, B. E. Boboye; Federal Univ. of Technology, Akure, Nigeria. of human serum albumin. Transfusion 42:1011-1018.
Malaria has been one of the major sources of human death and it causes sig-
nificant economic losses in Africa. Even in developed countries, such as Eu-
rope and United States, re-emergency of this disease has been reported. 101 (J)
Plasmodium spp— the etiologic agent—has shown resistance to common Bactericidal Activity of Antimicrobial Peptides Immobilized on
and cheapest drugs for the treatment and prevention of malaria. In other to Planar Surfaces
eradicate this disease, different control measures have since been exploited S. Arcidiacono, J. Uzarski, L. Doherty, W. Muller, C. Mello; U.S. Army Natick
by man. Control of Anopheles mosquito, the vector, is one of the ways our Soldier Ctr., Natick, MA.
research group is focusing on as a mean of controlling the disease. This re- Background: Substrates with antimicrobial functionality have utility in med-
search work aimed at the use of different leaf extracts of the medicinal plant; ical applications, food safety, decontamination, and prevention of materials
Vernonia amygdalina on the second and fourth instars larvae of Anopheles degradation. Most surface activity measurements are made in solution (e.g.
arabiensis for a period of 24 hours in-vitro and the phytochemical screening growth medium or buffer). However, some applications occur in which the
of the plant. Three different leaf extracts namely; water, ethanol and methanol antimicrobial compound needs to retain functionality in a non-solution state,
extracts showed different percentage mortality on the tested larvae. The high- (i.e. in a range of water content or humidity). To examine the effect of hu-
est mortality was recorded in ethanol treated larvae. The lethal concentra- midity on activity, bacteria were exposed to antimicrobial peptides (AMPs)
tion at LC50 and LC75 also varied with different extracts and instrars. In the attached a planar surface and incubated under varying relative humidity.
fourth instars larvae treatments; LC50 (15.43mg/mL) and LC75 (46.95mg/mL); Methods: Cysteine variants of Cecropin P1 and SMAP29 were attached to
LC50 (12.54mg/mL) and LC75 (16.64mg/mL); and LC50 (2.51mg/mL) and LC75 gold via self-assembled monolayers consisting of mixed oligo(ethylene gly-
(5.45mg/mL) were recorded in water, methanol and ethanol extracts respec- col) containing alkane thiols, functionalized with a sulfo-GMBS cross-linker.
tively. Phytochemical screening of this plant revealed the presence of some Surface antimicrobial activity was determined against cells in the presence of
chemicals that are insecticidal such as Tannins and Alkaloids. Summarily, our solution as well as cells dried on the surface and incubated at constant hu-
finding revealed that Vernonia amygdalina might be considered as a larvici- midity. Cells were also observed microscopically with LIVE-DEAD fluorescent
dal agent in the control of Anopheles mosquito larvae. stain to assess activity. Results: Immobilized SMAP29 was active against the
Gram-negative bacteria Escherichia coli and Acinetobacter baumannii in the
presence of solution. Fluorescent microscopy confirmed that the surface was
100 (J) active. However, no activity was observed against the Gram-positive bac-
Formaldehyde Disinfection Using Porcine Parvovirus as Indicator terium Staphylococcus aureus. Surfaces containing immobilized Cecropin P1
E. Emmoth, U. A. Bengtsson, I. Dergel, C. Hultén; Natl. Vet. Inst., Uppsala, were not active. Activity was evaluated against cells dried on a surface and
Sweden. incubated under constant humidity. Suitable incubation conditions allowing
We used porcine parvovirus (PPV) to validate a formaldehyde fumigation cells to retain viability during drying continues to be investigated. Conclu-
process of a biosafety level 3 laboratory of 190 m3 volume. PPV strain 893/76, sions: Covalent attachment of SMAP29 imparted antimicrobial functionality
in Eagle’s minimal essential medium (EMEM) with 1% foetal bovine serum to gold surfaces. The research presented here lays the foundation for a new
(FBS) was dried in 50 μl volumes on stainless steel discs. The discs were generation of non-leaching antimicrobial treatments. As a potential alterna-

52 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N DAY A B S T R AC TS

tive to antimicrobial agents currently in use, AMPs are not expected to pro-
mote emergence of resistant microorganisms.
103 (K)
Preparing Biodefense Professionals: MS in Biotechnology
Concentration in Biodefense and Certificate in National
102 (J) Security Studies
Investigating the Dispersion and Decontamination of Anthrax K. M. Obom1, A. I. Roth2, P. J. Cummings3; 1Johns Hopkins Univ., Rockville,
Spores Within Indoor Work Areas MD, 2Johns Hopkins Univ., Washington, DC, 3Johns Hopkins Univ., Baltimore,
A. Clohessey, S. Hoque; Rowan Univ., Glassboro, NJ. MD.
The threat of biological agents used as weapons on the civilian population To respond effectively to a biological threat, the nation will need a well trained
has become a very real concern, especially after the anthrax-based attacks workforce. Currently, there is a shortage of biodefense professionals, partic-
between October 4 and November 2, 2001. This study is aimed at gathering ularly individuals that have a strong background in the science and policy re-
information on the dispersion of particles in an office space and the subse- lated to biodefense issues. To prepare professionals who have this expertise,
quent decontamination of that area. The goal of this investigation is to un- students will be able to pursue a Certificate in National Security Studies and
cover answers to questions that first-responders would have to face in a Master of Science in Biotechnology with a concentration in Biodefense at
situations where an infectious agent has been released in a building. Here, Johns Hopkins University (JHU). This unique partnership between JHU’s Na-
furniture presence is investigated in the effects it has on particle dispersion. tional Security Studies and the Center for Biotechnology Education programs
Computational fluid dynamics software using LES method is used to simu- and the United States Army Medical and Research Institute of Infectious Dis-
lated particle releases in a modeled office space incorporating furniture. ease (USAMRIID) will provide students with a curriculum that includes grad-
100,000 Bacillus anthracis spores are released from 3 locations and the re- uate course offerings in Bioinformatics; Biothreat Response and Microbial
sults are compared. Areas where flow is more stagnant allow particles to re- Forensics; Science, Medicine & Policy in Biodefense; Vaccinology; Radiation
main within the room longer and disperse throughout. The timeframe of this Biology; and Biodefense Lab Methods, as well as Preserving American Se-
investigation has spanned September, 2011 until the present. Future goals curity; Crisis Management and The Art and Practice of Intelligence. As part of
aim at incorporating ClO2 disinfection agent into the room. The required con- the curriculum, USAMRIID offers a competitive fellowship program for up to
centration to decontaminate as well as time required to reach safe levels of five students each year to work at USAMRIID, while completing graduate
ClO2 will be investigated. courses at JHU’s campuses in Baltimore or Rockville, MD and Washington
DC. Graduates of the degree program and the certificate will have an in-depth
knowledge of the policy and practical laboratory skills in the field of biode-
fense and will fill a growing need for trained professionals skilled in this es-
sential discipline within the science community and for homeland security.

104 (K)
Recruiting First Responders from the Dental Profession
R. J. Boylan, D. L. Glotzer; NYU Coll. of Dentistry, New York, NY.
Significant advances in our nation’s ability to detect environmentally-dis-
seminated bioweapons have been made over the past ten years. Manipula-
tion of these bioweapons in the laboratory has given us much information
about potential mutants that could be produced as well as more effective
ways to treat them. However, a recent report from the bipartisan WMD Ter-
rorism Research Center states that “the primary means of defending the
American Homeland against bioterrorism is the capability to effectively re-
spond after an attack has occurred.” Our communities’ first responders will
have the primary responsibility to respond to a major disaster. However, in
the event of a massive attack employing bioweapons, it may be necessary to
call upon other healthcare personnel to participate. Experience has shown
that one critical area that needs to be considered in catastrophe response is
the available numbers of trained individuals in the public health workforce
that can, and will respond when necessary. Healthcare workers who are
knowledgeable about the potential agents and their properties, modes of
transmission and means of personal protection, as well as the most effective
therapies, are more likely to engage effectively and safely in such a response
than those who are not. Since shortly after 9/11 NYU’s Dental School made
it a priority to educate its students in such matters throughout its 4-year cur-
riculum. They are provided with extensive information about the bioweapons
in the CDC’s categories A and B in a two hour conference in Infectious Dis-
eases; given a comprehensive 12 hour course in Bioterrorism Preparedness
in which the AMA’s Core Disaster Life Support program is presented; and en-
couraged to prepare a plan to protect themselves, their staff and patients in
the event of a disaster. If HR 570, The Dental Emergency Responder Act
(which passed the House this year) passes the Senate, then dentists by Fed-
eral regulation will be considered emergency responders. The model for ed-
ucating dental students in these areas, which will be described more

BIODEFENSE PROGRAM & ABSTRACTS 53


M O N D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

thoroughly at the meeting, will potentially provide a larger pool of health care
workers willing to be effective agents, contributing importantly to a
107 (K)
bioweapons attack. Biosafety and Biosecurity: The Case of a Third World Country
and the One Health Spectrum
D. B. Ndumu1, R. Heckert2, C. S. Rutebarika1, R. W. Kaboyo3, M. Busuulwa4, P.
105 (K) Atimnedi5, S. Balinandi6, P. Delarosa7; 1Ministry of Agriculture, Animal In-
Tungiasis (Jigger Infestation) in Rural Kenya: An Emerging In- dustry and Fisheries, Entebbe, Uganda, 2Robert Heckert Consulting LLC,
fectious Disease Bowie, MD, 3Ministry of Hlth., Kampala, Uganda, 4AFENET, Kampala, Uganda,
5
N. N. N. N. Ngomi, Jr.; Jomokenyatta Univ. Kenya, Nairobi, Kenya. Uganda Wildlife Authority, Kampala, Uganda, 6Uganda Virus Res. Inst., En-
Objective: To describe the prevalence of tungiasis (jigger flea infestation) and tebbe, Uganda, 7Booz Allen Hamilton, Middletown, DE.
associated risk factors in a sentinel group (children 5-12 years of age) in rural The silent ‘bioterror war’ on mankind in the Third World is attributed to epi-
Central Kenya. Methods: A cross-sectional study was carried out in Murang’a demics and emerging infectious diseases. These developing countries are
South district during high transmission season (dry season, August - Sep- the front line of defense against the devastation that would arise from pan-
tember 2009). A total of 385 randomly selected households were visited. Chil- demics or deliberate release of a pathogen. While disease outbreaks are also
dren were examined for presence of tungiasis, and a questionnaire was linked to sociopolitical problems, integrated plans that prepare such coun-
administered to collect demographic, behavioral and environmental data. tries to deal with pandemics and possible bioterror attacks have not been
Results: Prevalence of tungiasis was 57% (218/385; 95% CI=51.7%-61.6%). devised. Recent efforts rely on a reporting system through the World Organ-
Itching (89.1%) was the most common associated symptom, followed by pain ization for Animal Health. Although important; this reporting alone is a frag-
upon pressure (67.3%), sleep disturbance (58.2%) and walking difficulties ile early warning system, as it relies on a single response generated after
(53%). In multivariate logistic regression analysis the following independent confirmation of an outbreak in as far as biodefence is concerned. Since many
factors were identified to be associated with tungiasis: living in houses with human diseases have their source in animals as do 80% of the pathogens
an earthen floors (adjusted OR=3.84; 95%IC: 2.09-7.06), walking barefooted that could potentially be used in bioterrorism, it is imperative to build mod-
(OR=3.28; 1.78-6.04), having a common resting place outside the house els that incorporate ecological health into preparedness and response to
(OR=2.36; 1.01-5.51) and presence of rats on the compound (OR=1.69; 1.03- human disease outbreaks. Using the One Health paradigm, a robust biose-
2.75). Conclusion: Tungiasis is an emerging neglected disease found in Africa. curity system can be developed that adds a layer of support to international
It is highly endemic in rural Central Kenya and associated with considerable disease reporting. This presentation examines a redefinition of traditional
morbidity. The disease is associated with poverty. Modifiable risk factors were biosafety and biosecurity within the context of One Health efforts in Uganda
identified that should be the focus of sustainable and effective control meas- with an emphasis on defining the elements of a One Health risk assessment
ures. and application of biosafety and biosecurity efforts across a One Health plat-
form. The infrastructure and organization, or the lack of it, in Uganda is used
as an example. The article also portrays opportunities to strengthen world-
106 (K) wide biosecurity by defining concentric layers of biosecurity and biosafety
using the One Health paradigm.
Essential Oils of Moroccan Thymus munbyanus as Natural
Product for Nosocomial Infection Prevention
M. Fatima Zohra, Mohammed Khatouf; Badiaa Lyoussi and Abdelfattah Ab-
dellaoui; Faculty of Sci. Dhar Mhraz, Fes, Morocco.
Diseases caused by Gram-negative bacteria resistance to antibiotics are an
increasing problem in Nosocomial infections, and they are responsible of
morbidity in the hospital. In such cases, evaluation of antibacterial activity
must run tests against those bacteria to determine new natural product. The
purpose of this study was to compare the activity antibacterial of essential
oils of thymus to antibiotics used for treatment of Nosocomial infection in
Reanimation Polyvalent and Anesthesia service; CHU Hassan II Fez city. Thy-
mus species have been recommended in Moroccan traditional medicine as an
antispasmodic intestinal and antidiarrheal. The essential oils were isolated by
hydrodistillation, from the aerial parts of Thymus munbyanus possess an-
timicrobial properties important against bacteria tested: Escherichia coli;
Pseudomonas aeruginosa; Klebsiella pneumoniae. The results obtained sug-
gest that the essential oils of the plant could become a source of biologically
active compounds for pharmaceutical industry.

54 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

and sporadic outbreaks of disease associated with its propensity to cause


Poster Session nosocomial infections are infamous. A rapid diagnosis makes useful contri-
018. Tuesday Posters butions to hospital and patient management and is especially pertinent dur-
ing simultaneous outbreaks of dengue fever which may also present with
Tuesday, February 28, 2012 | 1:00 PM - 3:00 PM haemorrhagic symptoms. Results: We have developed a novel and sensitive
Exhibit Hall real time RT-PCR assay for the detection of a broad range of CCHFV geno-
types. Based on a single probe the assay brings added benefits over other
140 (A) similar tests. It has also been transitioned to colleagues in Karachi where it
has proved invaluable for rapid clinical diagnosis. Conclusions: In Pakistan
Molecular Epizootiology of Street Rabies Virus Isolates in rapid diagnosis of patients with CCHF enables prompt admission to isolation
Ukraine units and efficient patient management. Foreknowledge of the likely course
I. Polupan, N. Ivanov, O. Deryabin, V. Skripnik; Inst. of Vet. Med., Kyiv, of disease and implementation of appropriate barrier nursing techniques
Ukraine. greatly reduces the risk of nosocomial transmission. The assay makes broad
To develop and implement an effective program of rabies eradication in contributions to differential diagnosis and supports the management of
Ukraine it is necessary to research the molecular genetic characteristics of haemorrhagic fever at Aga Khan University Hospital, Karachi.
the causative agent of this disease in the country. To conduct this research in
2008 was founded the unique collection of samples of pathological materi-
als confirmed as positive in rabies at the regional veterinary laboratories of 142 (A)
Ukraine. The collection is constantly updated and to present moment it in- Development of Differential and Broad Spectrum Immunologi-
cludes 1294 samples from all regions of Ukraine, selected from 16 animal cal Reagents for Identification and Characterization of Filovirus
species and humans. Identification of the rabies virus in samples of patho-
Sub-Types
logical material for their further selection was carried out using the test de-
C. Bruce, K. Richards, L. Easterbrook, H. Love, L. Hudson, J. Plank, H. Tolley,
veloped by us which based on RT-PCR with primers complementary to the
R. Hewson, A. Robers; Hlth.Protection Agency, Salisbury, United Kingdom.
conservative fragments of the 5’-end of nucleoprotein gene of rabies virus.
Background: With the ever increasing levels of international travel, trade and
For the study of the street rabies virus isolates from the collection we use RT-
threat of bio-terrorism, the need to quickly and efficiently identify filovirus
PCR with the primers pair (509, 304) flanking the variable 3’-end part of nu-
infection is critical. Specialised high-containment laboratories required to
cleoprotein gene of the reference strain of rabies virus CVS (fragment in 377
work with hazard group-4 (HG) agents are costly to run/maintain and can re-
bp). With this pair of primers it was conducted amplification of 76 street iso-
strict experimental procedures. Alternate work strategies are therefore re-
lates, 3 vaccine strains and reference strain CVS of rabies virus. These primers
quired to facilitate the swift and efficient development of new assays required
were complementary to the reference strain CVS and all of vaccine strains of
to support diagnostic and vaccine development programmes. Our work fo-
rabies virus. However from 76 street rabies virus isolates variable fragment
cuses on the development of differential immuno-assays for identification
of 3’-end was amplificated at 32 (42%) but at 44 (58%) amplification didn’t
and characterization of filovirus sub-types. Here we describe a strategy for
take place. Lack of amplification for the part of street rabies virus isolates is
producing monoclonal antibodies (mAb) to HG-4 agents in Biological Safety
indicative of polymorphism of the target nucleoprotein gene fragments. This
Level-2 (BSL) facilities using the baculovirus expression system (BES). Meth-
was used as a criterion for their genetic typing and makes it possible to divide
ods: Viruses Filoviruses were propagated in Vero E6 cells. The strains used
the researched isolates into two genetic clusters. The analysis of the results
were Ebola Zaire Manyinga 1976, Sudan Boniface 1976, Ivory Coast 1994
allowed to draw conclusions about the absence of relation of genetic clusters
and Bundibugyo 2007 and Marburg Ravn and Angola. Virus-Like Particles
with the host species. But the territorial ranking of these genetic variants of
Species-specific VLPs were produced using recombinant baculoviruses con-
rabies virus showed their distinct geographical distribution limited by the
taining glycoprotein (GP) and VP40. Monoclonal Antibodies Mice were im-
Dnipro river. Isolates belonging to cluster I circulate at the western side of
munized with EBOV or MARV VLPs. Monoclonal hybridoma populations were
Ukraine and at the eastern side circulate isolates belonging to cluster II.
assayed for antigen specificity by Enzyme-Linked Immuno-Sorbent Assay, na-
tive and denaturing Western blots. Results: EBOV and MARV recombinant
141 (A) VLPs were shown to exhibit a similar morphology to wild-type virus, and pro-
duced an immune response in mice; producing both GP and VP40 antibodies.
Utility of a New Real-Time RT-PCR Assay for the Detection of Arrays of poly and mAbs were identified that displayed species-specificity, or
Crimean-Congo Haemorrhagic Fever Virus in Pakistan cross-reactivity in the assays used. Conclusions: We have successfully shown
B. Atkinson1, Z. Hasan2, A. Samreen2, B. Jamil2, T. Moatter2, J. Chamberlain1, that the BES can be used to produce EBOV and MARV VLPs, and that they
N. Cook1, C. Logue1, C. Bruce1, S. Dowall1, R. Hewson1; 1Hlth. Protection are immunogenic in mice, producing specific and cross-reactive Abs to dif-
Agency UK, Salisbury, United Kingdom, 2The Aga Khan Univ., Karachi, Pak- ferent filoviruses. These reagents provide the basic building blocks for de-
istan. veloping differential and broad spectrum immunological detection assays,
Introduction: Crimean-Congo Haemorrhagic Fever virus (CCHV) is a seg- and potential therapeutics.
mented, negative stranded, RNA virus classified within the genus Nairovirus
family Bunyaviridae. It is serious human pathogen categorized at biosafety
level 4 (BSL4) mandating the use of maximum containment laboratory facil- 143 (A)
ities such as those at the HPA Porton Down in the UK. The virus is maintained Fighting Fire with Fire: The Development of Antiviral AAVs
in nature in a zoonotic cycle involving a variety of vertebrate species, with K. K. Wong, Jr.; City of Hope Natl. Med. Ctr., Duarte, CA.
animal-to-animal transmission mediated by ticks. Both vertebrate hosts and Adeno-associated virus (AAV) is a 5 kb nonpathogenic, helper dependent
tick vectors act as reservoirs of virus. Transmission to humans commonly oc- member of the Parvovirus Family. Helper viruses have included adenovirus
curs by bite from an infected tick. In addition to zoonotic transmission, CCHFV (Ad), all herpes viruses, human papillomavirus (HPV), and vaccinia virus, all
can also be spread from person to person. Case fatality rates ranging from 10- potential human pathogens. Classically, AAV has been utilized as a viral vec-
50% are common; however person to person transmission, especially in hos- tor by removing endogenous AAV genes, substituting a gene of interest, and
pital settings, often results in higher mortality. CCHF is endemic in Pakistan encapsidating with AAV Rep/Cap genes and specific Ad or herpes simplex

BIODEFENSE PROGRAM & ABSTRACTS 55


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

(HSV) genes. We have chosen another approach.In contrast to currently avail- line with the areas of probable change identified during the regulation re-
able rAAV vectors, we utilized full length wild type AAV (RC rAAV) and inserted view. Key changes have been identified and highlighted to aid in promotion
inhibitory siRNA cassettes targeting specific helper viruses. We were able to of the new regulations. Conclusion: The Center for Biodefense, Law & Public
significantly inhibit expression of the HPV31 E7 gene, which is essential for Policy will conduct a comprehensive analysis and comparison of these new
HPV replication, by 4 different siRNAs. There are several novel and unique regulations in relation to the old regulations.
features of this strategy. (1) When co-infected with a targeted helper virus
(THV), antiviral RC rAAVTHVi vectors replicate, amplify and potentiate their
antiviral effect(s) by increasing DNA template number and augmentation of 146 (A)
antiviral gene expression. In contrast, standard AAV Rep/Cap deficient (NR) Development of a Murine Model for Aerosolized Filovirus Infec-
rAAVTHVi vectors cannot replicate, and require a larger amount of vector for tion
the same level of inhibition. (2) During active infection, RC rAAVTHVi package E. E. Zumbrun, H. A. Bloomfield, T. B. Chance, D. K. Nichols, A. Nalca; US-
and spread to surrounding cells to provide additional protection to both THV AMRIID, Fort Detrick, MD.
infected and uninfected cells. (3) Antiviral RC rAAVTHVi express the AAV2 Rep Aerosol models of filovirus infection have been developed in guinea pig and
protein which possesses additional intrinsic antiviral activity against HPV, non-human primates; however, filovirus infection of wild-type mice by the
and other viruses. (4) RC rAAVTHVi vectors would be easier to produce at aerosol route has not been reported. A murine model of aerosolized filovirus
high titer using a different or irrelevant “helper” virus than the THV, because infection in mice will be useful for screening vaccine candidates and thera-
they can generate a full infectious cycle (replication->packaging->infection pies. In this study, various strains of wild-type and immunocompromised mice
of new cells, etc.) within helper virus infected 293 cells, while NR rAAVTHVi were exposed to aerosolized wild-type (WT) or mouse-adapted (MA) Ebola
vectors cannot. (5) From a Biosafety standpoint, these vectors would be no Zaire (EBOV). Upon exposure to aerosolized WT-EBOV, Balb/c, C57Bl6, and
more toxic than wild type AAV2, which is considered a nonpathogen by the DBA mice were unaffected, but 100% of SCID and 90% of Stat1 knock-out
CDC, assuming that the expressed siRNAs are themselves not toxic. Using (KO) mice succumbed to infection between 7-9 days post-exposure (dpe).
this approach, we have inhibited HSV (HSV UL29.2) and HPV (E7) genes, and Balb/C mice lost approximately 15% of their body weight upon exposure to
are examining inhibition of Ad genes for a potential human clinical trial. MA-EBOV, but all mice recovered. In contrast, 10-30% lethality was observed
in C57Bl6 and DBA mice exposed to aerosolized MA-EBOV, and 100% of SCID,
144 (A) Stat1 KO, Ifnγ -/- and perforin -/- mice succumbed to infection between 7-14
dpe. To identify one or more immunocompetent mouse strains in which ex-
Arenavirus Reverse Genetics for Vaccine Development posure to aerosolized MA-EBOV is uniformly lethal, 62 different BXD recom-
E. Ortiz Riano1, B. Y. H. Cheng1, J. C. de la Torre2, L. Martinez-Sobrido1; 1Univ. binant inbred (RI) mouse strains were tested for susceptibility or resistance
of Rochester Med. Ctr., Rochester, NY, 2The Scripps Res. Inst., La Jolla, CA. to infection. Aerosol exposure to a high dose of MA-EBOV produced a spec-
Arenaviruses are important human pathogens with no FDA-licensed vaccines trum of disease severity, with most strains losing weight but recovering, and
available and current antiviral therapy being limited to an off-label use of the a uniformly lethal disease within 7 to 12 dpe in five strains. Aerosol exposure
nucleoside analog ribavirin. Plasmid-based reverse genetics techniques for of these five BXD strains to 10-fold less MA-EBOV resulted in lethality rang-
the generation of recombinant arenavirus have proven valuable for virus re- ing from 0% in two strains to 100% lethality in only one strain. In the three
search; however, current approaches to generate recombinant arenaviruses BXD strains with lethality at the lower dose of MA-EBOV, analysis of post-
are based on transcription of genomic viral RNA by the murine RNA poly- mortem tissue showed liver damage as well as lung lesions in two of these
merase I promoter, allowing virus recovery only from murine cells. In order to strains, but no lung lesions in the third strain. A computational genetic analy-
develop an efficient reverse genetic system in human cells, we first gener- sis of the RI strains on the basis of outcome to aerosolized filovirus infection
ated a minigenome assay based on the human RNA polymerase I promoter, may allow for identification of gene loci involved in susceptibility or resist-
using Gaussia luciferase and the green fluorescent protein (GFP). Next, we ance to infection. Furthermore, the BXD strain for which even a low dose ex-
used this approach to successfully generate recombinant Lymphocytic Chori- posure to MA-EBOV is uniformly lethal may be useful for testing vaccines and
omeningitis Virus (rLCMV) from human cells and, more importantly, from FDA- therapies.
licensed Vero cells for the future development of potential vaccine seeds.
These novel reverse genetics techniques open new avenues for, both, the
identification of antivirals to treat arenavirus infections, and the generation 147 (B)
of recombinant arenavirus for the development of vaccines in FDA-approved Antibiotic Profile of Nosocomial Bacterial Pathogens
cell lines to prevent arenavirus infections in humans. N. N. Eduok1, I. P. Udoh2, C. I. Eleazar2, B. O. Ogeneh2; 1Ministry of Sci. and
Technology, Uyo, Nigeria, 2Univ. of Nigeria, Enugu, Nigeria.
145 (A) Background: Antibiotic resistance among nosocomial bacteria is a global
problem. This is a concern to medical specialist and microbiologists espe-
New Select Agent Regulations cially as the choice of antimicrobial agents available for the treatment of such
J. Garrett, V. Sutton, H. Carson; Texas Tech Univ. Sch. of Law, Lubbock, TX. infections become limited and inevitably more complicated. Methods: The
Background: The U.S. Departments of Health and Human Services (HHS) and sensitivity pattern of nosocomial bacterial isolates from various clinical sam-
Agriculture (USDA) were required to review and update the National Select ples was investigated. A total of 450 samples were obtained from subjects
Agent Registry rules as promulgated in 42 C.F.R. 73, 9 C.F.R. 121, and 7 C.F.R. both gender of various ages. Isolates were identified using standard micro-
331. The proposed final rules were published on October 3, 2011 in the Fed- biological techniques. Antibiotic susceptibility test was carried out using disc
eral Register and contained several key changes important to researchers. diffusion and modified agar dilution methods. The antibiotics tested against
Methods: Prior to the publication of the new regulations, a thorough exami- the isolates were ampicillin (10μg), chloramphnicol (10μg), cloxacillin (5μg),
nation of the existing regulations, comments from the public, and review of carbennicillin (30mcg), pefloxacin (5iu), ciprofloxacin (5iu), ofloxacillin (5iu),
the recommendations of the Federal Experts Security Advisory Panel and gentamicin (10μg) and augmentin (5iu). Erythromycin (5μg), and strepto-
areas of probable change were identified. Results: Some, but not all, of the mycin (10μg) were tested against Gram positive organisms only, tetracycline
recommendations were addressed in the new regulations, including those (10μg) and cotrimoxazole (25mcg) for Gram negative while nitrofurantoin
from previous comment periods. The majority of the changes made were in (200mcg), nalidixic acid (300mcg), were tested against the urine isolates

56 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

only.The MIC of ampicillin, gentamicin, ciprofloxacin used against some of this in fertile hen’s eggs. The two stocks were compared before aerosol stud-
the clinical isolates were also carried out.Various strains were used as con- ies. As a virulent US strain was not possible to obtain, HPA recovered a vial
trols. Result: Pathogens isolated were as follows: Staphylococcus aureus, of yolk sac (YS) material containing C. burnetii (dated 1993) obtained origi-
Klebsiella pneumoniae, Escherichia coli, Pseudomonas aeruginosa, Proteus nally from ATCC (RSA 423 9mi, 306GP/2EP, 4/26/71). This was used to infect
mirabilis, Enterococcus faecalis and Streptococcus pyogenes. Distribution of three male guinea pigs IP. Spleens were harvested and homogenised at peak
infections in the various sites was as follows: urinary tract (44.5%), blood febrile response and used to inoculate 80 hen eggs. After 11 days, YSs were
(11.1%), genital tract (22.2%), upper/lower respiratory tract (11.1%), and harvested and homogenised. Results: Both C. burnetii stocks were inocu-
wound (11.1%). Ampicillin and gentamicin produced the highest MIC against lated IP into guinea pigs at a dose adjusted to be equivalent to 1.5E+07
S. pyogenes, (3.75) and E. feacalis (1.875),while ciprofloxacin showed the copies by qPCR (sod gene). Both groups of guinea pigs showed clinical signs
lowest MIC against E.coli (0.235) and greatest activity against the various of infection with C. burnetii (body temperature increase and body weight re-
isolates. Conclusion: The high frequency of nosocomial infections places a duction) but, unlike the spleen stock dosed group which all survived, 75%
substantial burden on individual patients and on the health care system while of the group that received YS material had to be euthanised due to severity
emergence of antibiotic resistance has become a medical catastrophe. of disease. These data suggest that the YS material may be more virulent.
The YS material was then delivered via the aerosol route to 3 groups of ani-
mals (guinea pigs and 2 strains of mice). The guinea pig group displayed clin-
148 (B) ical signs of infection (increase in temperature and failure to gain body
Comparative Transcriptomics of Three Species of Fleas weight) but the mice displayed no or minimal clinical signs. These data sug-
(Siphonaptera: Pulicidae) gest that PCR alone is inadequate in determining the virulence of an inocu-
C. A. Whitehouse1, G. Koroleva1, J. R. Kugelman1, S. K. Williams2, S. W. Bear- lum of C. burnetii and guinea pigs are more susceptible than mice to aerosols
den2, K. L. Gage2, G. F. Palacios1; 1USAMRIID, Frederick, MD, 2CDC, Fort of this stock of C. burnetii. Conclusion: We have developed an aerosol model
Collins, CO. of C. burnetii infection in guinea pigs which replicates the clinical signs seen
Background: Fleas serve as vectors of several important diseases. In partic- in acute human infection.
ular, the oriental rat flea, Xenopsylla cheopis, is the primary vector of Yersinia
pestis, the causative agent of plague throughout much of the world. How-
ever, the ground squirrel flea, Oropsylla montana, is considered to be the pri-
150 (B)
mary vector of plague in the western U.S. The cat flea, Ctenocephalides felis, Automated Extraction of Genomic DNA from Food-Associated
can rarely transmit Y. pestis, but is a more important vector of bartonellosis, Pathogens and Environmental Samples
murine typhus, and the dog tapeworm Dipylidium caninum. Despite their im- P. Williams, T. Parrish, M. Duggan, J. Chapman, W. Burke, J. Birkenholz; Evo-
portance, very little is known at the genomic level of these flea vectors, es- gen, Lenexa, KS.
pecially related to their transcriptome. Methods: Pools of 20 individual Introduction: The extraction and purification of DNA from the bacteria Es-
colony-reared fleas from each of the three species were homogenized in 0.1 cherichia coli O157:H7 and Salmonella enterica are essential first steps for
ml of TRIzol reagent, and total RNA was extracted. cDNA libraries were con- the successful amplification of gene targets by PCR. Automated sample prep
structed using the SMART cDNA library construction kit to enhance se- systems allow for less hands-on time, provide greater consistency of ex-
quencing. Libraries were processed for either 454 or Illumina sequencing. tracted sample, and use fewer consumables. We have developed a fully Au-
Hybrid assemblies were produced using Lasergene nGen and BLASTx was tomated Sample Preparation (ASP) instrument which was used to extract and
used to compare contigs to the GenBank non-redundant database. Results: purify DNA from ground beef and turkey meat cultures and environmental
In total, we obtained 7.10 x 106 (Illumina) and 2.02 x 105 (454) reads for X. surface swab cultures. Method: 24 hour cultures of ground beef, turkey, pork,
cheopis, 9.90 x 106 and 1.88 x 105 reads for O. montana, and 8.90 x 106 and and environmental swabs were established using standard USDA protocols.
1.69 x 105 reads for C. felis, which resulted in 16,257 contigs, 16,831 con- 200 μl of culture media from the meat and swab samples were placed in the
tigs, and 14,307 contigs, respectively. BLAST searching of non-redundant ASP cartridge and then extracted and purified using the following automated
databases yielded ~15.4K significant matches to gene families potentially protocol: 200 μl of Evogen ONE was added to the samples which were then
encoding gene products for X. cheopis, ~15.6K for O. montana, and ~13.1K incubated at 95°C for 5 minutes. 250 μl of binding buffer was added to the
for C. felis. Here we compare transcript identity and abundance between the samples, followed by an incubation of 5 minutes. DNA bound to the magnetic
three flea species. Conclusions: This is the first study to elucidate the global beads was washed and then incubated at room temperature for 5 minutes to
transcriptome of three species of fleas that are important disease vectors elute the purified DNA. 200 μl of purified DNA was then automatically trans-
and lays the foundation for future studies of the molecular mechanisms in- ferred to an elution tube. DNA yields were measured with a NanoDrop sys-
volved in vector-pathogen interactions. tem. Validated primers and HyBeacons probes were targeted to the
Salmonella ttr gene and the E. coli eae (gama) gene. 3 μl of the purified DNA
was used in a standard PCR reaction using the following parameters: 95ºC for
149 (B) 15 seconds, 55ºC for 30 seconds, 72ºC for 30 seconds; for 40 cycles on a Bio-
Development of a Coxiella burnetii Aerosol Infection Model in Rad CFX 96 thermocycler. Results: DNA yields from ground beef and swab
Rodents cultures were 26 ng/μl and 17.2 ng/μl respectively. The mean DNA yields from
K. R. Bewley, J. K. Pitman, D. J. Atkinson, G. McLuckie, M. Charlwood, G. ground turkey and swab cultures were 21.9 ng/μl and 20.2 ng/μl respectively.
Hatch, J. Vipond, S. G. P. Funnell, A. Roberts; Hlth. Protection Agency, Salis- Complete analysis time was 3 hours. All gene targets were successfully am-
bury, United Kingdom. plified. Conclusion: The use of the ASP for genomic DNA extraction from S. en-
Background: Coxiella burnetii is the causative agent of Q fever. Infection can terica and E. coli O157:H7 proved to be a rapid and simple protocol which
take several forms, although in man disease ranges from asymptomatic, provided high quality DNA for PCR amplification.
through acute, to life-threatening chronic illness. Chronic disease develops in
approximately 5% of those infected. Methods: In order to develop an aerosol
model of infection, we needed to ensure that the working stocks we produced
were virulent. As a result, we initially prepared a Master stock of C. burnetii
(Nine Mile strain, 1993) in guinea pigs then prepared a working stock from

BIODEFENSE PROGRAM & ABSTRACTS 57


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

151 (B) 153 (B)


Monoclonal Antibody against Capsule of Bacillus anthracis Bacillus anthracis Acetyl-Transferases PatA1 and PatA2 and
Protects Mice from Enhanced Lethal Toxin Activity Due to Cap- Secondary Cell Wall Polysaccharide-Mediated Assembly of S-
sule and Anthrax Spore Challenge Layer Proteins
G-E. Rhie1, J. Jang1, M. Cho1, J-H. Chun1, J. Park2, K. Hong1; 1Korea NIH, Chung- J. M. Lunderberg1,2, S-M. Nguyen-Mau1,2, G. S. Richter1,2, Y-T. Wang1,2, J.
buk, Korea, Republic of, 2Hankuk Univ. of Foreign Studies, Yongin, Korea, Re- Dworkin3, D. M. Missiakas1,2, O. Schneewind1,2; 1Univ. of Chicago, Chicago,
public of. IL, 2Howard Taylor Ricketts Lab., Argonne, IL, 3Columbia Univ., New York, NY.
The poly-γ-D-glutamic acid (PGA) capsule is one of the major virulence factors The envelope of Bacillus anthracis, the causative agent of anthrax, is com-
of Bacillus anthracis, which causes a highly lethal infectious disease. The PGA prised of a poly-D-γ-glutamic acid (PDGA) capsule, a proteinaceous S-layer
capsule has been known to disguise B. anthracis from immune surveillance with S-layer associated proteins and the cell wall peptidoglycan. Recent work
and allow its unimpeded growth in the host. Recently, the importance of PGA suggests that, in addition to PDGA, S-layer and S-layer associated proteins
in the pathogenesis of anthrax infection is being emphasized. The PGA cap- contribute to the pathogenesis of anthrax. Protein assembly in the envelope
sule is associated with lethal toxin (LT) in the blood of experimentally infected of B. anthracis requires S-layer homology domains (SLH) within S-layer and
animals and PGA was shown to produce enhancement of the cytotoxicity of S-layer associated proteins. SLH domains fold into a three-pronged spindle
LT through increasing binding of protective antigen to its receptors resulting structure that associates with the secondary cell wall polysaccharide (SCWP),
more accumulation inside cells. In this study, we confirmed the enhancement a carbohydrate that is tethered to the peptidoglycan of B. anthracis. Earlier
effect of PGA on LT-mediated cytotoxicity ex vivo using mouse bone marrow work described the structure of SCWP as [→6)-α-GlcNAc-(1→4)-β-ManNAc-
derived macrophage (BMDM). Western blot analysis showed that the com- (1→4)-β-GlcNAc-(1→]n, where α-GlcNAc is substituted with α-Gal and β-Gal
bination of PGA and LT produced a greater degree of degradation of MAP ki- at O3 and O4, respectively, and the β-GlcNAc is substituted with α-Gal at O3.
nase kinases and an increased level of activation of the proform of caspase-1 The SCWP is modified with pyruvyl and acetyl moieties. Pyruvylation involves
to its processed form as compared to LT alone. For the evaluation antibody the csaB gene, which is essential for S-layer assembly and whose product is
against PGA on the prevention of progress of anthrax disease, mouse mono- thought to function as a pyruvyl-transferase. Here, the effects of two putative
clonal antibodies (mAbs) against PGA were produced. mAbs bind encapsu- acyl-transferases, PatA1 and PatA2, on SCWP acetylation are investigated
lated B. anthracis H9401 specifically and showed opsonophagocytosis through MALDI-TOF mass spectrometry and fluorescent protein binding as-
activity against encapsulated B. anthracis H9401. Anti-PGA mAbs also says. Our data suggest PatA1 and PatA2 act on the SCWP and affect the dep-
showed protective effect against both spore and PGA enhanced LT toxicity osition of some, but not all, SLH-domain containing proteins into the S-layer
challenges in mice. These results indicate that anti-PGA mAb protects the of B. anthracis.
host from the progression of anthrax not only by eliminating vegetative form
of encapsulated B. anthracis but also from by inhibiting the enhancing cyto-
toxic activity of LT by PGA and suggest a role of anti-PGA antibody in pre- 154 (B)
venting and treating anthrax infection. Autotransported (Type V Secreted) Proteins of Burkholderia
pseudomallei and Their Role in Biofilm Formation, Serum Re-
152 (B) sistance and Pathogenesis
N. R. Lazar Adler1, R. E. Dean2, M. P. Stevens3, J. L. Prior2, T. P. Atkins2,4, E. E.
The Cat Flea as a Laboratory Model for Understanding Yersinia Galyov1; 1Univ. of Leicester, Leicester, United Kingdom, 2Defence Sci. and
pestis Colonization of its Insect Vector Technology Lab., Salisbury, United Kingdom, 3Univ. of Edinburgh, Easter
T. M. Hermanas, L. E. Quenee, O. Schneewind; Univ. of Chicago, Chicago, IL. Bush, United Kingdom, 4Univ. of Exeter, Exeter, United Kingdom.
In North America, 28 species of flea have been confirmed as vectors capable Burkholderia pseudomallei, a saprophyte endemic to the soil and water of
of transmitting Yerinsia pestis and maintaining the zoonotic/epizootic cycle. tropical regions, is the causative agent of Melioidosis, a potentially fatal
Epizoonotic epidemics of plague are thought to initiate when fleas acquire febrile human and veterinary disease. The molecular mechanisms of B.
Yerinsia pestis by feeding on heavily septicemic rodents with subsequent pseudomallei pathogenesis remains to be fully characterised. Autotrans-
transmission to human hosts. Research on the rat flea (Xenopsylla cheopis) porters are a family of secreted or membrane-associated proteins which me-
suggests that roughly 50 percent of fleas that feed on a septicemic host are diate their own secretion across the Gram-negative membranes via the type
colonized by Yersinia pestis. Colonization requires the formation of a biofilm V secretion pathway. Autotransporters from different bacterial species have
in the flea’s midgut, which eventually causes the infected flea to starve and diverse functions, with many demonstrated to play a role in virulence. Eleven
die. Colonization of the flea midgut has been linked to the production of n- genes encoding predicted autotransporters are present in the B. pseudoma-
acetyl- glucosamine by the heamin storage (hms) locus and phospolipase D llei K96243 genome (AT1-11). Most of these autotransporters remain un-
activity of the Yersinia murine toxin gene (ymt). Our goal is to develop a model studied; hence, single cross-over insertional mutants were constructed and
for colonization of the cat flea (Ctenocephalides felis) to help identify the subjected to a range of investigations to elucidate their role in pathogenesis.
Yersinia pestis requirements for flea colonization. Using an artificial feeding All mutants were assessed for intracellular invasion and survival within the
system, we observe transmission of Yersinia pestis CO92 pCD1- from blood A549 epithelial-like cell line with two mutants, AT7 and AT11, demonstrating
to the flea with subsequent persistent colonization. The progress of the in- attenuation. Furthermore, AT11 also displayed attenuation for intracellular
fection is monitored using CFU enumeration and microscopy. Our results survival in the J774 macrophage-like cell line. Biofilm assays were performed
show that Y. pestis persistence requires gradual growth in the digestive tract for all mutants and AT5 demonstrated reduced biofilm formation at 37°C but
for 21 days after feeding of the initial infectious dose. Experiments with wild- wild-type biofilm levels at 27°C. Evaluation of the survival of all mutants in
type Y. pestis and isogenic mutants show that the hms and ymt genes are re- 45% normal human sera identified that AT6 was sensitive to complement-
quired for persistent infection of the flea digestive tract. Additionally, we mediated killing. Eight mutants have been screened in the BALB/c Melioido-
utilize the cat flea model to screen iron transport genes in an attempt to un- sis mouse model, with five demonstrating attenuation for virulence (varying
derstand the role of iron acquisition during flea colonization. from a 1 to a 1.6-log reduction). Taken together, our data indicate that several
putative Type V secreted proteins play a role in the virulence of B. pseudo-
mallei and warrant further study. (c) Crown Copyright. Dstl, 2011

58 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

155 (B) 157 (B)


S-Layer Assembly in Bacillus cereus G9241 Requires csaB Salmonella Enteritidis Gene SEN1140 Contributes to Acceler-
Y-T. Wang1,2, S-Y. Oh1,2, A. P. Hendrickx1, O. Schneewind1,2; 1Univ. of Chicago, ated Type III Secretion System-2 Dependent Early Inflamma-
Chicago, IL, 2Howard Taylor Ricketts Lab., Argonne, IL. tion Kinetics in Mouse Colitis Model
The surface (S)-layer is a structure of crystalline protein arrays deposited on V. VISHWAKARMA1, B. Periaswamy2, N. B. Pati1, W-D. Hardt2, M. Suar1; 1Sch.
top of the cell envelope of many bacterial species. We sought to identify and of Biotechnology, Bhubaneswar, India, 2Inst. of Microbiol., Zürich, Switzer-
characterize the S-layer in Bacillus cereus G9241, which causes an anthrax- land.
like respiratory illness in humans and immune-competent mice. Our results Salmonella enterica subsp. I serovar Enteritidis (S. Enteritidis) exhibits faster
suggest that B. cereus G9241 immobilize S-layer proteins in their envelope in type III secretion system 2 (TTSS-2) dependent early colonization and in-
a csaB dependent manner. The product of the csaB gene is thought to pyru- flammation kinetics compared to its closely related serovar Typhimurium. To
vylate the secondary cell wall polysaccharide of Bacillus anthracis and Ther- understand the molecular mechanistic differences, in particular to investi-
mus thermophilus, thereby capturing secreted proteins with an N-terminal gate the TTSS-2 dependent accelerated pathogenic potential of S. Enteritidis,
S-layer homology domain (SLH). B. cereus csaB variants cannot retain the we focused on genes unique to its genome. Previously published compara-
main S-layer proteins, Sap and EA1, in the bacterial envelope. Further, com- tive genomic findings revealed presence of mutually exclusive genes in both
pared to the wild-type, the csaB variant displays altered colony morphology serovars. As a pilot study we investigated the role of six unique genes of S.
and a long chain growth phenotype of its vegetative forms. When evaluated Enteritidis in-vivo. Wild type isogenic tagged strains (WITS) of S. Enteritidis
in an intraperitoneal challenge model with C57Bl/6 mice, the csaB mutant with plasmid carrying gene specific putative promoter constructs with a
was attenuated as compared to wild-type B. cereus G9241. In addition to S- downstream GFP reporter were used to coinfect C57BL/6 mice and were later
layer proteins, B. cereus G9241 also elaborates two capsular polysaccha- interpreted by differential fluorescence induction (DFI), fluorescence acti-
rides, which are encoded by plasmids pBCXO1 and pBC218. Our results vated cell sorting (FACS) followed by quantitative real-time polymerase chain
suggest that the assembly and function of the S-layer and the capsular poly- reaction (qRT-PCR) for WITS tags to identify differentially expressed promot-
saccharides is carefully coordinated to enable the pathogenesis of anthrax- ers in sorted fractions. Using this strategy, promoter construct for gene
like disease. SEN1140 was found to express GFP in gut lumen, lamina propria, mesenteric
lymph nodes, spleen and liver. To investigate the role of SEN1140, knockout
mutant of the same was compared with S. Typhimurium on a type III secre-
156 (B) tion system 1 deficient (TTSS1-) background. Interestingly, the S. Enteritidis
Antibody Interactions with the Capsule of Bacillus anthracis mutant lacking SEN1140 did not show the unique early colonization and in-
M. A. Hubbard, P. Thorkildson, W. Welch, D. AuCoin, T. Kozel; Univ. of Nevada flammation kinetics phenotype in comparison to S. Typhimurium. To sub-
Sch. of Med., Reno, NV. stantiate it, complementation of SEN1140 restored the TTSS-2 dependent
Bacillus anthracis is the etiologic agent of anthrax, a disease of large mam- accelerated inflammatory potential of S. Enteritidis. Furthermore, this pilot
mals including humans. The bacterium synthesizes a defensive capsule en- study represents a suitable combination of DFI, FACS, qPCR and WITS tech-
tirely composed of poly-gamma-D-glutamic acid (PGA). We previously niques to sensitively screen S. Enteritidis specific genes for their unique role
demonstrated that passive immunization with a murine monoclonal antibody in bacterial pathogenesis.
(mAb) reactive with PGA is protective in a pulmonary model of anthrax. These
results are part of a growing body of research that has demonstrated the
merit of capsule-targeted immunity, leading to considerable interest in the 158 (B)
incorporation of PGA into anthrax vaccines. In the present study we sought to Activity of Two Novel Antibacterial Agents XF-70 & XF-73
further understand the modality of antibody binding to this unique antigen. against Multidrug-Resistant Acinetobacter baumannii
The original mAb was subject to CDR3 randomization and phage display, K. L. Lohman1, J. Coffin1, W. Rhys-Williams2, I. Hayter2, W. G. Love2;
which lead to the recovery of a high affinity variant. Binding of both the par- 1
MRIGlobal, Rockville, MD, 2Destiny Pharma Ltd., Brighton, United Kingdom.
ent and mutant antibodies was analyzed using surface plasmon resonance. Background: XF-70 and XF-73 are members of a new class of membrane ac-
The results of this study indicate that the association of mAb to PGA consists tive topical drugs which have broad spectrum activity against a range of bio-
of two kinetic classes: the faster is first order in mAb concentration, the threat agents, as well as other clinically relevant bacteria such as MRSA. This
slower zero order in mAb concentration. Dissociation appears to occur as a intrinsic activity is, in certain cases, augmented by light activation to produce
single kinetic class. A molecular model was constructed to provide an atom- an additional photodynamic effect. Here we report the results of an in vitro
istic explanation. Molecular dynamics simulations in explicit solvent demon- screen against a panel of clinically relevant multidrug resistant Acinetobac-
strate that the polyvalent nature of PGA is exploited by multiple interactions ter baumannii isolates with and without light activation. Methods: Multidrug
with the surface of the antibody. Molecular contacts occur along a positively resistant A. baumannii isolates from burn, bloodstream and upper and lower
charged groove. Although traditional CDRs were involved, surprisingly, respiratory tract infections were selected from the ATCC culture collection.
residues outside the region were large contributors to PGA binding. These MICs were determined by broth microdilution according to standard CLSI
results may indicate an alternative method of antibody binding and selection methodology (M100S21: Performance Standards for Antimicrobial Suscepti-
to combat polyanions by the immune system. bility Testing, January 2011). Colistin and Imipenem were included as con-
trols. For light activation studies, the plates were illuminated with a light
source with a wavelength range of 380 - 480nm for 15 minutes. Staphylo-
coccus aureus and Pseudomonas aeruginosa were included as bacterial con-
trols. Results: The intrinsic A. baumannii MICs for XF-70 were in the range of
32 to 64 µg/ml, which reduced to <0.25 µg/ml after 15 minutes illumina-
tion. The intrinsic A. baumannii MICs for XF-73 were reduced from 64-128
µg/ml to <0.25 µg/ml after illumination. The antibiotic resistance profile of
the isolates had no effect on the activity of either XF-70 or XF-73. MICs for
the control drugs were as expected. Conclusions: The results reported here

BIODEFENSE PROGRAM & ABSTRACTS 59


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

demonstrate that light activated XF-70 and XF-73 have potent activity against against a virulent strain collection (A total of 37 strains were evaluated). All
all of the A.baumannii isolates tested, irrespective of their drug resistance phages revealed different lytic activity against virulent strains. Further re-
and infection source. The results suggest that XF-70 and XF-73 may be of util- search will be focused on research of biological peculiarities of the genetically
ity in the treatment of A.baumannii infections and is worthy of further inves- different B. anthracis phages, which could lead to a more elaborate and im-
tigation proved diagnostics capability of phage typing of B. anthracis.

159 (B) 161 (B)


Anthrax Surveillance System and Canonical SNP Typing of Characterisation of Inhalational Infection with Burkholderia
Bacillus anthracis Strains Isolated in the Country of Georgia pseudomallei in BALB/c mice
2009-2011 S. G. P. Funnell, G. Hatch, J. A. Kane, S. Bates, K. M. Tonks, C. H. Tsang, J.
G. Chanturia1, E. Khmaladze1, E. Zhgenti1, L. Malania1, M. Zakalashvili1, N. Fretwell, H. Shuttleworth, J. Vipond, A. Roberts; Hlth. Protection Agency, Sal-
Tsertsvadze1, M. Kekelidze1, P. Imnadze1, S. Tsanava1, R. J. Obiso2, S. isbury, United Kingdom.
Francesconi3; 1The Natl. Ctr. for Disease Control and Publ. Hlth., Tbilisi, Geor- Background: Burkholderia pseudomallei is the aetiologic agent of melioido-
gia, 2The Microbe Company, Christiansburg, VA, 3Naval Med. Res. Ctr., Fred- sis, a disease which is endemic in Southeast Asian and Northern Australia.
erick, MD. Natural infection can occur via a range of routes including contamination of
Bacillus anthracis, the causative agent of anthrax, due to its high virulence skin abrasions, inhalation of aerosols and ingestion of contaminated water.
and ability to persist as a spore, poses a great threat as an emerging bioter- The aim of this study was to characterise inhalational infection of BALB/c
rorism agent. The epidemiology of this pathogen was studied in “Active Sur- mice with B. pseudomallei. Methods: Groups of BALB/c mice were challenged
veillance of Especially Dangerous Pathogens in the Southern Caucuses by the aerosol route using a modified Henderson apparatus in conjunction
Region” project, focused on monitoring the prevalence and distribution of with the Biaera aerosol management platform with different doses ranging
several especially dangerous pathogens, natural foci of which are located on from 3343 cfu to 3 cfu of B. pseudomallei strain K96243. Animals were
the territory of the South Caucasus. Within the project framework 1989 en- weighed and observed for clinical signs. In selected studies, at scheduled
vironmental samples have been investigated in 2009-2011. Samples were time points, the lung, spleen, kidney, and liver tissues were collected for
plated on PLET and SDA media. Suspicious for anthrax colonies were inves- histopathological profiling and also for enumeration of bacterial load using
tigated according to SOPs provided by DTRA. The final confirmation was per- Luria Bertani Agar. Ashdown’s agar was also used to assess morphological
formed using Real-time PCR. Totally 63 out of obtained 148 B. anthracis variation. Enumeration was conducted using an SOP based on classical
strains were chosen for further investigation. These isolates in addition with colony counting, and morphology variation was recorded photographically.
archival strains from National Center for Disease Control and Public Health Results: At high doses (above 500 cfu), all mice develop an acute disease
(NCDC) were characterized using canonical SNPs in order to place them in a which resulted in death within ten days of infection. At low doses, the ma-
global phylogeographic context. From this analysis we discovered two very jority of mice survived beyond 14 days after infection. The disease in these
distinct and relatively distant populations of B. anthracis. Majority of the survivors appeared to take a more indolent course. The histopathology and
strains belonged to the A.Br.003 together with Australian 94 lineage and very bacterial loads of the sampled tissues indicated that the disease progression
few of them went to the A.Br.008/009 (WNA lineage). Within the small geo- in survivors at day 30 would inevitably have led to a relapse. In addition, the
graphic scale of Georgia, the identification of two distinct and genetically dis- degree of B. pseudomallei morphological variation observed in tissue iso-
tant populations of B. anthracis suggests that Georgia is the site of multiple lates was greater than that seen in the challenge inoculum. Conclusion:
waves of B. anthracis dispersals from different origins. Obtained results re- These results indicate that BALB/c mice can be used to model human infec-
vealed need for further in depth epidemiological study of the cases. tion with B. pseudomallei and that the challenge dose will determine whether
an acute or chronic manifestation of disease is observed.

160 (B)
Diversity of Bacteriophages Active to B. anthracis Bacterial
162 (B)
Strains Isolated in Georgia Reaerosolization of Bacillus Spores During the Bio-Response
S. Rigvava1, M. Natidze2, L. Gubeladze1, L. Kvachadze1, R. J. Obiso3, M. Ku- Operational Testing and Evaluation
tateladze1, R. Adamia1; 1G. Eliava Inst. of Bacteriophages, Microbiol. and Vi- S. C. Taft1, R. A. Lordo2, D. J. Chappie2, E. Silvestri1, T. L. Nichols1, J. A. Rosati
rology, Tbilisi, Georgia, 2G, Tbilisi, Georgia, 3The Microbe Company, Rowe1; 1Natl. Homeland Security Res. Ctr., Cincinnati, OH, 2Battelle Mem.
Christiansburg, VA. Inst., Columbus, OH.
Bacteriophages are bacterial viruses that reveal specific lytic activity against Background: The Federal Interagency Bio-Response Operational Testing and
host bacterial strains. The Eliava Institute, Tbilisi, Georgia is one of the most Evaluation (BOTE) project was conducted to evaluate the efficacy of three de-
experienced institutes for phage isolation, research, and application. Several contamination technologies on the Bacillus anthracis spore surrogate, Bacillus
bacteriophages active against B. anthracis have been characterized. The aim atrophaeus subspecies globigii (Bg), disseminated in a building. During BOTE,
of this research study is to isolate, characterize, and study new specific complex air and surface sampling was also conducted to quantify potential ex-
phages against B. anthracis. Several lytic phage were isolated from two, ge- posure from the aerosolized spores during the release, from the subsequent
ographically distinct loci: one from West Georgia (12 phage from the Kutaisi re-aerosolized spores pre-decontamination, and from the re-aerosolized resid-
region) and the second from Eastern Georgia (6 phage from the Gardabani re- ual spores post-decontamination. Methods: SKC Bioaerosol samples were col-
gion). The phages were isolated and then propagated on vaccine strains of B. lected at five different time stages (background, during dissemination, before
anthracis (34F2, 55 and STI1). Bacteriophage isolated in this study have the surface sampling, during pre-decontamination surface sampling, and during
same morphology as the internationally accepted B. anthracis bacteriophage post-decontamination surface sampling) for each of the three decontamination
Gamma— the phages have hexagonal heads and long, non-contractile tails. technology test rounds (amended bleach, chlorine dioxide fumigation, and va-
All phages are very specific and do not lyse any other Bacillus species other porized hydrogen peroxide fumigation). Wipes, sponge sticks, and vacuum sur-
than strains of Bacillus anthracis. Each of the phage isolated differ from each face samples were also collected in the same rooms during the last two time
other by DNA restriction and structural proteins. The phages were tested stages in conjunction with the SKC Bioaerosol samples. The surface concen-

60 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

tration measurements were linked to the air concentration measurements to


parameterize potential reaerosolization of the spores due to human surface
165 (B)
sampling activity within a contaminated indoor setting. Results: The back- Achromobacter Xylosoxidans: An Emerging Pathogen Carrying
ground and post-decontamination air and surface samples were primarily non- Different Elements Involved in Horizontal Genetic Transfer
detects. There were significant differences observed in the air sampling results G. Traglia1, C. Adams2, M. Almuzara3, C. Vay3, D. Centrón1, M. Ramirez2,1; 1Sch.
for the other the time stages (dissemination, before surface sampling, and dur- of Med., Buenos Aires, Argentina, 2California State Univ., Fullerton, CA, 3Hosp.
ing pre-decontamination surface sampling). The surface to air concentration de Clinicas, Jose de San Martin, Buenos Aires, Argentina.
measurement comparisons varied considerably among the different surface In the last few years numerous cases of multidrug resistant Achromobacter
sampling teams, surface sampling collection methods, and surface types. Con- Xylosoxidans infections have been documented in immunocompromised and
clusions: These data confirm that single, point reaerosolization rates or factors cystic fibrosis patients. To gain insights into the molecular mechanisms and
are not sufficient for predicting potential reaerosolization from different sur- mobile elements contributing to multidrug resistance of this bacterium, we
face types or human activities. studied 28 non-epidemiological A. xylosoxidans clinical isolates from Ar-
gentina. All isolates were identified using standard biochemical tests, API
20NE (Biomeriux), and 16S rRNA gene sequencing. Clonal relationships were
163 (B) determined by macrorestriction mapping. Total DNA was used as template
Regulation of the Yersinia Type Three Secretion Pathway for PCR amplification reactions. Specific primers for plasmids (IncP, IncW,
K. Kopaskie1,2, J. McCool3, K. Aly1, B. Blaylock1,2, O. Schneewind1,2; 1Univ. of IncA/C, IncN, IncFII, repAci1), transposons (Tn1331, Tn3, Tn7), insertion se-
Chicago, Chicago, IL, 2Howard Taylor Ricketts Lab., Argonne, IL, 3Boston Univ., quences (IS) (IS26, IS1999, IS440, ISPa12, ISPa13, IS18, IS125, IS1008,
Boston, MA. ISAba1), and the blaampC, intI1 and intI2 genes were used in PCR reactions.
Three pathogenic Yersinia species—Yersinia pestis, Yersinia pseudotubercu- The variable regions (vr) of integrons and their association with IS26 and
losis and Yersinia enterocolitica—use a virulence plasmid-encoded type III se- IS440 were analyzed by PCR cartography. IS26 and IS440 were found in 2
cretion machine to inject effector proteins into host immune cells. The Yersinia and 7 isolates, respectively. Wide host range IncP plasmids were identified in
type III secretion pathway is essential for the pathogenesis of human or animal 10 isolates. The intI1 and intI2 genes were present in 10 and 3 isolates, re-
disease and some of its subunits, which are currently being tested as purified spectively. PCR cartography showed 4 different arrays in the vr of class 1 in-
antigens for plague vaccine development. During host infection, Yersinia in- tegrons. Relevant gene cassettes found in these vr include the
sert their preassembled secretion needles into the plasma membrane of im- aminoglycoside resistance aac(6’)-Ib and aadA1, the trimethoprim resistance
mune cells. This encounter is perceived by the YscF needle protein as a drop in dfrA1 and dfrA16, and the beta-lactamase blaOXA-2. In only one of the class 2
calcium concentration, triggering the synthesis and secretion of effector pro- integrons a vr was amplified, which included dfrA1. No association of inte-
teins (YopE, YopH, YopJ, YopM, YopO, and YopT). A hydrid protein, YopN-TyeA, grons with IS26 and IS440 were found. The blaampC gene was found in all
which is comprised of two other regulatory factors foe effector transport, com- isolates, confirming its ubiquitous nature. Our results show that A. xylosox-
pletely blocked Yersinia type III secretion and rendered bacteria non-respon- idans clinical isolates contain a rich variety of genetic elements commonly
sive to calcium signaling. YopN-TyeA was localized to the bacterial membrane associated with resistance genes and their dissemination. This supports the
fraction. Detergent solubilized membrane proteins were used for YopN-TyeA hypothesis that A. xylosoxidans is becoming a reservoir of resistance deter-
affinity chromatography experiments to identify machine components that may minants included in mobile elements.
contribute to the shut-down of the Yersinia type III pathway.
166 (B)
164 (B) Increase in Temperature of Cultivation Above 37°C Influenced
Real-Time Monitoring of Bacillus thuringiensis as a Surrogate Production of Yersinia pestis Major Protective Antigens F1 and
for Bacillus anthracis in Potable Water Distribution System LcrV
S. P. Sherchan, I. Pepper; Univ. of Arizona, Tucson, AZ. V. A. Feodorova1, T. I. Polyanina1, M. A. Khizhnyakova1, A. M. Lyapina1, O. V.
Following the events of September 11, 2001, and the subsequent anthrax Ulianova2,1, V. L. Motin3; 1Saratov State Vet. Inst., Saratov, Russian Federa-
mail attack on the Eastern United States, the focus on bioterrorism in partic- tion, 2Saratov State Univ., Saratov, Russian Federation, 3Univ. of Texas Med.
ular has become significant. Therefore, there is a need for integrated and in- Branch, Galveston, TX.
telligent sensors to operate in real-time,with the ability to recognize and Background: Human plague, which is an acute and often a fulminant disease,
diagnose day-to-day and perhaps minute-to-minute water quality distur- is characterized by a rapid onset of high fever up to 40-42°C. The purpose of
bances. Currently there is no available technology that will specifically iden- this study was to investigate whether Y. pestis responds to elevated tem-
tify a microorganism in real-time; however, there are commercial sensors perature by changing its production of specific antigens. Methods: Y. pestis
available that can detect microbial water quality changes in less than five EV NIIEG was grown in Heart Infusion Broth (HIB) supplemented with cal-
minutes with a sufficient sampling rate as to be considered “real-time.” The cium, magnesium, glucose, or a combination thereof at physiological con-
objective of this study was to evaluate the potential for real-time monitoring centrations, either at pH 7.2 or 5.5 and temperatures in the range of
of B.thuringiensis as a surrogate for B. anthracis using a variety of water qual- 26°C-42°C. Production of two major Y. pestis virulence factors and protective
ity sensors including: the HACH Guardian Blue Monitoring Platform; the JMAR antigens, such as capsular antigen F1 and LcrV, was evaluated in dot-ELISA,
BioSentry unit; and the S::CAN spectro::lyser technology. The minimum SDS-PAGE and immunoblots by using monospecific polyclonal antibodies to
threshold responses were examined by injecting B.thuringiensis either in these proteins. In addition, the existence of a capsule was visualized by using
deionized (DI) or tap water with spore concentration ranging from 102- 105 both light and electron microscopy. Results: Biosynthesis of both F1 and LcrV
colony forming units (cfu)/mL. Most sensors responded to increase in spore by Y. pestis EV NIIEG at neutral pH in HIB with no supplements was signifi-
concentration over the range of 102 - 105 cfu/ml and below this range; sen- cantly enhanced at growth temperatures above 39°C with a maximum at 40-
sors provided signals undistinguishable from noise. The data obtained thus 42oC. Addition of either calcium or magnesium in combination with glucose
far suggest that select sensors can detect microbial water quality changes elevated the production of LcrV, but not of F1. The medium containing either
and these advanced technologies can be integrated to monitor intrusion all three components or HIB with magnesium alone also stimulated the pro-
events in water distribution systems. duction of both antigens at an elevated temperature with a peak at 38°C-

BIODEFENSE PROGRAM & ABSTRACTS 61


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

39°C. When production of F1 and LcrV was tested at acidic pH, we observed Novel SNPs were discovered and converted into molecular assays which were
similar trends indicating that production of these antigens correlated with the used to screen the A.Br.013/015 Georgian isolates as well as other isolates
increase in cultivation temperature. Moreover, a 3.7-5.3-fold increase in the in our world-wide collection from this group. We identified a new sub-lineage
bacterial yield, depending on the combination of supplements, was seen at a within the A.Br.013/015 clade that contained most of Georgian isolates and
growth temperature of 39°C-42°C. The largest capsule size was found at 39°C- twelve isolates from Turkey. Conclusion: This finding is consistent with the
40°C. Conclusions: Elevation of temperature above 37°C influenced the pro- idea that this new genetic group may be geographically restricted to border
duction of two major Y. pestis antigens F1 and LcrV that can be critical during regions of the European/Asian continents. Within this new sub-lineage, eight
the infectious process and/or in eliciting protective immunity against plague. new genetic subgroups were identified. This restricted geographic distribu-
tion, as well as the high levels of genetic diversity within the lineage, is con-
sistent with a relatively older origin and localized differentiation. Within the
167 (B) small geographic scale of Georgia identification of two distinct and geneti-
Infection and Dissemination, Francisella tularensis holarctica cally distant populations of B. anthracis suggests that Georgia is the site of
in Mosquito Vector multiple waves of B. anthracis dispersals from different origins.
S. Backman; CBRN Defence and Security, Umea, Sweden.
Background: A number of different insect species are suggested to transmit
Francisella tularensis (F. t.), the etiological agent of the zoonosis tularemia,
169 (B)
to susceptible hosts. Mosquitoes are traditionally considered mechanical Polymorphisms in the lcrv Gene of Yersinia enterocolitica Do
vectors of F. t. holarctica and the main vector for transmission of tularemia to Not Provide for Escape from Plague Protective Immunity
humans in Sweden. It is however not clear how mosquitoes acquire and trans- N. C. Miller, L. Quenee, D. Elli, N. Ciletti, O. Schneewind; Univ. of Chicago,
mits the bacterium. The aim of this study was to investigate the hypothesis Chicago, IL.
that the mosquito already as larvae acquire the bacterium and as adult trans- Current efforts to develop plague vaccines focus on LcrV, a polypeptide that
mit the disease to susceptible hosts. Methods: In order to confirm presence resides at the tip of type III secretion needles. LcrV-specific antibodies block
of F. t. holarctica in the Swedish mosquito population field sampled mosqui- Y. pestis type III injection of Yop effectors into host immune cells, thereby en-
toes (sampled in an area with endemic tularemia, Örebro) were pooled (266 abling phagocytes to kill the invading pathogen. Earlier work reported that
pools with 1-50 mosquitoes) and screened by PCR targeting F. t. in general antibodies against Y. pestis LcrV cannot block type III injection by Yersinia
(lpnA2) and subsp. holarctica in specific (FtM19InDel). Further, a mosquito enterocolitica strains and suggested that lcrV polymorphisms may provide
model, Aedes aegypti, was established. Mosquitoes were infected in the lar- for escape from LcrV-mediated plague immunity. We show here that poly-
val stage and the persistence of the bacterium was monitored through the de- clonal or monoclonal antibodies raised against Y. pestis KIM D27 LcrV
velopmental stages using PCR. To evaluate transmission, laboratory infected (LcrVD27) bind LcrV from Y. enterocolitica O:9 strain W22703 (LcrVW22703) or
mosquitoes were allowed to feed on mice. Results: Twenty out of 266 pools O:8 strain WA-314 (LcrVWA-314), but are otherwise unable to block type III in-
of field sampled mosquitoes were positive for presence of F. t. specific se- jection by Y. enterocolitica strains. Replacing the lcrV gene on the pCD1 viru-
quences and 18 of those contained sequences specific for the clinically rele- lence plasmid of Y. pestis KIM D27 with either lcrVW22703 or lcrVWA-314 does
vant subsp. holarctica. Based on our model experiments F. t. holarctica persist not affect the ability of plague bacteria to secrete proteins via the type III
through mosquito development and 25 % of the flying adults (infected as lar- pathway, to inject Yops into macrophages or to cause lethal plague infections
vae) were PCR positive. None of the animals developed tularemia in the trans- in mice. LcrVD27-specific antibodies blocked type III injection by Y. pestis ex-
mission experiment. Conclusion: F. t. holarctica circulates in the mosquito pressing lcrVW22703 or lcrVWA-314 and protected mice against intravenous
population in a Swedish area with endemic tularemia. Further, the bacteria lethal plague challenge with these strains. Thus, although antibodies raised
persist through the developmental stages of laboratory infected mosquitoes. against LcrVD27 are unable to block the type III injection of Y. enterocolitica
Taken together this supports the hypothesis of an aquatic reservoir for F. t. strains, expression of lcrVW22703 or lcrVWA-314 in Y. pestis did not provide for
holarctica between outbreaks. However, in order to establish vector compe- escape from LcrV-mediated plague protective immunity in the intravenous
tence and transmission rates further investigations are needed. challenge model.

168 (B) 170 (B)


Phylogeography of Bacillus anthracis Isolates from the Coun- Multiple-Locus Variable Number Tandem Repeat Analysis of
try of Georgia Brucella melitensis from Azerbaijan Using Eight Markers
E. Khmaladze1, D. Birdsell2, E. Zhghenti1, G. Chanturia1, M. Kekelidze1, M. X-Z. Huang, E. B. Zelazowska, M. P. Nikolich; Walter Reed Army Inst. of Res.,
Nikolich3, P. Imnadze1, S. Tsanava1, L. Malania1, N. Tsertsvadze1, S. Beck- Silver Spring, MD.
strom-Sternberg4, J. Beckstrom-Sternberg4, T. Pearson2, D. Wagner2, P. Keim2; Background: Brucella melitensis is one of the pathogens that causes brucel-
1
Natl. Ctr. for Disease Control and Publ. Hlth. of Georgia, Tbilisi, Georgia, losis, a common bacterial zoonosis with global spread that is endemic in For-
2
Northern Arizona Univ., Flagstaff, AZ, 3Walter Read Army Inst. of Res., Fort mer Soviet Union countries including in the South Caucasus and Central Asia.
Detrick, MD, 4Translational Genomics Res. Inst., Phoenix, AZ. To better understand the epidemiological and evolutionary relationships of B.
Background: Bacillus anthracis, the causative agent of anthrax, is a proven bi- melitensis strains isolated in Azerbaijan, Multiple-Locus Variable number tan-
ological weapon. Very little is known about the genetic diversity of B. an- dem repeat Analysis (MVLA) was conducted using eight selected markers
thracis endemic to Georgia. To address this knowledge gap, we (MLVA-8). Methods: 25 B. melitensis isolates collected in Azerbaijan and
phylogenetically characterized 92 Georgian B. anthracis isolates based on three reference control strains were examined in this study. Genomic DNAs
Multiple-Locus Variable-Number Tandem Repeat Analysis (MLVA 15) and Sin- were extracted and used as templates for a multiplex PCR to amplify eight
gle Nucleotide Polymorphisms (SNP) to place them into a global phylogeo- Variable Number Tandem Repeat (VNTR) loci. The sizes of PCR products for
graphic context. Results: We discovered two very distinct and relatively each locus were determined using Beckman Coulter CEQ capillary sequencer.
distant populations of B. anthracis: A.Br.008/009—Transeurasia and BioNumerics 6.1 was used for cluster analysis of MVLA patterns. Results: In
A.Br.013/015 groups—Australian94 across the Georgian landscape. Se- most cases, B. melitensis strains from Azerbaijan clustered together upon
quenced Georgian B. anthracis isolate belonged to the A.Br.013/015 clade. based on MLVA-8 when compared with previously determined genotypes

62 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

from Georgia and Kazakhstan (human and animal isolates), though some All the isolates were resistant to one or multiple antibiotics, but the highest
strains from Azerbaijan shared identical MVLA-8 patterns with strains from resistance of 91.1% was to tetracycline and nine different resistance patterns
Kazakhstan. Strains from Georgia were genetically distinct from strains of the were observed among the isolates. Indiscriminate antibiotics usage in live-
other two countries. The Bruce21 locus was the most conserved locus across stock predisposes meat consumers to risks of antibiotic resistant Escherichia
the isolates compared, while the Bruce3, Bruce20 and Bruce28 loci showed coli O157:H7 in southwest Nigeria. Regulatory control of antibiotics usage in
higher discriminatory power to differentiate strains from different geographic livestock production, meat hygiene and pharmaco-epidemiological surveil-
locations and sources. Conclusions: This MVLA-8 subtyping approach proved lance in food animals is hereby recommended to ensure consumer safety.
to be a generally useful tool to distinguish B. melitensis strains from differ-
ent geographic origins toward source attribution. However, with the excep-
tions already evident in this small data set, the possibility of distant 173 (B)
disseminations of B. melitensis subpopluations must be considered. Further Contribution of OxyR to Burkholderia mallei Survival and Viru-
resolution may be obtained by including additional VNTR loci. lence In Vitro and In Vivo
D. A. Revelli, J. Boylan, F. Gherardini; Rocky Mountain Lab./NIAID/NIH,
171 (B) Hamilton, MT.
Background: Burkholderia mallei, the causative agent of glanders, is classi-
Rapid Single Nucleotide Repeat Sequence-Based Detection and fied as a Category B Bioterrorism Agent. B. mallei is a facultative intracellu-
Identification of B. anthracis Using Pyrosequencing Technology lar pathogen that replicates in a number of eukaryotic cell types, including
K. R. Hahn, E. Valle, M. Thomas, T. Janzen, M. Shields, N. Goji, K. Amoako; macrophage. Reactive oxygen and nitrogen species are important for the in-
Canadian Food Inspection Agency, Lethbridge, Canada. nate immune response and contribute to the clearance of B. mallei from in-
Bacillus anthracis is a biothreat agent with potential use in foodborne bioter- fected macrophages. However, the regulatory mechanisms for protection
rorism and early detection and identification is paramount to a successful against reactive oxygen and nitrogen species in B. mallei are poorly under-
mitigation strategy. Even though several technology platforms have been de- stood. The gene encoding the oxidative stress regulator, OxyR, is involved in
veloped for the detection and strain identification of B. anthracis, these meth- protecting a number of organisms against oxidative challenge. B. mallei en-
ods do not fully discriminate between outbreak strains due to the highly codes an oxyR homologue that may play an important role in protection
clonal nature of B. anthracis. Single Nucleotide Repeats (SNRs) are stretches against oxidative stress. To assess the role of OxyR in the survival of B. mallei
of single nucleotides repeated anywhere from 6 and up to 30 times in the intracellularly, we constructed an oxyR mutant. Methods: Mutants in B. mallei
genome of B. anthracis and can be used as genetic markers to genotype ATCC23344 were generated through insertional inactivation by transfer of
strains in the case of a disease outbreak. SNRs are susceptible to homoplasy, the plasmid pZSV. Sensitivity to reactive oxygen species was performed by
and thus are useful for detection and identification of anthrax. Pyrose- disc diffusion assay using 100 μM hydrogen peroxide. Intracellular survival in
quencing technology is a relatively rapid and inexpensive, easy-to-use murine J774.1 and human macrophage was assessed using a kanamycin pro-
method for sequence based detection. Results are generated in a fraction of tection assay. For in vivo experiments, 6-8 weeks old, BALB/c female mice
the time it takes to produce standard sequence reads. Here we report a novel were anaesthetized and inoculated intratracheally. An inoculum of 6x105 cfu
application of pyrosequencing technology for the rapid analysis of SNR re- was used to infect groups of 5 mice with wild-type or ΔoxyR B. mallei strains.
gions as a detection and identification tool for B. anthracis. Five SNR targets PBS was used as a mock infection control. Results: The ΔoxyR mutant was
specific for B. anthracis with repeats of 7-10 bases were analysed using sev- sensitive to hydrogen peroxide compared to the wild-type B. mallei at a con-
eral strains from disease outbreaks. The results indicate that this is a repro- centration of 100 μM. OxyR-deficient B. mallei did not replicate in either
ducible method for confirmation of SNR number and can potentially be used J774.1 or human macrophages. All wild-type infected mice presented with
to rapidly detect and discriminate between B. anthracis isolates during dis- late stage disease by day 5; in contrast 4 out of 5 mice infected with the
ease outbreaks. ΔoxyR mutant survived. Conclusion: Inactivation of oxyR increased sensitiv-
ity of B. mallei to hydrogen peroxide in vitro, demonstrating its protective
role against reactive oxygen species. The oxyR mutant was also attenuated
172 (B) for virulence in macrophages and in mice using an acute model of infection.
Beef and Chicken Contamination with Resistant Escherichial Collectively, these data reveal an important role for OxyR in B. mallei patho-
coli O157 from City Abattoirs in Southwest Nigeria genesis.
O. Olatoye, A. E. Amosun; Univ. of Ibadan, Ibadan, Nigeria.
This study investigated the contamination of meat from cattle and chicken
slaughtered for human consumption with E. coli 0157:H7 at the metropolitan
174 (B)
abattoirs and slaughtered slabs of selected poultry farms in Lagos and Growth Inhibition of Burkholderia pseudomallei by Onion Iso-
Ibadan, Nigeria. The aim was to compare the prevalence and antibiotic sus- lates of Burkholderia cepacia
ceptibility patterns across the different locations and climatic seasons. The J. M. Inamine, T. M. Eckstein; Colorado State Univ., Fort Collins, CO.
organism was isolated by cultural method using selective media and con- Burkholderia pseudomallei (BP) is the causative agent of melioidosis, an
firmed serologically using latex agglutination kits (OxoidR UK). Antibiotic sus- emerging infectious disease that is endemic to Southeast Asia and Northern
ceptibility to ten antimicrobial agents was performed by disc diffusion Australia. It is a major biodefense concern because of its potential use as a
method using commercial Gram negative discs (AbtekR). Out of 800 meat bioterror agent along with its inherent antibiotic resistance. Although the
samples collected, the overall prevalence of 17.1% (comprising of 19.8% and ecological niche of BP hasn’t been clearly identified, it is usually described as
14.5% of beef and chicken respectively) was obtained. The prevalence of E. an environmental organism. Given that Burkholderia species isolated from
coli O157:H7 in beef from Ibadan and Lagos were 28.5% and 11.0%, while soil and plants can produce bacteriocins, bacteriophages and anti-biologics,
those of chicken from Ibadan and Lagos markets were 13.0% and 14.0%, and we addressed the hypothesis that plant isolates of Burkholderia can inhibit
from Ibadan and Lagos farms were 18.0% and 13.0% respectively. The preva- the growth of BP. Such inhibitors could provide a new approach to the de-
lence of E. coli O157 was significantly higher in beef compared to chicken velopment of anti-BP agents. In this study, plant isolates of Burkholderia (pro-
(p<0.05), while during wet season, contamination of beef was also higher vided by Dr. Jan Leach, Colorado State University) were screened by agar
than in dry and significantly higher in beef from Ibadan than Lagos abattoir. diffusion for their ability to inhibit the growth of B. pseudomallei strain Bp82

BIODEFENSE PROGRAM & ABSTRACTS 63


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

(provided by Dr. Herbert Schweizer, Colorado State University). In brief, eight identification. The aim was to test the broad-range microbial DNA extraction
plant isolates were individually cross-streaked with Bp82 on LB and Nutrient capability of the molecular analytic kit, Universal Microbe Detection™, using
Agar plates (supplemented with 0.6 mM adenine and thiamine because Bp82 a variety of clinical material coming in for routine molecular analysis. Meth-
is a delta-pur M mutant), and incubated at room temperature for two days. ods: Specimens originated from patients under suspect of an infection. Sam-
None of the four Burkholderia andropogonis or two Burkholderia gladioli ples were processed as advised (Molzym, Bremen, Germany). Microbial DNA
strains showed any inhibitory activity against Bp82. However, the two Burk- was detected using universal rRNA gene PCR assays. Amplicons were se-
holderia cepacia strains completely inhibited the growth of Bp82 as evi- quenced and BLAST-analyzed to identify the organisms. Results: In total, 430
denced by clear zones of inhibition. Both of these strains were isolated from clinical specimens were analyzed, including blood, synovia, swabs (wounds,
onion, and both produce a bright yellow, diffusible pigment when grown on prostheses, pus, bones), CSF, and heart valve/aortal tissues. Among 186 pos-
LB or Nutrient Agar plates. Work is in progress to see if the pigment is a key itive samples, 95 showed typical pathogens (e.g., S. aureus, E. faecalis/fae-
inhibitor, as well as determine the chemical nature of the inhibitor(s). Inves- cium, E. coli, C. albicans) and 91 opportunistic strains (e.g., CoNS, viridans
tigators have recently shown that human isolates of B. cepacia can produce streptococci, Moraxella osloensis). Rare etiologies comprised bacterial
bacteriocin-like inhibitors active against other clinical B. cepacia complex pathogens, among them Bacteroides fragilis, Bartonella quintana, Borrelia
strains, while another group reported that Burkholderia multivorans strain graninii, Coxiella burnetii, Listeria monocytogenes, Providencia stuartii, and
NKI379 antagonizes the growth of BP in soil; however, to our knowledge this Tropheryma whippleii, and fungal pathogens (non-albicans Candida, black
is the first report of BP inhibition by B. cepacia plant isolates. yeast). One blood sample from a malaria patient was positive in a specific
assay for the etiological agent, Plasmodium falciparum. Conclusion: The pre-
analytical processing comprises the removal of human DNA, concentration
175 (D) and enzymatic lysis of microbes, and DNA extraction. The data from randomly
Quality Assurance for Select Agents: An Australian Approach collected, diverse clinical material indicated that a broad range of microor-
T. Theis1, D. Liu1, J. Gray1, P. Roffey2, W. Rawlinson3,1; 1RCPA BioSecurity QAP, ganisms was lysed, including Gram-positive and Gram-negative bacteria,
Surry Hills, Australia, 2Australian Federal Police, Weston, Australia, 3SEALS yeasts and other fungi, as well as a protist. The supply of DNA from diverse
Microbiol. Prince of Wales Hosp., Randwick, Australia. microbial taxons to the PCR analysis greatly supports the detection and pre-
In 2008 the Australian Government established a two tiered list of Security cise molecular identification of common and rare etiologies.
Sensitive Biological Agents (SSBA). Tier 1 agents include Bacillus anthracis,
Yersinia pestis, botulinum toxin and ricin, and pose the highest security risk
to Australia, while Tier 2 agents such as Clostridium botulinum, Salmonella
177 (D)
typhi, and Francisella tularensis, pose a high security risk. The RCPA BioSe- WITHDRAWN
curity QAP has been established to provide a quality assurance and profi-
ciency testing program (PTP) for Tier 1 and Tier 2 SSBAs. Participants in this
PTP require a regular supply of testing material from the RCPA BioSecurity
178 (D)
QAP to assess their laboratory competency in detecting SSBAs. The dispatch Laboratory Surveillance of Tick Born Causing Agents and Other
of virulent material has significant issues for regulation, potentially for na- Relevant Pathogens in the State of Maryland
tional security. Therefore, RCPA BioSecurity QAP developed a set of surro- M. Dembele1, L. Trivedi2, T. Lawson2, S. Montgomery2, M. P. Carlos2,1; 1Johns
gate agents that can be shipped and handled safely, while displaying testing Hopkins Sch. of Publ. Hlth., Baltimore, MD, 2Maryland Dept. of Hlth. and Men-
characteristics identical to Tier 1 and Tier 2 SSBAs. The RCPA BioSecurity QAP tal Hygiene Lab. Admin., Baltimore, MD.
has developed proficiency testing surveys for the SSBAs B. anthracis, Y. Background: The purpose of study is to analyze the immunological test re-
pestis, C. botulinum, botulinum toxin and ricin. The non-virulent surrogates sults from suspected Lyme disease (LD), Ehrlichiosis (HME and HGE), Tu-
include carefully selected and evaluated bacterial strains that are similar to laremia, Rocky Mountain Spotted Fever (RMSF), Babesiosis, Coxiella burnetii,
B. anthracis, Y. pestis, and C. botulinum. We have established a comprehen- and Yersinia pestis specimens submitted to the Maryland Department of
sive plasmid library which includes more than 20 of the most common gene Health and Mental Hygiene (DHMH) Laboratories Administration Division of
targets used in nucleic acid testing of SSBAs. The proficiency testing surveys Virology and Immunology. This analysis will provide a better understanding
for botulinum toxin and ricin were developed using inactivated material, or of the occurrence of emerging and re-emerging tick and rodent born diseases
non-toxic protein domains. We were able to show that immunological tests in Maryland. Methods: From 2008 to 2011, de-identified data (N= 3578) were
performed by reference laboratories yielded results comparable to those ob- obtained from the MD DHMH Laboratories Administration Laboratories In-
tained with holotoxins, making them ideal surrogates to be used in a profi- formation Management System. Tests were performed at Division of Virology
ciency test. All specimens developed for these proficiency testing surveys and Immunology, however tests for Babesiosis, C. burnetii and Y. pestis were
can be handled at Biosafety level 2. This allows for training and education of sent out to CDC. Immunological methods included: immunofluorescent assay
laboratory staff with minimal risk of infection. The educational component of for LD, HME, HGE, RMSF, Babesiosis and C. burnetii; western blot for LD con-
this proficiency testing program is further strengthened by specimen-free on- firmation; agglutination test for Tularemia; and fluorescent antibody test for
line modules. Y. pestis. Results: Lyme disease was the most prevalent disease with male
cases accounting for 71.4 % and the 25-59 years old age group for 45%.
RMSF was the second most predominant disease, with male cases and the
176 (D) 25-59 years old age group accounting for the majority of the detected cases
Broad-Range Microbial DNA Isolation from Clinical Specimens with 58.6 % and 69%, respectively. There were 3 positive cases of C. burnetii
for Universal PCR Diagnosis and Babesiosis. Ehrlichiosis was diagnosed in 47.6% of the female popula-
C. Disqué1, M. Linow1, N. Murphy2; 1Molzym GmbH & Co. KG, Bremen, Ger- tion, and 38.1% in the 25-59 and > 60 years old age groups. No case of Tu-
many, 2CaerusBio Inc., Downingtown, PA. laremia was detected. Conclusions: Laboratory diagnosis can alleviate the
Objective: Molecular analysis has proven to be a valuable tool in the timely burden of an outbreak by the immediate identification of early exposure
diagnosis of the etiological agents of infectious diseases such as sepsis, en- cases, and confirmation allows appropriate treatment. Moreover, a compre-
docarditis and meningitis. DNA extraction of a broad range of microorgan- hensive laboratory surveillance of emerging and re-emerging pathogens in
isms is a key issue for sensitive detection and precise sequence-based the most susceptible populations is a valuable tool in public health preven-
tion and control.

64 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

good candidates for improved EF detection by MS. Performance of the EF


179 (D) method for these new mAb’s were compared to anti-EF mAb’s described pre-
Development of Single Domain Antibodies for Bacillus an- viously. The EF method is specific, fast (≤4h) and sensitive with a lower limit
thracis Spore Proteins of detection of 16 fg/mL (180 zmoles/mL) for 200 µl sample. The EF-MS
S. A. Walper1, P. Lee2, J. Liu3, G. Anderson3, E. Goldman3; 1Natl. Academy of method is the most sensitive reported for quantitative detection of any an-
Sci., Washington, DC, 2Nova Res. Inc, Alexandria, VA, 3Naval Res. Lab., Wash- thrax toxin. Combined with our MALDI-TOF-MS method for LF, the EF method
ington, DC. expands our capability to detect and measure both toxins in early asympto-
Background: The spores of Bacillus anthracis are considered a potential matic infection and throughout treatment.
bioweapon of high concern. Assays are needed to rapidly analyze samples at
the point of concern which is often far removed from a laboratory facility. The
high affinity and stability exhibited by single domain antibodies (sdAb) from 181 (E)
camelids make these elements suitable for integration into assays for use in Simultaneous Detection of Ricin and Abrin DNA by Real-Time PCR
such environments. Targeting proteins specific to B. anthracis spores will I. Mossbrugger, E. Felder, R. Wölfel; Bundeswehr Inst. of Microbiol., Munich,
allow for the development of highly specific assays capable of rapidly de- Germany.
tecting the pathogen. Methods: Two llamas were immunized with six protein Ricin is one of the most potent plant toxins known, and the castor plant from
constituents of the B. anthracis spore. A sdAb library was constructed and which it is derived, Ricinus. The harvesting of castor beans exceeds one mil-
enrichment of antigen binders accomplished using phage display. Unique lion tons annually, and the toxin may be easily obtained in high yield from
clones were characterized for binding kinetics to the antigen and ability to the castor seeds using rather simple technology. As a result, ricin is a potent
detect spore material from B. anthracis. The melting temperature and ability and available toxin for criminal or terrorist acts. Abrin is a small toxic protein
to refold following thermal denaturation was assessed using circular dichro- similar to ricin, found in the beans of the Jequirity plant (Abrus precatorius),
ism and ELISAs. Results: Representative sdAbs showed nanomolar or better and shares structural and functional principles similar to those of ricin. As
affinities for target antigens as determined by SPR. SdAb targeting BclA and the production of highly purified ricin or abrin toxin is labour intensive and re-
gerQ antigens were able to detect spore material from B. anthracis in ELISAs quires sophisticated lab equipment, it is more likely that crude, rather than
and did not recognize vegetative cell samples. Those sdAbs targeting BclA purified, extracts would be used by non-governmental perpetrators. In such
exhibited excellent specificity when tested with spores of other Bacillus crude extracts remaining nucleic acids provide appropriate surrogate ana-
species. SdAb directed towards several other spore proteins however, were lytes for the toxins themselves and allow the PCR detection and identifica-
not able to detect spore material, potentially due to a low abundance of the tion of abrin or ricin. As part of an effort to provide protocols for the detection
target antigen. Melting points ranged from 58 - 72 °C, and many of the sdAbs of various biological toxins we developed a real-time PCR assay for simulta-
were successful in antigen detection following iterations of heating and cool- neous detection of ricin and abrin nucleic acid. Oligonucleotide primers were
ing. Conclusions: We successfully isolated sdAbs to six proteins of the B. an- designed manually based on an alignment of R. communis, A. precatorius
thracis spore, all with affinities in the nanomolar range. Only sdAbs targeting and other lectine-producing plant genome information. Two primer pairs were
the BclA and gerQ antigens were able to directly detect spore material in identified to be efficient for amplification of lectine-specific target sequences.
ELISAs . These sdAbs may potentially serve in combination with other de- The differentiation between both ricin and abrin was enabled by toxin-spe-
tection elements for the development of fieldable assays for the detection cific, dye-labelled 5’exonuclase hybridizations probes. The resulting assay
and differentiation of B. anthracis spores. reliably amplified the targeted region of ricin and abrin DNA. To determine
the analytical sensitivity of the assay, cloned and quantified fragments of ricin
and abrin DNA were amplified in limiting log10 dilution series. The 95% de-
180 (D) tection limit of the real-time PCR was 3.0 genome copies per reaction for the
Zeptomole/ML Quantification of Anthrax Edema Factor by LC- detection of ricin and 1.2 genome copies per reaction for the detection of
ESI-MS/MS: Activity Enhancement by Monoclonal Antibodies abrin. In summary, the novel ricin and abrin DNA real-time PCR assay pro-
R. C. Lins1, Z. Kuklenyik2, C. P. Quinn2, K. Bedi2, J. Goldstein2, A. Lyons2, M. Gal- vides a rapid and sensitive method for the detection and identification of ricin
legos2, U. Ansari3, R. Pitts3, D. Bagarozzi3, K. Isbell1, C. E. Leysath4, Z. Chen4, and abrin nucleic acid. It may prove useful for the investigation of deliberate
S. H. Leppla4, J. R. Barr2, A. Boyer2; 1Battelle CDC, Atlanta, GA, 2Ctr. for Disease castor or Jequirity bean contaminations.
Control and Prevention, Atlanta, GA, 3Ctr. for Disease Control and Prevention
Battelle, Atlanta, GA, 4NIH, Bethesda, MD.
Anthrax is caused by the pathogenic bacterium Bacillus anthracis. It secretes
182 (E)
two major exoproteins: lethal factor and edema factor (EF) that combined Low-Cost, Highly Efficient Airborne Pathogen Collection and
with protective antigen form lethal toxin and edema toxin respectively. EF is Detection System for Field Use
a highly active calmodulin-dependent adenylyl cyclase (AC) which converts J. Gordon1,2, P. Gandhi1; 1Inspirotec LLC, Chicago, IL, 2Northwestern Univ.,
ATP to cyclic AMP (cAMP) causing edema and immune dysfunction in anthrax. Evanston, IL.
We have developed a high sensitivity AC activity-based method for quantify- Efficient and cost effective collection of airborne pathogens, and subsequent
ing EF. Analytic sensitivity is in part dependent on prior purification of EF with detection, is a critical need for biodefense. We describe use of an ionic
monoclonal antibodies (mAbs). Here we report screening of 12 murine anti- propulsion device as a platform for air sampling. A plasma is generated at a
EF mAbs for their ability to bind and modulate EF. The mAbs were tested for high voltage wire. The ions generated are propelled by a geometric arrange-
1) reactivity with EF by western blot (WB), 2) interference/enhancement of EF ment of electrodes, creating an airflow. Particulates acquire an induced
catalytic activity, 3) immunoprecipitation (IP) of EF and 4) combined EF IP/ac- charge and are deposited on plate electrodes. From electrical field theory,
tivity. For EF-MS mAbs were immobilized on magnetic protein-G or tosyl-ac- non-conductive elements will not affect the electrostatic field. We evaluated
tivated beads to purify and concentrate bound EF which was then reacted the use of non-conductive textile envelopes on the electrodes, for collection
with ATP/calcium/calmodulin to generate cAMP, subsequently quantified by of samples for analysis. We used cat allergen Fel d1 and ELISA assay as a
LC-ESI-MS/MS. All 12 mAbs were reactive for EF in WB. Three mAbs enhanced model system. Possible effects of the textile on the air-flow pattern and ve-
and two highly neutralized EF activity. Combined capture/activity was almost locity were investigated with a smoke gun and an anemometer. Captured al-
doubled for two mAbs. In summary, two of the anti-EF mAbs identified were lergen was measured with an ELISA. The recovery of allergen extracted from

BIODEFENSE PROGRAM & ABSTRACTS 65


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

textile was as efficient as with a published swabbing procedure. Of materials


evaluated, silk gave the most efficient capture of the allergen. Diluent from
184 (E)
the ELISA was effective as an extraction medium. Extractables from the silk Development of Multiplex Rapid Tests Allowing Warfare Agents
had no effect on the ELISA performance. The detection limit was less than 5 Detection
pg/M3. We are further developing the system to include a specialized capture T. Berthelot, J. Credou, H. Volland; CEA, Gif sur Yvette, France.
electrode, which will functional as a potential well and trap the sample on a Background: Biological warfare agents are inconspicuous, their detection is
small area. The small area will permit collection of the sample in a small vol- difficult and, within the limit of our current knowledge, far from being in-
ume with consequent increase in sensitivity. The sample collection means stantaneous. This point is even more crucial because the effects of these
may be optimized for delivering analyte to any relevant rapid test. The tech- agents, dramatically effective, are insidious. The development of powerful
nology will be developed into a cost-effective, handheld device for use in the systems of detection and allowing identification of these agents, as early as
field by first responders and military personnel. their dispersion, is therefore crucial to be efficient against those. These sys-
tems will have to be easy to use, because they are susceptible to be used by
non specialists. Methods: Among the various methods of detection devel-
oped, immunochromatography, while preserving the property of specificity of
classical immunological assays, presents several advantages: fast, ease of
use, adapted to measurements on the field and very low cost. Unfortunately
classical lateral flow immunoassays could not detect several agents at the
same time in the same sample. Based on the Whitesides research group work
we have developed immunochromatographic tests which could allow the de-
tection and identification of up to 16 different agents simultaneously in the
same sample. This project implies the development of chemistry which en-
ables both the binding molecule on paper and the conservation of their bio-
logical activity. We also compare different kind of colorimetric labels linked to
monoclonal antibodies. Results: The developed fluidic system constituted
by paper allows the fraction of one sample in 16 isolate spots. We are also
able to bind one different biological molecule, like monoclonal antibody, or
chemical component to each spot. Moreover specific signals visually detected
were associated to the presence of molecules in the sample. Conclusions:
Our device is suitable for a field use and authorizes a multi detection/iden-
tification of up to 16 agents. Currently we work on different parameters (flow
183 (E) rate, label, monoclonal antibody density on the spot) in order to increase the
Evaluation of Automated and Manual Commercial Nucleic Acid limit of detection.
Extraction Methods for Detection of Ricinus communis DNA by
Real-Time PCR
A. S. Hutchins1, M. Astwood2, J. R. Saah1, L. A. Dauphin2; 1North Carolina State
185 (E)
Lab. of Publ. Hlth., Raleigh, NC, 2CDC, Atlanta, GA. Validation of Unique Signature Discovery in Select Agents and
Background: The castor bean plant, Ricinus communis, produces ricin toxin, an Emerging Pathogen
which has been used as a biological weapon in the past. Ricin is classified by B. J. Haley1, E. Taviani1, A. Chen1, A. Phillippy2, A. Huq1,3, R. R. Colwell1,4,5, I.
the CDC as a Category B critical biological agent. Currently, diagnostic labo- Knight3,6; 1Maryland Pathogen Res. Inst., College Park, MD, 2Ctr. for Bioin-
ratories use real-time PCR assays for rapid detection of R. communis DNA in formatics and Computational Biology, College Park, MD, 3Maryland Inst. for
environmental samples, which is an indicator that ricin toxin could be pres- Applied Environmental Hlth., College Park, MD, 4Univ. of Maryland Inst. for
ent. DNA extraction methods can impact the sensitivity of real-time PCR as- Advanced Computer Studies, College Park, MD, 5CosmosID, College Park,
says; therefore, determining the optimal extraction method is important for MD, 6Canon U.S. Life Sci., Inc., Rockville, MD.
effective detection of R. communis DNA. Methods: This study evaluated com- The Insignia informatics pipeline (http://insignia.cbcb.umd.edu/), which rap-
mercial nucleic acid extraction methods for their efficiency in isolating R. com- idly detects unique DNA sequences of bacteria, viruses, and bacteriophage
munis DNA from castor bean preparations. Four extraction methods, against a background of RefSeq sequences, was used to determine unique
including two automated methods, the MagNA Pure Compact and MagNA genomic signatures of three select bacterial agents (Yersinia pestis, Brucella
Pure LC, and two manual methods, the QIAamp DNA Blood Mini Kit and the abortus, and Clostridium botulinum) and one emerging bacterial pathogen
Masterpure Complete DNA and RNA Purification Kit, were compared. Castor (Campylobacter jejuni). These species-specific signatures were validated by
beans were homogenized in phosphate buffered saline, and the extraction TaqMan Real-Time PCR assays. For Y. pestis, C. botulinum, and C. jejuni; 43,
methods were performed in triplicate upon dilutions of the castor bean ho- 54, and 82 unique sequences of at least 100 nucleotides in length were dis-
mogenates. The effectiveness of nucleic acid extraction with the four meth- covered. B. abortus signatures were too short for primer/probe generation,
ods was determined by comparisons of cycle-threshold (CT) values with a R. so at most 100 bp of non-unique sequence was permitted between the
communis-specific TaqMan® real-time PCR assay with an internal positive shorter signature sequences, from which the primers and probes were se-
control (IPC), as well as comparisons of DNA yields and DNA purity. Re- lected. When validated against a DNA from Y. pestis and its near-neighbors,
sults/Conclusion: Although the four extraction methods yielded DNA that re- 42 primer and probe sets were discriminatory between Y. pestis and near-
sulted in differences in real-time PCR CT values; the limit of detection was neighbors, while 1 set showed late amplification. All primer and probe sets
equivalent for all extracts. There was no evidence of PCR inhibition for any of were unique for Clostridium dificile when compared with the near neighbors
the extraction methods as measured by the IPC assay. The results from this C. glycolicum, C. thermocellum, and C. proteoclastium. All primer and probe
study indicate that diagnostic laboratories have the option to select from au- sets generated amplification for all Campylobacter jejuni strains except for 2
tomated or manual extraction methods without loss of sensitivity when using which did not show amplification for a strain whose genome had yet to be
real-time PCR for detection of R. communis DNA. sequenced. Eleven other primer and probe sets did amplify regions of four

66 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

near neighbors whose genomes were not yet deposited in RefSeq. Seven phylogenetic trees, assign annotations to new genomes, and store results
primer and probe sets from non-overlapping regions of one gene sequence from any search or analysis within a personal Workbench space on the ViPR
were able to discriminate between B. abortus and the near neighbors B. suis server for future use. We expect our current (and future) suite of tools to fur-
and B. melitensis. Results of this study validate the ability of the Insignia in- ther assist viral researchers in developing diagnostics, prophylactics, and/or
formatics pipeline to generate a suite of unique genomic DNA targets for the therapeutics for viruses that are pathogenic to humans.
detection of bacteria, including select agents.

188 (F)
186 (E) Effects on Host Responses of Air-Borne and Aerosolized Rab-
Novel Surface Chemistry for the Development of Multiplex bitpox Infections in New Zealand Rabbits
Immunochromatographic Tests L. DaSilva1, A. Porter1, A. Nalca1, R. Settlage2, C. L. Galindo3, L. McIver2, K.
J. Credou, H. Volland, T. Berthelot; CEA, Gif sur Yvette, France. McMahon2; 1USAMRIID, Frederick, MD, 2Virginia Bioinformatics Inst. at Vir-
Background: The development of new chromatographic format as lab-on- ginia Polytechnic Inst., Blacksburg, VA, 3Eagle Eye Data Analysis, Houston,
paper by the Withesides group [1] involves the replacement of conventional TX.
material (nitrocellulose) used for lateral flow immunoassays by paper. This Rabbits are commonly used research animals. The understanding of the ge-
technological breakthrough imposes to develop novel surface chemistry to netic structure of the rabbit genome represents therefore an important step
functionalize paper with biomolecules without altering their biological activ- towards the full characterization of the rabbit animal model in biodefense re-
ity. Methods: Our project implies the development of chemistry which en- search. We have developed a custom microarray covering 35,058 probe set
ables both the biomolecules grafting on paper and the conservation of their identifiers representing the leporine genome to determine the effects of an
biological activity. Moreover, our process must be operated in soft conditions aerosol exposure to two doses of the rabbitpox virus in the host’s lungs. Se-
(aqueous systems, compounds and methods compatible with biological ma- quence information was obtained from the 2X coverage of the Oryctolagus cu-
terial) but also must be compatible with paper sheet format. In fact, it is well niculus genome provided by the Broad Institute. Microarrays were performed
known that cellulose is not water-soluble which implies that its chemical using RNA isolated from lungs of rabbits challenged with aerosolized rabbit-
modification is performed under harsh conditions in organic solvents. To over- pox virus (RPXV) at a low dose (165 PFU) (n=10) or at a high dose (4,171 PFU)
come these drawbacks, we chose two synthetic pathways to functionalize (n=10), and compared to environmental control rabbits (n=4), yielding sta-
paper sheets based (i) on an innovative surface chemistry, named GraftFast™ tistically significant (p<0.05) changes in gene expression in the low- and
[2, 3], and (ii) on a photochemical process. Results: By our approaches, paper high-dose exposure groups. Hierarchical clustering and phylogenic analyses
sheets have been successfully modified and bear different functionalizable of the rabbit genome array data confirmed the existence of at least three dif-
groups, as carboxyl, amino or thiol groups for example, to graft biomolecules ferent groups: non-responders, mild responders, and high responders among
by classical bioconjugate techniques. These paper modifications were per- the significant transcripts. The high responders group consisted mostly of
formed in aqueous environment and at room temperature. Conclusions: The the animals receiving the lower dose of the aerosolized RPXV. Host responses
chemical ways developed here are suitable for new immunochromatographic in the low- and high-RPXV-dose groups revealed the involvement of genes of
format as lab-on-paper. Currently, our work is focused on different parame- the cell cycle, growth and development and carbohydrate metabolism. In ad-
ters (chemical groups, linker density onto the surface…) which can play a role dition, rabbitpox produced a transcriptional response resembling humoral
in the detection limit. [1]. A.W.Martinez, et al. Analytical Chemistry, 2010, 82 responses and more so than the classical antiviral responses. Interestingly,
[2]. Mévellec, V. et al. Chem. Mater. 2007, 19, 6323-6330. [3]. Mevellec, V. et the data have also suggested an effect on RNA post-transcriptional modifi-
al. Patent n° WO2008/078052; 2006. cation genes to account for viral survival. Potential biomarkers of rabbitpox
infection were also identified, including transcripts for FLRT2, TPM3, CD14
and HP. The present work should contribute to a more comprehensive eval-
187 (F) uation of the rabbit when used as an animal model of orthopox viral disease
Virus Pathogen Resource (ViPR): An Open Bioinformatics and of other human infectious diseases as well.
Database and Analysis Resource for Human Virus Pathogen
Research
Y. Zhang1, B. Pickett1, E. Sadat1, J. Noronha1, R. B. Squires1, V. Hunt1, M. Liu2,
189 (F)
S. Kumar3, S. Zaremba3, Z. Gu3, L. Zhou3, C. Larson4, J. Dietrich3, E. Klem3, R. Analysis of Next-Generation Sequencing for Detecting Rare
H. Scheuermann1; 1Univ. of Texas Southwestern Med. Ctr., Dallas, TX, 2South- Variants in Bacillus anthracis
ern Methodist Univ., Dallas, TX, 3Northrop Grumman Hlth. Solutions, R. E. Colman1, J. Schupp1, J. Gillece1, A. Rawat1, D. M. Engelthaler1, J. Foster2,
Rockville, MD, 4Vecna Technologies, Greenbelt, MD. P. Keim2,1; 1Translational Genomics Res. Inst., Flagstaff, AZ, 2Northern Arizona
The Virus Pathogen Resource (ViPR, www.viprbrc.org) is a freely available, Univ., Flagstaff, AZ.
NIAID-funded resource providing integrated datasets and analysis tools for Next-generation sequencing is rapidly advancing capabilities for bioforen-
fourteen different virus families: Arenaviridae, Bunyaviridae, Caliciviridae, sics. Bacterial populations, whether grown in the lab or found in the envi-
Coronaviridae, Flaviviridae, Filoviridae, Hepeviridae, Herpesviridae, Paramyx- ronment, will represent a mixture of genotypes even if the population arose
oviridae, Picornaviridae, Poxviridae, Reoviridae, Rhabdoviridae and Togaviri- from a single cell. Detection and use of rare variants as a forensic diagnostic
dae. ViPR includes information from: GenBank sequence records, gene tool for identifying particular preparations is an important capability, e.g. the
annotations, strain metadata, Gene Ontology classifications, UniProtKB pro- Amerithrax investigation, and a potentially powerful application of next-gen-
tein annotations, relevant 3D protein structures from the Protein Databank eration sequencing. We have developed and synthesized plasmid constructs
(PDB), immune epitopes from the Immune Epitope Database (IEDB), clinical with engineered SNP and INDEL mutations for use in characterizing se-
and surveillance metadata and novel data derived from comparative ge- quencing error and as an internal sequencing control (ISC) in forensic analy-
nomics analysis. ViPR data can be searched and analyzed through user- sis. We Illumina sequenced mixtures of these different plasmid constructs to
friendly web interfaces for calculating: BLAST, multiple sequence alignments, extremely high coverage (~100,000X), as well as DNA from the well charac-
phylogenetic trees, single nucleotide polymorphisms, sequence feature vari- terized Ames and Sterne strains of Bacillus anthracis (~1,000X). We then cal-
ations, and metadata-driven comparative analyses. Tools also exist to: view culated average baseline error rates of 0.41% and 0.55% for plasmids and B.

BIODEFENSE PROGRAM & ABSTRACTS 67


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

anthracis, respectively. Using the plasmid standards, we characterized three data required for the detection SNPs that are present in 0.1% of a sample
factors that contribute to error: library preparation variation, multiple PCR (ultra-rare variants). We have conducted computational simulations and deep
steps on a single preparation variation, and lane to lane variation. We found sequencing experiments on Illumina’s GAIIx using Enterobacteria phage
very little variation in the average baseline error rate ranging from 0.36- phiX174 (NC_001422), Pseudomonas aeruginosa (NC_008463), and PCR am-
0.44%. We determined that the reference base differentially contributes with plified regions from Mycobacterium tuberculosis (mixed at 1:1,000 mutant
C to A, A to C, G to T, and T to G errors being 2-3 times more likely to occur to wildtype ratios). Results suggest that Next Generation Sequencing ap-
than other errors. In our sequence analysis of the Ames strain, the distribu- proaches can, in a cost acceptable manner, produce sufficient sequence data
tion of error is not Poisson distributed, indicating that error is non-random. to successfully detect ultra-rare variants. However, distinguishing true ultra-
The majority of positions have a low error rate, 87% of positions with error rare variants from false positive SNPs is a major limiting factor when the vari-
have an error of 0.5% or less; however there are notable error hotspots. We ant SNP is present at less than 1%. We have performed a detailed analysis of
have also sequenced a known artificial mixture of B. anthracis with our syn- factors contributing to false positive SNP identification. Both systematic and
thesized plasmid spiked in to serve as ISC. This enabled us to characterize random sources of errors have been identified. The systematic sources of
within-lane error rate and true variant limits of detection, increasing the con- error include: machine sequencing error, alignment algorithm errors, and bias
fidence in minor variant detection. induced by features of the reference sequence. Random sources of errors
identified as related to multiple hypotheses testing during SNP detection.
Corrections for systematic and random errors have been designed and found
190 (F) to increase the specificity (reduce the number of false positive SNPs identi-
Nested Amplification for Improved Genotyping of Coxiella fied) of ultra-rare variant detection.
burnetii
R. Vipond, A. Pepler, G. Vincent, J. Latham; Hlth. Protection Agency, Salis-
bury, United Kingdom.
192 (G)
Coxiella burnetii is one of the few biothreat agents naturally present in the UK PANACEA Broad-Spectrum Antiviral Therapeutics
and has been responsible for large outbreaks of disease as well as sporadic T. H. Rider; Massachusetts Inst. of Technology, Cambridge, MA.
cases. Diagnosis is often by serological screening for an immune response to Background: Although there is great concern over emerging and category A-
the bacterium, though direct detection by PCR early during infection is also C priority viruses, there are relatively few prophylactics or therapeutics for
possible. Because this is a zoonotic infection, linking human cases to out- these viruses, and most which do exist are highly pathogen-specific or have
breaks in animals or reservoirs of infection is important for disease manage- undesirable side effects or other disadvantages. We have developed a radi-
ment and risk analysis. The highly infectious nature of the pathogen and cally new and very broad-spectrum antiviral therapeutic/prophylactic that
intracellular lifestyle mean that genotypic analysis, in our hands at least, is has the potential to revolutionize the treatment of viral infections, including
often directly on extracted clinical or veterinary samples rather than cultured those due to emerging, category A-C, and common clinical pathogens. Our
material. Using published Multi-spacer typing (MST) and Multiple Locus Vari- Double-stranded RNA (dsRNA) Activated Caspase Oligomerizer (DRACO) ap-
able Number Tandem Repeat Analysis (MLVA) methods we found direct analy- proach selectively induces apoptosis in cells containing any viral dsRNA, rap-
sis frequently failed even with samples which were known to have pathogen idly killing infected cells without harming uninfected cells. Methods: We have
nucleic acid present by real time Polymerase Chain Reaction (PCR). Thus we produced preliminary DRACO designs and evaluated their toxicity, antiviral
designed sets of flanking primers for nested PCR reactions and adapted the efficacy, and other properties in quantitative cell assays and in mouse trials.
method to improve the sensitivity of the assay and permit genotyping of ex- Results: We have demonstrated that DRACOs are nontoxic in all 11 cell types
tracted samples without enrichment or culture of PCR-positive samples. The tested thus far, including human, monkey, and mouse cells representing nu-
nested MST and MVLA approach was applied to a range of clinical, veterinary merous tissue types. Using these cells, we have shown that DRACOS are ef-
and environmental samples. Results showed a significant increase in the fective against all 15 viruses tested thus far, including DNA and RNA viruses,
number of samples for which complete genotype profiles were obtained (from enveloped and nonenveloped viruses, human and rodent viruses, and viruses
50% up to 85%, n= 22 samples) using the nested methods compared to the that use a variety of receptors. Among the viruses against which DRACOs
original direct PCR approach. The limit of analysis, not surprisingly, correlated have proven effective in vitro are dengue hemorrhagic fever virus, multiple
to the abundance of template as determined by real time PCR, whilst there arenaviruses, and multiple bunyaviruses. In mice, we have demonstrated that
was variance between loci, a 15-100 fold increase in the lowest level of tem- DRACOs rapidly penetrate into all organs tested, persist for over 24 hours
plate which yielded a result was observed. In summary, we have shown a after each dose, and are nontoxic. We have shown that DRACOs rescue mice
nested approach improves the sensitivity of existing typing methods for C. from lethal H1N1 influenza and Amapari arenavirus challenges, and we are
burnetii which, given the difficulty of working with this particular organism, conducting efficacy trials against dengue in mice. Conclusions: We have
is of great utility in genotypic analysis of a variety of sample types which may demonstrated that DRACOs are nontoxic and effective against a broad spec-
be encountered during outbreak investigations or routine surveillance. trum of viruses. We hope to optimize our DRACO designs and demonstrate
them against additional viruses and in additional animal models. This work
should greatly advance DRACOs toward ultimate utility as safe, broad-spec-
191 (F) trum therapeutics/prophylactics for priority and emerging viral pathogens,
Utility and Challenges Associated with Use of Next Generation filling a large gap in existing therapeutics.
Sequencing in Ultra-Rare Variant Detection
H. Koshinsky1, V. Fofanov2, J. Liu2; 1Eureka Genomics, Hercules, CA, 2Eureka
Genomics, Houston, TX.
Presence of rare variants in bacterial samples could act as a sample’s fin-
gerprint and be important in investigation and prosecution of bioterrorism
attacks or attempts. Existing PCR-based approaches cannot detect unknown
variants if present in less than 10% of the sample. The recent advances in
Next Generation Sequencing technologies may have sufficient throughput to
make feasible the cost-acceptable generation of the amount of sequence

68 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

of annotated small molecule inhibitors for their ability to block intracellular


193 (G) growth of C. burnetii, B. abortus and R. conorii. Using this inhibitor screening
Efficacy of a Doxycycline Treatment Regimen in Rhesus assay, we have identified host functions that likely play a role in regulating in-
Macaques Aerosol Challenged with Brucella melitensis tracellular growth of C. burnetii and rickettsial species. Experiments are un-
L. Henning, E. McGuinness, D. Pak, S. Miller, C. Briscoe, M. Anderson, R. War- derway to elucidate the molecular mechanisms underlying this phenotype.
ren; Battelle, Columbus, OH.
Background: The non-human primate inhalational brucellosis model was pre-
viously characterized at Battelle. This large animal model was used to assess 195 (G)
the efficacy of a doxycycline treatment regimen when administered to rhe- Restoring Antibiotic Sensitivity with Antisense Inhibitors of
sus macaques (RMs) therapeutically after aerosol challenge with B. meliten- Gene Expression
sis. Methods: Fourteen RMs were aerosol challenged with a targeted dose P. L. Iversen1,2, D. V. Mourich1,2, F. Schnell1,2, B. Geller1,2; 1AVI BioPharma, Inc.,
of 2.50E+05 cfu B. melitensis. Beginning 21 days post-challenge, doxycycline Bothell, WA, 2AVI BioPharma, Bothell, WA.
was administered orally to 10 RMs. Treatment was continued for 14 consec- The objective of the current study was to demonstrate a reduction in the min-
utive days (BID). Body temperature, blood culture, splenomegaly and tissue imum inhibitory concentration (MIC) of aminoglycosides, beta-lactams, chlo-
burden were assessed. In addition, biomarkers were evaluated by the mi- ramphenicol, tetracycline and sulfonamides by inhibiting bacterial genes
croagglutination assay and ELISAs (IgG, IgM, and neopterin) throughout the linked to their drug resistance phenotype. Phosphorodiamidate morpholino
in-life period. Results: Aerosol challenged RMs developed fevers and sero- oligomers (PMOs) are short synthetic DNA mimics that inhibit gene expres-
converted (≥ 1:80) by Day 21 post-challenge. Ninety-three percent (13/14) of sion by binding specifically to mRNA with complementary base sequences.
challenged animals were bacteremic by enriched blood culture on Day 21. Previously, we synthesized peptide conjugated PMOs (PPMO) targeted to es-
On Day 28, while 9/10 treated animals were negative, all untreated animals sential genes of bacteria, and found that these PPMOs can be potent bacte-
remained bacteremic. While treated animals had either no tissue burden ricidal compounds. Escherichia coli SMS-3-5 represents an excellent
(6/10) or below the limit of quantitation (4/10) by Day 37 post-challenge, all organism to evaluate PPMO inhibitors of gene expression linked to antibac-
untreated animals had a tissue burden in the lung, liver, spleen and/or kid- terial resistance because it is resistant to a record number of antimicrobial
ney. In addition, splenomegaly resolved for most treated animals one to two compounds from all major classes. We evaluated 11 different PPMOs in liq-
weeks after treatment was initiated, while it was still visualized by ultrasound uid cultures with a range of dilutions of the selected antibiotics to establish
in untreated animals. Conclusion: The oral doxycycline treatment regimen the minimum inhibitory concentration (MIC indicated as µg/mL) over a range
appears to be effective against B. melitensis infection in RMs. The results of concentrations of PPMO from 0.5 up to 32 uM. A scrambled PPMO se-
from this study support the use of the inhalational brucellosis model for test- quence had no influence on the MIC value for at least three different antibi-
ing therapeutics. This work was supported by the Transformational Medical otics. Antibiotic sensitivity was nearly completely restored for streptomycin
Technologies program through CBRNIAC contract number SP0700-00-D- by PPMOs targeting strA and strB. Less effective restoration of sensitivity was
3180, Task CB-09-0018 Task 769 from the Department of Defense Chemical observed for streptomycin with PPMOs targeting aadA and aph genes. These
and Biological Defense program through the Defense Threat Reduction observations further indicate the PPMO alone is not an effective inhibitor of
Agency (DTRA). bacterial growth. The observations may suggest that both aadA and aph con-
tribute less to streptomycin resistance than strA or strB, or that these two
target genes may not have been effectively inhibited. indicating a need for
194 (G) continued effort to identify improved targets. The restoration of sensitivity
Host-Directed Antimicrobial Therapeutics for Intracellular to ampicillin and chloramphenicol was also substantial while restoration of
Bacterial Pathogens sensitivity to tetracycline and trimethoprim was minimal. This results show
A. Levi1, D. Arvans2, P. Robins2, L. P. Potluri1, B. Heaton1,2, S. Riley1,2, S. that sensitivity to antibiotics can be modulated and restored by PPMOs, and
Crosson1,2, J. Martinez1,2, J. E. Gabay1, H. A. Shuman2,1; 1Univ. of Chicago, that variations in PPMO design can greatly affect the results.
Chicago, IL, 2Howard Taylor Ricketts Lab., Lemont, IL.
Although antibiotic therapy is often successful, it is becoming increasingly
clear that alternatives to conventional pathogen-directed antibiotic therapy 196 (G)
must be developed in the face of increased antibiotic resistance. We aim to Post-Exposure Administration of a Monoclonal Antibody Cock-
identify human genes that encode functions required for successful intracel- tail Is Protective in Orthopoxvirus-Infected Mice and Cynomol-
lular infection by a variety of bacterial pathogens. We hypothesize that novel gus Macaques
host-directed antimicrobial therapies can be developed by targeting specific J. L. Nordstrom1, G. J. Rainey1, H. Li1, P. Silvera2, F. Koide2, M. Buller3, J.
host pathways used by individual or groups of pathogens. We have identified Schriewer3, A. Dekonenko4, K. Shah1, L. Huang1, V. Ciccarone1, S. Burke1, M.
shRNA libraries as the method with the highest potential to discover human Bowe1, D. Farb1, M. Kadan1, E. Bonvini1, K. Stein1, S. Koenig1, S. Johnson1;
genes required for a successful infection. As a proof of principle experiment, 1
MacroGenics, Rockville, MD, 2Southern Res. Inst., Frederick, MD, 3Saint
we have prepared 18 cell lines containing candidate shRNA constructs. The Louis Univ., St Louis, MO, 4Univ. of New Mexico, Albuquerque, NM.
candidate genes were chosen based on preliminary data and literature re- An orthopoxvirus-neutralizing immunotherapeutic with post-exposure effi-
ports that certain receptors or regulators were important for successful in- cacy would be valuable in case of a bioterrorism event. One candidate is a
fection by specific pathogens. Additionally, we have also transferred an monoclonal antibody (mAb) cocktail consisting of h7D11, a humanized ver-
shRNA library that consists of several shRNA constructs per human gene and sion of 7D11 (Hooper Virol 266:329) which targets L1 protein of intracellular
covers the entire human genome, to HEK293T cells in which several differ- mature virions, and ch12F, a human/chimpanzee chimera of 12F (Chen J Virol
ent intracellular pathogens are able to grow. We have begun to test the abil- 81:8989) which targets A33 protein of extracellular enveloped virions. Re-
ity of phase II Coxiella burnetii and Rickettsia conorii to replicate in the cell search lots and a GMP lot of the mAbs were produced; the GMP product was
lines in which the candidate genes have been knocked down. In addition, we formulated as a 1:1 cocktail and has shown good stability. Individual mAbs
have constructed a Brucella abortus strain that expresses mCherry, a red flu- and the mixture were administered as single i.p. injections at doses ranging
orescent protein that facilitates the measurement of intracellular growth. In from 10 to 100 mg/kg to vaccinia-infected Balb/c mice (5e5 PFU/intratra-
order to discover additional potential host targets we screened a collection cheal) at 3 days post infection (dpi), ectromelia-infected A/NCR mice (15

BIODEFENSE PROGRAM & ABSTRACTS 69


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

PFU/intranasal) at 5 dpi, or monkeypox (MPXV)-infected CASTEiJ mice (4400 sponse and protection against lethal anthrax challenge. BMDC pre-pulsed
PFU/intranasal) at 3 dpi. The cocktail significantly improved survival and/or with heat-killed B. anthracis, rPA and CpG have upregulated expression of
reduced disease compared to vehicle-treated animals, with h7D11 as the key CD80, CD86 and MHC-II, and by seven days post transfusion have induced a
contributor in the vaccinia model and ch12F in the ectromelia model. A phar- specific T cell response in recipient mice. When rPA in alum is co-delivered
macokinetic study of the cocktail in naïve cynomolgus monkeys showed ter- with pulsed BMDC we see enhanced T cell and antibody responses compared
minal t values of ~9 and 13 days for h7D11 and ch12F, respectively. to rPA and alum alone. Mice vaccinated ona a single occasion with a sub-op-
Cynomolgus macaques challenged i.v. with 5e7 PFU of MPXV and treated i.v. timal rPA and alum dose together with pulsed BMDC have 100% survival fol-
at 1 dpi with 100 mg/kg mAb cocktail, 1000 mg/kg vaccinia immune globu- lowing lethal B. anthracis challenge, compared to 80% for rPA and alum
lin (VIG) or vehicle demonstrated 100, 67 and 0% survival, respectively. The vaccinated control mice and 40% for mice receiving pulsed BMDC only.
mAb cocktail reduced viremia, the number of pox lesions and body weight
loss to a greater extent than VIG. In summary, the data show the mAb cock-
tail to be an effective poxvirus-neutralizing immunotherapeutic that is more 199 (H)
potent than VIG when administered post-exposure. (Support: NIAID grant Radioprotective Mn-DP-Pi Complexes Preserve Immunogenic-
5U01AI070504-04 and NIAID contract N01AI30063 Task Order C24) ity of Inactivated VEEV During Gamma Irradiation: A New Ap-
proach for Vaccine Development
197 (H) M. Gayen1,2, E. K. Gaidamakova1, P. Gupta1,2, A. Sharma1, P. J. Glass3, M. J.
Daly1, R. K. Maheshwari1; 1Uniformed Services Univ. of Hlth. Sci., Bethesda,
Clinical Development of a Plant-Produced Subunit Vaccine MD, 2Birla Inst. of Tehnology and Sci., Pilani, India, 3U.S. Army Med. Res. Inst.
against Bacillus anthracis of Infectious Diseases, Frederick, MD.
V. Yusibov, J. Chichester, S. Manceva, K. Musiychuk, M. Shamloul, A. Rhee, J. Venezuelan equine encephalitis virus (VEEV) is a mosquito borne New World
Trefethen, M. Jones; Fraunhofer USA CMB, Newark, DE. alphavirus in the family Togaviridae. VEEV causes biphasic disease with ini-
The potential use of biological agents or biowarfare threatens the security of tial replication in lymphoid and myeloid tissues followed by infiltration of the
populations globally. Biological weapons can be easily concealed, trans- central nervous system initiating neuropathology and sometimes fatal en-
ported and released with devastating consequences, making rapid response cephalitis. Currently there are no FDA approved vaccines for VEEV prophy-
capabilities a task of paramount importance. A quick response to a potential laxis. A live attenuated strain of VEEV, V3526, was shown to provide excellent
bioterrorist attack requires the immediate availability of reagents for mass immunogenicity in Phase I clinical trial however caused adverse effects in
therapeutic treatment or for mass vaccination. Bacillus anthracis is one of vaccinees. Therefore, attempts to generate an inactivated VEEV vaccine are
the most-often cited biological threats. Extensive research efforts have been ongoing. A high dose of gamma irradiation ensures complete virus inactiva-
directed towards the development of recombinant subunit vaccines based tion but also leads to loss of immunogenicity due to oxidative damage of epi-
on protective antigen (PA) of anthrax. Among the emerging technologies for topes. Here, we describe a novel approach of gamma inactivation of V3526
the production of these vaccine antigens is our “launch vector” system. Plant while preserving the viral immunogenicity. Use of the attenuated V3526 over
virus-based transient expression systems are suitable for rapid engineering, the virulent TrD strain provides an additional layer of protection, the ability to
ease of scale-up and cost-effective production of target antigens. In addition, work in BSL-2 conditions and an additional protective epitope on the surface
this technology is particularly well suited for producing large quantities of of the virus. In previous work, we identified radioprotective Mn2+-peptide-Pi
vaccine antigens in a short period of time, which is particularly important complexes in highly radioresistant bacteria, Deinococcus radiodurans. In this
when faced with natural outbreaks or intended release of bio-treat agents study we show that reconstituted Mn2+-DP-Pi complexes protect V3526 epi-
such as anthrax. To this point, we have successfully engineered and ex- topes from high doses of gamma radiation, which obliterated its genome.
pressed a PA antigen in Nicotiana benthamiana plants using agroinfiltration. Viral envelope protein immunogenicity was examined using PE2 monoclonal
This plant-produced antigen elicited high toxin neutralizing antibody titers antibody. RNA infectivity was examined by transfection into sensitive cell
in mice and rabbits after two vaccine administrations with Alhydrogel. In ad- lines. Our study demonstrates that Mn2+-peptide-Pi complexes should be
dition, immunization with this vaccine candidate protected 100% of rabbits further explored for generation of highly immunogenic gamma irradiated
from a lethal aerosolized B. anthracis challenge. These results support the VEEV vaccine. This work was supported by funding from the DTRA and Air
testing of this vaccine candidate in human volunteers and the utility of our Force Office of Scientific Research. Opinions presented here are of the au-
plant expression system for the production of a recombinant anthrax vaccine. thors and should not be construed as that of USUHS, DoD, DTRA, and US-
The availability of safe, efficient and easily delivered vaccines against this AMRIID.
pathogen is of paramount importance to protect civilian populations and mil-
itary personnel.
200 (H)
198 (H) Longevity of Vaccine Protection against Pneumonic Plague in
BALB/C Mice: Part 2
Adoptive Transfer of Pulsed BMDC Enhances Specific Immune Z. N. Llewellyn1, F. Koide2, L. E. Bowen1, P. William2, S. Lin2, J. A. Boydston1, N.
Responses and Provides Protection against Lethal Anthrax Harman-Richardson3, P. M. Silvera2; 1Southern Res. Inst., Birmingham, AL,
Challenge 2
Southern Res. Inst., Frederick, MD, 3Alpha StatConsult, LLC, Damascus, MD.
I. J. Thompson1, M. G. Stokes1, B. Hallis2, A. J. Simpson1, E. D. Williamson1; Background: Yersinia pestis is the etiological agent of bubonic and pneu-
1
Defense Sci. and Technology Lab., Salisbury, United Kingdom, 2Hlth. Pro- monic plague. Subunit vaccine approaches comprising recombinant forms of
tection Agency, Salisbury, United Kingdom. the fraction 1 (F1) and V (virulence) proteins have demonstrated protection
Dendritic cells are potent activators of the immune system and have a key against pneumonic plague in mice. At last year’s meeting, we reported on
role in linking innate and adaptive immune responses. In the current study we the protection conferred by rYP vaccine at early times post-vaccination and
have used Bacillus anthracis as a model to develop an adoptive transfer strat- showed that F1 and V antibody levels correlated with protection against chal-
egy, using pulsed Bone Marrow Dendritic Cells, to provide protection against lenge with Y. pestis CO92 [Abstract number 207 (H)]. Here, we have extended
infectious agents. Recombinant Protective Antigen (rPA) formulated with our studies to further investigate the longevity of protection induced by rYP
alum provides a high titre antibody response to rPA, a detectable T-cell re- vaccine. Methods: On day 0 and 21, three sets of Balb/c mice (N=110/set)

70 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

in Groups 1-6, were vaccinated with 2.50, 0.63, 0.16, 0.04, 0.01 and 0μg vac-
cine plus Alhydrogel, respectively. Blood collected on various times post-vac-
202 (H)
cination was analyzed for F1 and V antibodies by ELISA. On day 79, 109 and Stabilization Technology for Biodefence Vaccines: Exemplifica-
139, the 3 sets of mice were exposed to 1.1x106, 1.4x106 and 2.0x106 CFU/L tion Using a New Viral Vaccine and an Adjuvanted Vaccine
Y. pestis CO92, respectively, via nose-only inhalation exposure and monitored J. Drew, C. Andrews, E. Abbott, S. Ward; Stabilitech Ltd., London, United King-
for 14 days. Results: On Day 21, mean F1 and V antibody titers in Groups 1- dom.
5 were 2.72, 2.61, 2.34, 1.74, 1.32 and 1.56, 1.55, 1.55, 1.56, 1.56 log10 Improved stability against temperature excursions for vaccines and biophar-
assay units, respectively. F1 and V antibodies continued to rise in Groups 1- maceuticals would have a significant and obvious impact, particularly with re-
5 following the 2nd vaccination on Day 21. Notably, V-specific antibodies were spect to the logistics of distribution in challenging conditions or where
significantly boosted in Groups 1-4 by Day 29 (p<0.01). As expected, F1 and temperature control variances are prone to occur. Stabilitech Ltd is a UK-
V antibodies were not detected in Group 6. Following challenge with Y. pestis based company which has overcome thermostability issues for a wide range
CO92 on day 79 (set A), day 109 (set B) and day 139 (set C), survival rates in of viruses, vaccines, biopharmaceuticals and Alhydrogel adjuvant using our
Groups 1-6 were 95, 90, 85, 35, 0, 0, 90, 60, 85, 55, 5, 0, and 90, 90, 75, 45, proprietary stabilization technology. We have developed proprietary excipi-
15, and 0%, respectively. Moreover, survival probability correlated with F1 ent systems which, when mixed with suspensions of target vaccine or pro-
and V antibody titers (p<0.001). Conclusions: These results corroborate our tein dramatically improve stability when stored at elevated temperatures (in
previous findings demonstrating that F1 and V antibodies induced by an F1/V- dry or liquid presentations) or when taken through multiple freeze thaw cy-
based vaccine correlated with protection against pneumonic plague. This cles. The protein-free technology can be readily incorporated into cGMP man-
work was funded by NIAID contract N01-AI-30063 ufacturing processes using standard equipment. In order to illustrate the
application of this technology, results will be presented showing enhanced
stability of a novel recombinant adenovirus-based vaccine, at a range of tem-
201 (H) peratures and timepoints, both in a iquid and lyophilized form. Data for an ad-
Dose Response of IMVAMUNE, a Third Generation Smallpox juvanted subunit protein vaccine of biodefense significance will also be
Vaccine Candidate, in a Lethal Monkeypox Virus Nonhuman Pri- shown, showing long-term retention of both Alhydrogel and protein-antigen
mate Inhalation Model structure. Stabilitech’s technology has overcome several major hurdles af-
T. Brasel, K. Agans, K. Dearen, T. Bailey, A. Russo, S. Storch, R. Sherwood; fecting vaccine and biopharmaceutical storage and distribution. This propri-
Lovelace Respiratory Res. Inst., Albuquerque, NM. etary technology has the potential to facilitate significantly the deployment
First and second generation smallpox (SPX) vaccines composed of replicating of vaccines in harsh conditions in the field.
vaccinia are associated with adverse health risks. Because of this, testing of
new, safer vaccines is desirable. IMVAMUNE®, a formulation of live Modified
Vaccinia Ankara, is a promising candidate due to its inability to replicate in
203 (H)
mammalian cells. We have previously shown that a prime-boost regimen of A Marmoset Model for Lethal Orthopox Virus Infection Enables
IMVAMUNE® is fully protective in a lethal aerosol monkeypox virus (MPXV) Study of Vaccines
nonhuman primate (NHP) challenge model. The objective of this study was to C. Yue1, M. Kramski1,2, C. Stahl-Hennig3, K. Mätz-Rensing3, F-J. Kaup3, G.
determine the minimum effective dose of the vaccine using the same model. Pauli1, H. Ellerbrok1; 1Robert Koch Inst., Berlin, Germany, 2Univ. of Mel-
NHP were vaccinated with IMVAMUNE® at 1 x 108, 1 x 107, or 1 x 106 TCID50 bourne, Parkville, Australia, 3German Primate Ctr., Goettingen, Germany.
or vehicle on Study Days -67 and -39. MPXV neutralizing antibody (Ab) pro- Variola virus is under continuous discussion as a biological threat agent. This
duction was assessed at key time points post vaccination. Animals were chal- discussion is paralleled by increasing numbers of zoonotic infections in hu-
lenged with aerosolized monkeypox virus (1 x 105 pfu) on Study Day 0. mans with other orthopox viruses like monkeypox or cowpox viruses. While
Clinical observations were performed daily for up to 28 days post-challenge. the development of safer vaccines and antiviral therapies is desirable the
Blood and pharyngeal swab analysis, lesion counts, and clinical pathology question remains how to test their efficacy. Recently, we have developed a
were conducted at routine intervals. Upon necropsy, tissues from each animal lethal non-human primate infection model using calpox virus and marmosets
were collected and analyzed for MPXV burden and histopathological changes. to study the pathogenesis of orthopox virus infections (Kramski et al. 2010,
A dose response in neutralizing Ab titers was measured in non-control ani- PLoS one 5, e10412). We have applied this model to test vaccines and vacci-
mals following vaccination. Alterations, predominantly associated with res- nation strategies using Vaccinia Virus Lister-Elstree (LE) and Modified Vac-
piratory distress, were noted in all groups post-exposure, though were most cinia virus Ankara (MVA) as vaccines. Marmosets were immunized with LE or
severe in controls. Among the 24 vaccinated animals, four succumbed to MVA and challenged with a lethal dose of calpox virus either four or ten weeks
MPXV-associated complications. Alterations in these monkeys were compa- after immunization (p.im.). Animals were routinely checked for clinical symp-
rable to those observed in controls. Death was attributed to severe fi- toms. Viral load in blood and in saliva were analyzed by real-time PCR p.im.
bronecrotic pneumonia. Surviving animals demonstrated minimal alterations, and post challenge. Virus-specific antibody titers were determined by im-
though severity was inversely proportional to the IMVAMUNE® dose. Cu- munofluorescence test and by plaque-reduction neutralization test. All four
mulatively, results demonstrated that the protective efficacy of IMVAMUNE® marmosets immunized with LE and challenged ten weeks p.im. survived with-
was directly proportional to the administered dose. Though a definitive min- out any clinical symptoms although calpox virus was transiently detectable
imum effective dose was not determined, results demonstrated a waning ef- in blood. Only one out of four animals challenged four weeks p.im. survived
ficacy as the vaccine dose decreased. the lethal calpox virus challenge. In contrast, none of the animals (n=4) vac-
cinated with MVA and challenged 10 weeks p.im. survived and only 2 out of
7 animals challenged 4 weeks p.im. survived. In some of the 5 animals suc-
cumbing to disease the onset of symptoms was only slightly delayed. In con-
clusion, the non-human primate model is suitable for the study of orthopox
virus pathogenesis as well as for different vaccination strategies but exten-
sion of the observed results to humans needs to be analyzed in more detail.

BIODEFENSE PROGRAM & ABSTRACTS 71


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

elicited antibody titer of 1000 GMT compared to 43000 - 66000 GMT from
204 (H) constructs co-expressing signal sequences. Similarly, MVA/V307 conferred
New Adjuvants for Anthrax Vaccine 25% protection against challenge with 50MLD of Y. pestis (CO92) compared
Y. Gauthier1, A. Borel2, Y. Trescos1, J. Mathieu1, B. Verrier2, J-N. Tournier1; to 100% protection conferred by constructs co-expressing signal sequences.
1
IRBA, La Tronche, France, 2CNRS-IBCP, Lyon, France. Conclusions: The adoption of MVA as a second-generation smallpox vaccine
We developed a vaccine based on PA and spore (FIS) with two adjuvants. Alu- makes our recombinant MVA candidate vaccines attractive alternatives to
minum hydroxide (Al) has side effects. The mucosal adjuvant cholera toxin protect against smallpox and plague.
(CT) is not usable in humans. These limitations led to the search of new ad-
juvants. Particular synthetic oligodeoxynucleotides (CpG ODN) are potent ad-
juvants. The safe and biodegradable PLA-based (Poly (D,L)-Lactic Acid) 206 (H)
nanoparticles (NPs) have adjuvant properties. The synthetic RNA polyri- Second-Generation Inactivated Vaccines Protect Mice from
boinosinic polyribocytidylic acid (poly(I:C)) also has adjuvant effects. Our aim Lethal Venezuelan Equine Encephalitis Virus Infection
was to assess the efficiency of these potent adjuvants in a mouse model of K. Spurgers1, S. Honnold1, R. Bakken1, J. Cohen1, L. Rowan1, C. Lind1, P.
inhalational anthrax. Mice were immunized with rPA. Animals were IN infected Gupta2, A. Sharma2, R. Maheshwari2, P. Glass1; 1USAMRIID, Frederick, MD,
with spores of either the 7702 Sterne strain or the 9602 strain of B. anthracis. 2
USUHS, Bethesda, MD.
PA-PLA was as efficient as both PA-Al and PA+CT for SC and IN immunizations Venezuelan (VEEV), eastern (EEEV), and western equine encephalitis (WEEV)
respectively, and elicited 100% protection against an IN lethal challenge of viruses, members of the genus Alphavirus, are causative agents of debilita-
DBA/2 mice with 10.LD50 of the Sterne strain. PA+Poly(I:C) elicited full pro- tive and sometimes fatal encephalitis in North, Central, and South America.
tection as well, through either the SC or the IN route of immunization. Inter- Natural human cases are rare, and occur from the bite of an infected mos-
estingly, PA alone was fully protective when administered SC, and elicited no quito. However, these viruses pose a threat to military personnel, and to pub-
protection through the IN route. PLA+Poly(I:C), without PA, elicited partial lic health, because of their potential use as bioweapons. There are currently
protection when administered IN. In SWISS mice immunized with PA+FIS and no licensed vaccines to prevent alphavirus infections. To address this need,
challenged IN with 10.LD50 of the fully virulent 9602 strain, SC immunization our laboratory group derived and tested an attenuated, PE2 cleavage site
with CpG did not elicit better protection than Al. Total anti-PA and anti-spore mutant of VEEV, termed V3526. V3526 was a safe and effective vaccine plat-
IgG titers, both in the sera and BALs from CpG immunized mice, were not form in rodents and primates, and advanced to phase I clinical trials. How-
modified compared with titers obtained with Al. Nevertheless, when PA+FIS ever, human recipients of V3526 exhibited an unacceptably high frequency of
were delivered both SC and IN, anti-PA titers were decreased in an average adverse events requiring cessation of the study. To increase the safety of this
range of 10 times in mice immunized with CpG compared with Al. TNA titers vaccine candidate, several inactivated V3526 formulations were tested for
were decreased 4 times as well after CpG immunization. PA+Poly(I:C) was as efficacy against VEEV infection. Inactivation methods included formalin,
efficient as PA-Al for inducing 100% protection against an IN lethal infection gamma irradiation, and 1,5 iodonaphthyl azide (INA). In initial studies, small
with the Sterne strain. Immunization of DBA/2 mice with biocompatible 200 doses of vaccine were administered, and afforded only partial protection
nm PA-PLA NPs was as fully protective as PA-Al. CpG-ODN 1826 used as sin- against aerosol VEEV challenge. The current study tests higher doses of three
gle adjuvant for vaccination with (PA+FIS) did not improve the immune re- inactivated V3526 formulations without adjuvant, administered by three dif-
sponse and protective activity against lethal inhalational anthrax compared ferent routes (intranasal, intramuscular, and subcutaneous). Cohorts of mice
with Al. received either one dose, or two doses of vaccine (28 d apart). Mice were
then challenged with aerosolized VEEV (Trinidad). Complete protection was
205 (H) observed when two doses of gamma irradiated-V3526 or INA-V3526 were
administered i.m. For each cohort of mice, total serum anti-VEEV antibody,
Effects of Signal Sequences on the Immunogenicity and Effi- and neutralizing antibody levels are reported. Best performing vaccine for-
cacy of an MVA Vectored Plague Vaccine mulations, route, and dose will be down-selected for further testing with ad-
J. Brewoo1, T. Powell2, G. Young1, C. Partidos1, D. Stinchcomb2, J. Osorio3; 1In- juvants.
viragen, Inc., Madison, WI, 2Inviragen, Inc., Fort Collins, CO, 3Univ. of Wis-
consin, Madison, WI.
Background: Signal sequences have been employed in vaccine development 207 (H)
to facilitate the expression of target antigens and significantly enhance their Evaluation of Inactivated and Live-Attenuated Bivalent Vac-
immunogenicity. We report on the effects of different post-transcriptional reg- cines That Confer Protection against Rabies and Ebola Viruses
ulatory signals on the immunogenicity and protective efficacy of a recombi-
in Mice
nant modified vaccine Ankara (MVA) vector expressing a truncated version
J. Blaney1, A. Papaneri1, C. Wirblich2, J. Cann3, M. Holbrook3,4, A. Freiberg4, P.
of the low-calcium virulence antigen (V307) from Yersinia pestis. Methods:
Jahrling1,3, M. Schnell2; 1 NIAID, NIH, Fort Detrick, MD, 2 Jefferson Med. Coll.,
MVA recombinants were constructed expressing antigen from Y. pestis (V307)
Thomas Jefferson Univ., Philadelphia, PA, 3Integrated Res. Facility, NIAID,
under the translational control of the encephalomyocarditis virus (EMCV) in-
NIH, Fort Detrick, MD, 4Univ. of Texas Med. Branch, Galveston, TX.
ternal ribosomal entry site (IRES) and/or fused to the tissue plasminogen ac-
Our objective is to generate an Ebola virus vaccine with maximum likelihood
tivator (tPA) or a putative vaccinia virus secretory signal (C13L). The candidate
of licensure based on considerations of safety, manufacture, and cost. To this
vaccines were tested for antigen expression in vitro, immunogenicity and pro-
end, we developed (A) replication-competent, (B) replication-deficient, and
tection in a mouse plague challenge model. Results: Western blot analysis
(c) chemically inactivated rabies virus (RABV) vaccines expressing Zaire
showed increased antigen expression from vaccine constructs co-expressing
ebolavirus glycoprotein (GP) using a reverse genetics system based on the
V307 and tPA (MVA/tPA/V307) or C13L (MVA/C13L/V307) compared to con-
SAD B19 RABV wildlife vaccine. Each vaccine candidate is apathogenic upon
structs expressing only the V307 (MVA/V307). Antigen expression from the
peripheral administration in mice, induces anti-RABV and anti-Ebola humoral
construct co-expressing V307, IRES and tPA (MVA/IRES/tPA/V307) was signif-
immunity, and confers protection from lethal mouse-adapted Ebola and RABV
icantly lower than the other constructs. Interestingly, immunogenicity of the
challenge. We have further characterized the safety and immunogenicity of
candidate vaccines was superior with constructs co-expressing signal se-
these live and inactivated vaccine candidates. Replication-deficient vaccine
quences. Following prime and boost (28 days apart) vaccination, MVA/V307
candidates expressing GP with a deletion in the RABV glycoprotein were

72 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

found to be avirulent after intracerebral inoculation of adult and suckling lenged on Day 149 by aerosol exposure to Y. pestis CO92 at a target dosage
mice and peripheral administration in SCID mice. Analysis of replication-de- of 5.5 x 103 cfu. Animals were observed for signs of disease and changes in
ficient viruses in the brain by quantitative PCR, determination of virus titer, clinical pathology, body temperature and activity. Samples for evaluation of
and immunohistochemistry indicated greatly restricted virus replication. Fur- humoral immune responses were taken throughout the study and on the day
ther examination of the immune response to these RABV vectored vaccines of challenge and were tested in a Bridge ELISA for presence of anti-rF1, anti-
by ELISPOT assay indicated that a memory T cell response is induced by both rV and anti-rF1V antibodies. The data generated from these studies were sta-
live and killed vaccines. Finally, we demonstrated that a multivalent antibody tistically analyzed to provide a more precise estimate of the relationship
response can be induced by co-administration of the RABV/Ebola vaccine between vaccine dosage, Bridge ELISA titers and survival. Histopathology
and another RABV vectored vaccine expressing the botulinum neurotoxin A and residual Y. pestis were evaluated in the lungs, liver, kidney and spleen in
HC50 fragment. Based on the strong safety and immunogenicity profiles of all animals that succumbed to disease. Results: A full survival curve was ob-
these bivalent vaccines directed against RABV and Ebola in mice, we are cur- tained over the course of both studies. The protective titer and protective
rently planning studies in nonhuman primates. vaccine dosage at 50% survival were estimated to be 878 U/mL of anti-rF1V
and 2.05 µg rF1V vaccine, respectively. Conclusions: The results of these
studies indicate a significant relationship between vaccine dosage and sur-
208 (H) vival (p<0.001) and between Bridge ELISA titer and survival (p<0.001) for all
Improved Efficacy of a Multiagent Equine Encephalitis Virus analytes (rF1, rV and rF1V).
Vaccine Candidate
P. J. Glass1, R. R. Bakken1, J. Ennis2, K. B. Spurgers1, K. I. Kamrud3, K. Alter-
son3, M. Custer3, J. Turner2; 1USAMRIID, Fort Detrick, MD, 2Defyrus Inc.,
210 (I)
Toronto, Canada, 3Alphavax, Research Triangle Park, NC. Peptide Mapping of Anthrax Toxin Protective Antigen Using
Background: Virus-like replicon particles (VRPs) were produced that express Anti-AVA Human Sera
the Venezuelan (VEEV), eastern (EEEV) or western (WEEV) equine en- V. A. Semenova, P. Svoboda, J. Pohl, S. Soroka, C. Quinn; CDC, Atlanta, GA.
cephalitis virus E1 and E2 glycoproteins. Previous studies showed that a mul- Anthrax toxin protective antigen (PA) is a major protective immunogen in the
tiagent equine encephalitis virus vaccine consisting of an equal mixture the anthrax vaccine adsorbed (AVA, BioThrax). Identification of antigenic se-
VEEV, EEEV and WEEV VRPs (1x107 IU each) administered subcutaneously quences of PA is important for future vaccine designs and immunotherapeu-
protected mice from lethal aerosol challenge with VEEV, EEEV and WEEV. tics. We characterized PA linear epitopes using sera with anti-PA IgG
Methods: In this study, we examined the efficacy of a lower dose of the mul- concentrations ≥ 300 μg/ml from human volunteers in the CDC Anthrax Vac-
tiagent VRP (maVRP) vaccine (1x105 IU each) administered by intramuscular cine Research Program (AVRP). Fmoc synthesis was used to prepare 37 N-
(IM) or intranasal (IN) routes, with or without DEF201. DEF201 consists of a terminally biotinylated peptides of 30 amino acid (AA) residues, overlapping
proprietary adenovirus type 5 (Ad5) vector expressing interferon (IFN)-α, that by 10 AA. Except for the C-terminal peptide made as the free acid, sequences
is administered IN. BALB/c mice were given one or two doses of the maVRP were Fmoc synthesized as C-terminal amides, HPLC-purified and prepared as
vaccine or PBS, with or without DEF201 (primary vaccination only) and chal- trifluoracetic acid salts. Peptides were captured on streptavidin coated (2
lenged 28 days post-final vaccination with VEEV, EEEV, or WEEV. Mice were μg/ml) microtiter plates. Seroreactivity was evaluated colorimetrically using
monitored daily for mortality and clinical signs of disease for 21 days sera of 60 volunteers who had received 8 subcutaneous (SQ) or at least 3 in-
postchallenge. Results: Mice given one or two doses of the maVRP vaccine tramuscular (IM) injections of AVA with the licensed regimen. A peptide was
alone by the IN or a single dose by the IM route were not protected against considered antigenic if its OD value was greater than the mean OD+3SD of
lethal aerosol challenge with VEEV, EEEV or WEEV. DEF201 administered in control group sera (n=24). Sera from all vaccinees reacted with peptide se-
conjunction with one or two IN doses of the maVRP vaccine provided en- quences corresponding to all 4 domains of PA. There were four predominant
hanced protection (80-100%) against lethal EEEV and WEEV aerosol chal- seroreactive PA peptide regions. These peptides corresponded to amino acid
lenge. Two IM doses of the maVRP vaccine alone resulted in 70-90% (AA) sequences in the chymotrypsin sensitive loop 2ß2-2ß3 (AA 301-330),
protection. Administration of DEF201 in conjunction with two IM doses of the 2ß3- 2α1(AA321-350), 2ß10-12(AA421-450) in domain 2, and 4ß5-8(AA641-
maVRP vaccine completely protected mice against lethal VEEV, EEEV, or WEEV 670) in domain 4 of mature protein sequence Bacillus anthracis V770-NP1-R
challenge. Conclusions: IM vaccination with a lower dose of the maVRP vac- (BioThrax Vaccine Strain). We demonstrated that serum antibodies from
cine effectively protected mice from lethal challenge with VEEV, EEEV, or human AVA recipients exhibit reactivity to a range of different sequences in
WEEV when administered as a single dose regimen with DEF201 or a two- PA. The major antigenic regions were located in domains 2 and 4. These data
dose regimen with or without DEF201. Future studies will examine onset and in humans concur with the previous findings in non-human primates that pro-
duration of the maVRP vaccine efficacy when administered in conjunction tective epitopes can be attributed to specific PA domains (Flick-Smith et al.,
with DEF201. 2002; Williamson et al., 2005).

209 (H) 211 (I)


Relationship between Vaccine Dosage, Immunogenicity and Serum Opsonization Promotes the Efficient Uptake and Clear-
Survival in RF1V-Vaccinated Cynomolgus Macaques ance of Burkholderia pseudomallei by Human Neutrophils
P. Fellows1, J. Price1, R. Krile2, H. Lockman2, L. Wolfraim1; 1Dynport Vaccine M. E. Woodman, R. G. Worth, R. M. Wooten; Univ. of Toledo Coll. of Med.,
Company Llc, Frederick, MD, 2Battelle, Columbus, OH. Toledo, OH.
Objective: The objective of these studies was to determine minimum protec- Burkholderia pseudomallei is the causative agent of melioidosis and is a
tive titers in cynomolgus macaques (CM) vaccinated with a recombinant major mediator of sepsis in its endemic areas. Because of the low LD50 via
plague vaccine (rF1V) and subsequently challenged by aerosol exposure to aerosols and resistance to multiple antibiotics, it is considered a Category B
Yersinia pestis CO92. Methods: Two dosage ranging studies were performed bioterrorism agent by the CDC. B. pseudomallei is an encapsulated bacterium
to establish a full survival curve (0 to 100% survival) associated with rF1V that can infect, multiply, and persist within a variety of host cell types. Burk-
vaccination. CMs were vaccinated with rF1V on Days 0, 56 and 121 by intra- holderia thailandensis is closely-related to B. pseudomallei, but lacks a poly-
muscular injection with dosages ranging from 0.016 to 20 µg and were chal- saccharide capsule and is relatively avirulent in infection models. In vivo

BIODEFENSE PROGRAM & ABSTRACTS 73


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

studies suggest that macrophages and neutrophils are important for con-
trolling B. pseudomallei infections, however few details are known regard-
213 (I)
ing how neutrophils respond to these bacteria. Our goal is to describe the A Quantitative PCR-Based Method for the Evaluation of Anti-
capacity of human neutrophils to control highly virulent B. pseudomallei com- Vaccinia Neutralization Titers
pared to the relatively avirulent B. thailandensis using in vitro analyses. B. A. D. Garcia, C. A. Meseda, J. Weir; FDA/CBER, Bethesda, MD.
thailandensis was more readily phagocytosed than B. pseudomallei, but both The plaque reduction neutralization test (PRNT) is a standard method used
displayed similar rates of persistence within neutrophils, indicating they pos- for measuring poxvirus neutralizing antibodies in vaccinees. However, the
sess similar inherent abilities to escape neutrophil clearance. Serum op- PRNT is labor- and time consuming and can require relatively large amounts
sonization studies showed that both were resistant to direct killing by of immune serum, making this neutralization test impractical for use in large
complement, although B. thailandensis acquired significantly more C3 on its scale studies and in the mouse challenge model. Therefore, the goal of this
surface than B. pseudomallei. Both Burkholderia spp. showed significantly work was to develop a rapid and efficient method to measure vaccinia virus
enhanced uptake and killing by neutrophils after serum opsonization. Killing neutralization based on viral DNA sampling performed in a multi-well format.
of serum-opsonized B. pseudomallei and B. thailandensis was prevented with Here, we have applied a SYBR green endpoint quantitative PCR methodology
inhibition of the NADPH-oxidase indicating the importance of reactive oxy- to quantitate the changes or reduction in vaccinia genome copy numbers, as
gen species in this process. Serum-opsonized Burkholderia induced a sig- the assay readout, in response to neutralization of virus infectivity by vari-
nificant respiratory burst by neutrophils compared to unopsonized bacteria. ous immune sera. To evaluate the feasibility of a vaccinia qPCR assay method,
In summary, while complement opsonization is important for efficient uptake we have designed and tested PCR primer sets specific to 3 vaccinia virus
and killing of both strains, the absolute levels of surface C3 does not corre- genes: J2R, G9R, and E9L. Our preliminary studies have determined that
late with increased killing of B. pseudomallei and B. thailandensis. Based on primers to the J2R and G9R genes were suitable for further development of a
these findings, neutrophils appear able to play a significant role in clearing vaccinia qPCR method. Next, the qPCR assay was optimized for both primer
B. pseudomallei infection. sets in which the PCR reactions contained ~102 genomic equivalents of the
respective control reference plasmid, pCR-vvJ or pCR-vvG. The assay quanti-
tative range was determined to be ~102 to >108 genomic equivalents. In ad-
212 (I) ditional feasibility studies, we have demonstrated that the qPCR assay can
Priming of Alveolar Macrophages Augments Inflammatory Re- reproducibly measure virus genomic copies in total cell extracts prepared
sponse on Anthrax Lethal Toxin Stimulation from Vero cell monolayers (in 48-well plates) that were infected with ~100
J. Thomas, L. Xiao, J. W. Christman, J. L. Cook, I. Levitan; Univ. of Illinois Med. pfu of vaccinia virus (WR strain or NYCBOH strain). Based on neutralization
Ctr. at Chicago, Chicago, IL. experiments done with rabbit anti-vaccinia virus polyclonal antibodies, our re-
Background: The cause of rapid death in anthrax infection both in animals sults indicate that the qPCR method represents a viable approach for the de-
and humans are unknown and concern over the use for biological warfare tection and measurement of vaccinia-specific neutralizing antibodies in
has renewed interest in elucidating the mechanisms of anthrax induced in- immune sera.
flammation. Interleukin-1β (IL-1β) secretion is an important inflammatory re-
sponse against anthrax lethal toxin (LeTx) a virulence factor of Bacillus
anthracis and Lipopolysaccharides (LPS), an endotoxin of all gram-negative
214 (I)
bacteria. Recently, we reported that in mouse macrophages LeTx activates, in- The Chemokine Receptor CCR7 is Expressed in Cells of the
wardly-rectifying K+ (Kir) and voltage-gated K+ channels (Kv), the 2 types of K+ Human Lung in Association with Bacillus anthracis Spore In-
channels in mouse macrophages. Blocking these channels inhibits IL-1β se- fection
cretion. Susceptibility to LeTx is augmented by a two-fold increase in IL-1β se- M. Langer1, J. Booth1, E. Duggan1, V. Patel1, T. Veres2, F. Prenzler2, K. Sewald2,
cretion primed overnight with LPS compared to unprimed cells challenged K. Coggeshall3, A. Braun2, J. Metcalf1; 1Univ. of Oklahoma, Oklahoma City, OK,
with LeTx alone. We hypothesize that reactive oxygen species (ROS) gener- 2
Fraunhofer Inst. for Toxicology and Experimental Med., Hannover, Germany,
ated by LPS activate K+ channels and may prime cells for increased activation 3
Oklahoma Med. Res. Fndn., Oklahoma City, OK.
upon LeTx challenge. Methods: Whole-cell patch clamp was used to deter- Background: The lung is the entry site for Bacillus anthracis in inhalation an-
mine if toxin and ROS activates K+ channels in macrophages. ROS activation thrax, the most deadly form of the disease. B. anthracis does not cause dis-
of K+ channels was measured by a brief exposure to H2O2 or a ROS scavenger ease in the lung. Instead, spores are internalized by resident lung cells and
MnTmPyP. Release of IL-1β was determined by ELISA. Results: LPS primed disseminate to cause systemic disease as the vegetative form of the organ-
LeTx stimulation yielded a higher IL-1β secretion and Kir current compared to ism. Three cell types resident in the lung have been previously shown to in-
unprimed LeTx stimulation suggesting priming affect of LPS on K+ channels ternalize spores: alveolar macrophages (AM), dendritic cells (DC) and alveolar
mediated inflammatory response. As a first report we show that LPS alone is epithelial cells (AEC). One of these cells may contribute to dissemination by
sufficient to illicit K+ channel activation and IL-1β secretion. LPS stimulate K+ migrating to the lymphatic system after uptake of spores. In particular, we
channels in a dose and time depended manner. Lastly, H2O2 alone can acti- hypothesized that DC may responsible for carrying spores to the lymph
vate K+ channels and inhibition of LPS-generated ROS by MnTmPyP inhibits nodes. If this occurs, the chemokine receptor CCR7, a receptor involved in
K+ channel activation , suggesting the role of ROS in K+ channel activation. DC chemotaxis, may be upregulated after spore uptake. Methods: We used
Conclusion: In macrophages LeTx or LPS alone is sufficient to induce both IL- fluorescent immunohistochemistry and confocal microscopy, in addition to
1β secretion and activation of K+ channels. LPS-generated ROS may be a prim- immunofluorescent staining and flow cytometry to investigate spore uptake
ing mechanism of K+ channel activation and IL-1β secretion. This model may and CCR7 expression. Our models were human lung organ culture and iso-
be useful to design new therapeutic strategies to block endotoxic-shock re- lated human AM obtained by bronchial-alveolar lavage. Results: We found
sponse to anthrax infection. that the majority of spores were ingested by AM and DC, while AEC ingested
significantly fewer spores. Spore ingestion was associated with increased ex-
pression of CCR7 in cells that take up numerous spores, either AM or DC.
However, the relevant cell type is not likely AM because isolated AM do not
express CCR7 after taking up spores. Conclusions: Our results suggest that
DC may increase expression of CCR7 after internalization of spores and may

74 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S DAY A B S T R AC TS

migrate to the lymph nodes carrying spores. Our results further suggest that
CCR7 is a potential therapeutic target in preventing dissemination of B. an-
217 (K)
thracis from the lung, and thereby preventing systemic disease. WMD Training Courses at Dugway Proving Ground, Dugway UT
P. B. Brown, J. Montague, J. Sorenson; U.S. Army, Dugway Proving Ground,
Dugway, UT.
215 (J) How do you collect a powder sample aseptically and limit the spread of the
Forensic Entomology and Biodefense contamination? What does Bacillus anthracis look like on an agar plate? Do
A. Lindström; Natl. Vet. Inst., Uppsala, Sweden. all Bacillus spores look the same under the microscope? What are the signa-
Forensic entomology is the application of the science of entomology to any tures for a bacterial production lab? Where is the best sample point when
legal case. Traditionally it is used for postmortem interval estimates in homi- sampling a clandestine biological laboratory? All of these questions and more
cide investigations where the age of insects on the corpse is used as an ap- will be answered during courses offered at US Army, Dugway Proving Ground.
proximation of the time that have elapsed since the person died. For Courses are taught by microbiologists that emphasize hands on training. Stu-
biodefense purposes the science of forensic entomology is more involved in dents will gain experience in aseptically collecting powder and liquid sam-
predicting scenarios where insects are used to inflict economic or bodily harm ples. They will practice collecting samples from powder disseminations; they
to humans, livestock or crops. If such an attack was discovered the forensic will learn to recon and sample a rogue laboratory, and work with attenuated
entomologist would also have an important task in collecting and interpeting strains of Bacillus anthracis, Yersinia pestis and Francisella tularensis. Stu-
the evidence so that the offenders can be brought to justice. Unfortunately dents will gain knowledge and experience using Biological Detection equip-
from a biodefense viewpoint insects are a problem since they present a rel- ment such as Hand Held Assays, RAZOR, RAPID, HazMat ID, and Microscopes.
atively cheap low tech, high impact, low risk way for terrorists to spread Customers include First Responders, Civil Support Teams and other Military
pathogens or at least the fear of pathogens. Instead of obtaining pathogens units.
by stealing them from high security facilities, which is a common scenario,
they can be found naturally infecting wild animals and livestock in several
nations of concern in for example North Africa and the Middle East as well as
218 (K)
USA. Diseases such as bubonic plague and Rift Valley Fever could easily be WITHDRAWN
obtained together with appropriate insect vectors. Propagation of the vector
and the pathogen would then be straightforward utilizing information read-
ily available on the internet. For the medical entomology community as well
219 (K)
as for medical and veterinary practitioners it is important to have forensic Biological Security: Identifying Vulnerabilities, Preventing
awareness when it comes to vector borne diseases in order to discover in- Damage
tentional release of disease vectors. C. Uhlenhaut; Robert Koch Inst., Berlin, Germany.
Our goal is to identify vulnerabilities within the biosecurity context in Ger-
many by combining different approaches in an innovative way. One main pil-
216 (J) lar of this program is developing generic scenarios. Being build from blocks,
Rapid Detection of Endospores by Capillary Electrophoresis on these should ideally work for several settings or several pathogens, e.g., de-
a Hybrid Channel-to-Digital Microfluidic Platform for Steriliza- pending on the infectious route. These scenarios will then be used to work on
risk assessments, involving scientists but also specialists from other fields.
tion Validation
Since the scenarios are scalable, several theoretic outcomes can be modeled
L. Li, W. Wu, W. Chan, H. Cheong, P. Yung; The Chinese Univ. of Hong Kong,
and evaluated for each pathogen or setting. The scenarios will be tested by
Shatin, Hong Kong.
entering data and information from previous outbreaks and incidents. These
We present the design, implementation and demonstration of a microfluidic
can then be modified by changing the setting, climate conditions, mode of
device for the rapid detection of endospores by time-gated fluoresecnce and
transmission etc. The information obtained will be used to identify knowl-
bioluminescence. Endospore formation is a very successful survival and dis-
edge gaps, prioritize resources (e.g. financial, research) and to provide im-
persal strategy. They are found in nearly every conceivable natural habitat
proved information. This information will be custom-tailored, e.g., for policy
on Earth and persist for long times due to their extreme resistance. There-
makers, first responders or the general public. To achieve this, we identify
fore, endospores have been fascinating subjects of investigation for a num-
the risk communication needs and networks for which we then produce our
ber of important applications, from medical sterilization validation, food
own videos, print material and other online resources. One of these will be a
safety, biodefense to astrobiology. Detection of low levels of dormant en-
new databank which will contain all relevant information about biological
dospores under various extreme environments presents great challenges to
materials, not only biothreat agents or highly pathogenic organisms. We
researchers. Here we propose a rapid and automated device to determine
chose this broad approach in order to provide one format for all potential bi-
endospore viability from different perspectives, namely germination propen-
ological hazards, not only intentional releases but also accidental releases or
sity, metabolic activity, protein degradation and membrane integrity. En-
naturally occurring infections. We will develop different output formats for
dospores are detected based on a series of unique physiological changes
different users, e.g., first responders will have all the relevant information at
during germination, exemplified by dipicolinic acid release, ATP synthesis,
hand while someone trying to assess the potential economic impact of a cer-
protein degradation and membrane stainability. Total endospore population
tain biothreat agent will get a different set of information. However, this will
is determined by monitoring dipicolinic acid release upon electroporation.
be done to facilitate working with the database and if necessary more or all
Sample handling, processing and analyses are incorporated into a hybrid mi-
information will be available. We are working on a biothreat safety net, com-
crofluidic chip highlighting capillary electrophoresis and channel-to-digital
bining known facts with generic scenarios and expertise from a broad range
conversion for smaller reagent volumes, shorter reaction times, and a better
of experts. In the end we will have a set of tools, such as generic scenarios
separation efficiency. This lab-on-a-chip design enables portability and on-
and an intelligent database that will allow assessing risks and identifying vul-
the-spot analysis to probe the in situ viability state of endospores in their na-
nerabilities.
tive environments, providing us valuable information in medical sterilization
validation and food safety.

BIODEFENSE PROGRAM & ABSTRACTS 75


T U E S D A Y P O S T E R S E S S I O N | 1:00–3:00 p.m. | Exhibit Hall

220 (K)
Transitional Operations: Guiding Owners through Start-Up Op-
erations & Regulatory Compliance
M. L. Weiss, D. C. Sharpe; The WorkingBuildings Companies, Atlanta, GA.
Objective: To introduce a new process in construction project delivery that
incorporates SOPs, Regulatory Compliance, Safety and Maintenance as the
drivers for successful design, commissioning and operations. The focus is on
the transition period between acceptance of a new building and full opera-
tions. Methods: This topic will be explored through a case study that includes
various testing protocols in BSL-2 and BSL-3 spaces. The presentation will
outline the importance of the Owner’s responsibility for information gather-
ing, organization and documentation on the successful transition between
acceptance and operations. Discussion: Laboratories are becoming more
complex to design, build and operate, and in many cases have been deliv-
ered but never become fully operational. In response to this, commissioning
has become an accepted practice to ensuring that the complex systems are
aligned with the Design Intent. Conclusion: Transitional Operations is a
process utilizing a team of knowledgeable facility operators and regulatory
compliance experts that is on-site full-time during the transitional period to
ensure the Design Intent is met and to guide Owners through the task of op-
erating and maintaining their new laboratory. It is a process that builds upon
traditional commissioning activities and extends into through the first year of
operations allowing Users and Facilities Management time to focus on the
ongoing operations of their existing facility and coordinating the move and
assistance with the laboratory regulatory compliance and certification
process. This time also allows for facility and regulatory compliance training
and shortening the users learning curve on the operations and management
of the laboratory’s new building systems.

76 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N D AY O R A L A B S T R A C T S

dicated that C. burnetii spreads into the environment in the direction of the
Highlighted Oral Abstract Presentations prevailing wind, but at 200-400 meters from the breeding areas very limited
009. Environmental Detection amounts of C. burnetii could be detected. These results demonstrate that al-
though large amounts of C. burnetii were released into the environment at
Monday, February 27, 2012 | 3:00 PM - 4:00 PM these farms during the birthing of infected goats, conditions at these farms
Diplomat Ballroom resulted in limited spreading to locations outside of the birthing area.

108 110
Case-Clusters of Melioidosis Associated with Climate Condi- Rapid and Sensitive On-Site Identification of Biothreat Agents
tions and Geographical Topography in Southern Taiwan Using Electrochemical Biochips and a Portable Detection Plat-
Y-L. Chen1, Y-S. Chen2, H-S. Lin3, P-J. Liu1; 1Natl. Kaoshiung Normal Univ.,
form
Kaohsiung, Taiwan, 2Kaohsiung Veterans Gen. Hosp., Kaohsiung, Taiwan, 3E-
T. Elssner, C. Pöhlmann; Bruker Daltonik GmbH, Leipzig, Germany.
DA Hosp., Kaohsiung, Taiwan.
Rapid detection of toxins and pathogenic bacteria is a prerequisite for a huge
As an emerging disease, melioidosis case clusters have been occurred in
number of applications, including bio-security and defense, medical diag-
2009 (n=8) and 2011 (n=12), in a small area (13 km2), in southern Taiwan
nostics, environmental and food analysis. Over the last decade, the anthrax
after violent typhoons go through. The geographical topography of this cer-
or ricin findings in the US postal system clarified the urgent need to develop
tain area appears the hills (50-400 m) are surrounding on north, east and
rapid, sensitive and specific detection systems for biothreat agents. Currently,
south as well as seashore on west. By culture-confirmed and PCR-based de-
the most widely used test to detect the presence of toxins is ELISA. However,
tection using soil (n=80) or water (n=36) specimens, the positivity to Burk-
these techniques cannot be performed by untrained personnel in the field
holderia pseudomallei was found among specimens that collected from the
and they are time-consuming. Remarkable advantages of electrochemical
north of this certain area but negativity in that collected from south of this
biosensing compared to common used optical methods are the high sensi-
area or out of surrounding by hills. By PFGE (pulsed-field gel electrophoresis)
tivity, simple operation, and the possibility of being used in portable instru-
analysis, the molecular linking of isolates between environments and pa-
ments for on-site testing. Here, we present an electrochemical detection
tients was demonstrated. In following as typhoon history, at least, Taiwan
technology applying biochips with immobilized antibodies against BoNT/A,
has been occurred 20 typhoon invasion from 2008 to 2010. If the typhoon
BoNT/B, BoNT/F, ricin and SEB. The detection platform allows simultaneous
appeared the maximum wind velocity of 40 m/sec and wind direction from
identification of these toxins within less than 25 minutes in an automated
northwest to southeast, the soil or water contaminated with B. pseudomallei
process. Detection limits for BoNTs, ricin and SEB are in the pg/mL to low
could be brought by wind vector from north and it associated with the oc-
ng/mL-range. We demonstrate identification of ricin from different Ricinus
currence of melioidosis case clusters in south of this certain area after ty-
communis varieties, whereas closely related lectins result in no signal. Fur-
phoon go through. However, out of this area (appeared hill barriers), no
thermore, we show specific detection of BoNTs and SEB directly from various
melioidosis case was occurred. We suggested that occurrence of melioidosis
Clostridium botulinum or Staphylococcus aureus strains, respectively. Finally,
case clusters was associated with the certain climate condition and geo-
we present the detection of different toxins in various sample matrices inoc-
graphical topography in southern Taiwan.
ulated with toxins applying minimal sample preparation. Furthermore, we in-
troduce the application of the electrochemical detection platform for rapid
109 identification of bacteria possessing high potential for use in biological war-
fare. After rapid PCR amplification labeled amplicons of different Bacillus sp.
Environmental Contamination of Goat Farms with Coxiella bur- are detected within ten minutes using DNA/DNA hybridization on biochips.
netii During a Q Fever Outbreak Limits of detection and specificity using several Bacillus sp. strains are pre-
G. J. Kersh, K. A. Fitzpatrick, A. V. Kondas, A. J. Roche, J. S. Self, R. A. Priest- sented. The presented results demonstrate the potential of electrochemical
ley, A. Bjork, A. D. Anderson; CDC, Atlanta, GA. biochips for sensitive and universal on-site detection of biothreat agents.
Coxiella burnetii is an obligate intracellular bacterium that cause4s the
zoonotic disease Q fever. It is considered a potential bioweapon due to its
stability, low infectious dose, and ease of transmission by inhalation. A con- 111
cern for C. burnetii is its ability to maintain infectivity and spread easily Detection of Botulinum Neurotoxin Serotypes A and B Using
through the environment carried by the wind or on animals. However, a quan-
Chemiluminescence and Electrochemiluninescene Immunoas-
titative evaluation of C. burnetii environmental spreading from a known
says in Food and Serum Matrices
source has not been reported. In the summer of 2011, an outbreak of Q fever
L. W. Cheng, L. H. Stanker; USDA, Albany, CA.
on multiple goat farms in Montana and Washington took place. Large num-
Botulinum neurotoxins (BoNT) are some of the most potent b4iological tox-
bers of organisms in the placentas of goats resulted in goat abortions and at
ins with BoNT serotypes A and B being most prevalent in foodborne contam-
least 17 cases of Q fever in humans. Here we report quantitative detection of
inations. BoNTs are also likely targets for use in intentional adulteration of
C. burnetii DNA on farms involved in the outbreak. Sponge, vacuum, and bulk
food or animal feeds and are thus classified as Select Agents. In our labora-
samples were taken, and inhibitor-free DNA isolated. Quantitative PCR tar-
tories, high-affinity monoclonal antibodies (mAbs) have been developed for
geting the IS1111 insertion sequence was performed to determine genome
the detection of BoNT/A and BoNT/B in traditional sandwich type ELISA as-
equivalents (GE) of C. burnetii in the environmental samples. Seven farms
says. To improve toxin detection sensitivity in complex matrices of food and
were included and between 14 and 138 samples were collected on each farm.
sera, we tested the use of electrochemiluminescence (ECL) type immunoas-
C. burnetii DNA could be detected at all seven of these farms, and of 390 total
says using a Meso Scale Diagnostic instrument. Both ELISA and ECL im-
samples, 274 (70.3%) were positive. Of the 274 positives, 202 (73.7% of pos-
munoassay formats detected BoNT/A and BoNT/B with better detection
itives) contained less than 100 GE per gram, but on the two farms that were
sensitivities than the “gold standard” mouse bioassays. Both immunoassays
most closely linked to human Q fever cases, 20 samples contained greater
had limits of detection of 5 pg/ml for BoNT/A and 20 pg/ml for BoNT/B in
than 10,000 GE per gram, with 5 samples from goat breeding areas having
buffer matrices, below mouse lethal dose levels. Although detection sensi-
greater than 1 million GE per gram. Further sampling on these two farms in-
tivities of ECL and ELISA platforms were similar in buffer matrices, ECL as-

BIODEFENSE PROGRAM & ABSTRACTS 77


M O N D A Y O R A L A B S T R A C T S | 3:00–4:00 p.m.

says outperformed ELISA in detection sensitivity in most liquid food matrices. each strain were exposed inside a whole-body exposure chamber to aerosols
ELISA and ECL immunoassay platforms remain fast and simple alternatives to of RVFV generated with a Collison 3-jet nebulizer controlled by the AeroMP ex-
the mouse bioassay for use in the routine detection of BoNTs. posure system. Results: After aerosol infection, Wistar-Furth rats died of a
severe hepatic disease within 4 - 6 days post-infection (LD50 = 2 pfu). High
viral loads were seen in the liver, lung, and other peripheral organs. The LD50
Highlighted Oral Abstract Presentations for RVFV in ACI rats was 124 pfu, with a survival time between 8 and 14 days
post-infection. ACI rats succumbed to neurological disease which included
010. Viral Agents signs such as rolling in cage, head tilt, circling, and abnormal behavior. High
Monday, February 27, 2012 | 3:00 PM - 4:00 PM levels of virus were detected in brain tissue but not in peripheral tissues. Un-
Ambassador Ballroom like what has been reported for parenteral inoculation with RVFV, Lewis rats
succumbed to neurological disease after inhalation of RVFV; the LD50 was
112 pfu and the time to death ranged from 7 - 9 days. Conclusions: WF and
112 ACI rats appear to be potential models for the hepatic/hemorrhagic and neu-
First Evidence of Hantavirus Circulation in Terrestrial Small rological disease outcomes in humans, respectively. However, Lewis rats were
Mammals from Madagascar susceptible to neurological disease, which contradicts historical data for par-
J-M. Heraud1, N. Razafindralambo1, T. Andrianaivo2, M-M. Olive1, V. L. Soari- enteral inoculation of RVFV. It is not clear whether this difference is a result
malala2, S. M. Goodman2, J-M. Reynes3; 1Inst. Pasteur, Antananarivo, Mada- of the route of exposure or other factors. Additional studies are ongoing to
gascar, 2Association Vahatra, Antananarivo, Madagascar, 3Inst. Pasteur, Lyon, further characterize the disease outcome after aerosol exposure to RVFV in
France. all 3 strains of rat.
Background: Hantaviruses infect terrestrial small mammals and occasionally
humans. In human, the symptoms can be severe hemorrhagic fevers or acute
pulmonary syndromes. Previous study have shown the presence of antibod-
114
ies directed against Hantaan virus in R. rattus and R. norvegicus specimens. Interplay between RVFV and the Host NF-KappaB Signaling
Nevertheless, no study have shown viral circulation in small terrestrial mam- Cascade
mals of Madagascar. Material and Method: During two years, we collected A. Narayanan, K. Kehn-Hall, L. Hill, S. Senina, R. VanDuyne, I. Guendel, R.
serum and organs from animals obtained in the forest of Anorana. We de- Das, A. Baer, C. Bailey, F. Kashanchi; George Mason Univ., Manassas, VA.
tected hantavirus RNA using RT-PCR performed with degenerated primers. Background: Rift Valley fever virus (RVFV) is a category A select agent, an
The products of amplification of the expected size were sequences and the emerging infectious agent and an agricultural pathogen. There are currently
sequences obtained were analyzed. Results: 586 small terrestrial mammals no FDA approved therapeutics targeted against RVFV and understanding the
from 17 different species were captured between October 2008 and March virus-host interactions will be critical to the development of novel thera-
2009. About half of the animals (n = 283) represents endemic taxa, while the peutics. Our studies of host phospho-signaling responses to RVFV infection
rest (n = 303) was R. rattus. We detected RNA of hantavirus and phyloge- have revealed that infection causes activation of multiple signaling events
netic analysis of the partial sequences obtained suggested the presence of including the NFkB cascade and that inhibitors of host signaling responses
two new hantavirus species. One was associated to the introduced species down regulate virus. Methods: We analyzed p65 phosphorylation following
R. rattus and an endemic rodent Eliurus majori of the family Nesomyidae. RVFV infection in cells by western blots. We utilized size exclusion chro-
These viral sequences were different from the sequence of Seoul virus asso- matography to study the nature of IKK complexes in infected cells. Using Ki-
ciated with R. rattus. The other hantavirus detected was associated to the nase assays, we demonstrate the enzymatic activity of the IKK complex. We
Tenrecidae species Oryzorictes hova and was close to sequences of viral utilized multiple inhibitors of the NFkB pathway to determine whether they
species associated to Soricomorpha from elsewhere in the world. Raw preva- can inhibit viral replication. We evaluated inhibitor toxicity using Cell Titer
lence was different between endemic and introduced species (2 and 50% re- Glo. We utilized nanoparticles with immobilized inhibitors to identify the
spectively). Conclusion: Studying arenaviruses and hantaviruses are of host targets. We performed LC-MS/MS to understand the nature of the IKK
interest in Madagascar due to high rate of endemism (100%) among native complex in infected cells. Finally, we carried out animal experiments to
small mammal species. Our study allowed us to detect for the first time, two demonstrate in vivo efficacy of NFkB inhibitors. Results: Our results suggest
different hantaviruses in rodents and tenrecs. These results will open new that the MP12 strain of RVFV causes an increase in the phosphorylation of
research projects, which will address the question of infection of these p65 by the classical pathway in different human cells. Our experiments point
viruses in the malagasy population. to the existence of a novel low molecular weight IKK complex in MP12 in-
fected cells which retains kinase activity. Among the multiple NFkB inhibitors
tested, we have identified inhibitors of the IKK complex that effectively down
113 regulate virus and the mechanism may involve binding to IKK-β. Finally, we
Distinct Differences in Disease Presentation after Aerosol show that INFAR-/- mice, when treated with NFkB inhibitors, have up to 96%
Exposure to Rift Valley Fever Virus Dependent on Inbred Rat lower viral load in the liver. Conclusions: Our data suggests that RVFV in-
Strain fection induces formation of a novel IKK complex in infected cells that is crit-
ical to RVFV replication. Inhibition of IKK down regulates virus in both in vitro
J. M. Bales, D. Powell, L. M. Bethel, D. Reed, I. Pandrea, A. L. Hartman; Univ.
and in vivo systems.
of Pittsburgh, Pittsburgh, PA.
Background: Humans infected with RVFV generally have a self-limiting febrile
illness, but a small percentage of infected people develop complications in-
cluding acute hepatitis, fatal hemorrhagic fever, and delayed-onset en-
cephalitis. Inbred rat strains differ in their susceptibility to disease after
parenteral infection with RVFV. The objective of our study is to determine the
disease course and outcome of inbred rat strains after aerosol exposure to
Rift Valley Fever virus (RVFV) with the goal of using different inbred strains to
model the spectrum of clinical outcomes in humans. Methods: Female rats of

78 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N D AY O R A L A B S T R A C T S

A structural analysis showed that the LpxD1 preferentially added a 3-OH-


115 C18 at 37ºC, whereas the LpxD2 added a 3-OH-C16 at 21ºC. Transcriptional
Interactions of Venezuelan Equine Encephalitis Virus with Host analysis using microarray confirmed that the lpxD1 is up regulated at 37ºC
MicroRNA Processing Machinery and lpxD2 is up regulated at 21ºC. Post-transcriptional analysis using puri-
C. A. King, N. Shafagati, I. Guendel, F. Kashanchi, K. Kehn-Hall; George fied enzymes in an in vitro assay showed that these enzymes have different
Mason Univ., Manassas, VA. optimal temperature (LpxD1 at 37ºC, and LpxD2 at 21ºC). Finally, to evalu-
Venezuelan equine encephalitis virus (VEEV) is a new-world alphavirus which ate the potential role of these enzymes in pathogenesis, we tested both
has been classified as a Category B select agent and potential bioterror deletion mutants in mice using a subcutaneous route of infection. The
weapon, both for its severe disease course in humans and equines, and for LpxD1-null mutant was completely attenuated in mice and protected mice
its ability to be transmitted by aerosol exposure. There are no current FDA li- from lethal wild-type challenge, whereas the LpxD2 was as virulent as the
censed vaccines or specific therapies against VEEV, making identification of wild-type organism. Thus, by restricting the bacteria’s ability to remodel its
potential therapeutic targets a priority. With this aim, our research focuses on membrane in response to temperature may represent a novel pathway for in-
the interactions of VEEV with the host cell microRNA (miRNA) machinery. tervention or vaccine development.
MiRNA are small non-coding RNA which act as master regulators of the cell
by downregulating or degrading messenger RNA, thus suppressing produc-
tion of the resultant proteins. Recent publications implicate miRNA interac- 117
tion in the pathogenesis of HIV, rabies virus, HCV, and other viral diseases, Development of a Luciferase Reporter System to Monitor
leading to the hypothesis that interactions with host miRNA machinery would Plague Infection
be important in a human cell model of VEEV infection. These interactions Y. Sun, J. Pennington, M. G. Connor, J. M. Warawa, M. B. Lawrenz; Univ. of
were tested by infecting cells in which specific miRNA machinery was inacti- Louisville, Louisville, KY.
vated through null mutation or siRNA knockdown. Inactivation of cytoplasmic Background: No effective plague vaccine is currently approved for use in the
components Dicer and TRBP caused little to no decrease in viral replication US, and concerns over antibiotic resistant strains of Yersinia pestis have em-
when tested in null-mutant cells or by siRNA knockdown, whereas siRNA phasized a need to develop new vaccine and therapeutic candidates. A
knockdown of cytoplasmic Ago-2 caused a decrease of approximately 2 log rapid, high-throughput plague model that allows for real time monitoring of
in viral genomic copies as measured by PCR, and 1.5 log in viral replication infection, while minimizing group sizes, would improve the early screening
as measured by plaques. Inactivation of nuclear components Drosha, DGCR8 steps of new anti-plague drug discovery. Methods: The lux operon from P. lu-
(Pasha), or Exportin-5 also resulted in significant decreases, of 3.5-5 log in minescens was integrated into the Y. pestis chromosome. Three promoters
viral genomic copies as measured by PCR, and a 2-3 log decrease in viral repli- were cloned to drive the lux operon, and light output was monitored using
cation as measured in plaque assay. Taken together, these findings indicate a luminescent plate reader (Biotek) or CCD camera (IVIS Spectrum). As con-
that the nuclear processing and mRNA binding steps are the most critical for trols, reporters were integrated into a αphoPQ mutant (attenuated for sur-
VEEV infection, and suggest that the host cell may use redundant or alter- vival in macrophages) and a αpla mutant (attenuated during animal
nate means of cytoplasmic miRNA processing during VEEV infection. This infection). Results: We tested three reporters during in vitro culture and in-
question presents ample opportunity for further study. tracellular infection of macrophages. We observed differences in light pro-
duction for each promoter. We were also able to differentiate survival
phenotypes in the macrophage model. Using one reporter, we compared in-
Highlighted Oral Abstract Presentations fection by WT Y. pestis or an attenuated mutant in both the bubonic and
011. Bacterial Agents pneumonic plague models. The reporter allowed us to visualize differences
in bacterial dissemination in real time. We also used lux reporters to show
Monday, February 27, 2012 | 3:00 PM - 4:00 PM differences in colonization phenotypes between Y. pestis and B. pseudo-
Palladian Ballroom mallei intranasal infections. Conclusions: Conventional animal models re-
quire groups of animals to be sacrificed at various time points to monitor
progression of disease. Here we demonstrate a lux -reporter strain can be
116 used to rapidly visualize differences in the progression of Y. pestis infection
Temperature-Regulated Membrane Remodeling is Essential for in real time. Advantages of this system are: 1) smaller number of animals
Francisella Pathogenesis per group, 2) less time required at each time point, 3) ability to follow the
Y. Li1, D. A. Powell1, S. A. Shaffer2, D. A. Rasko3, D. R. Goodlett2, C. R. H. Raetz4, course of the disease in the same animal over time, and 4) identification of
X. Wang5, R. K. Ernst1; 1Univ. of Maryland, Baltimore, MD, 2Univ. of Massa- unexpected differences in disease progression. We expect that this reporter
chusetts Med. Sch., Worcester, MA, 3Univ. of Maryland Sch. of Med., Balti- system will allow for rapid screening of drugs during early steps of drug dis-
more, MD, 4 Duke Univ. Med. Ctr., Durham, NC, 5State Key Lab. of Food Sci. covery. Furthermore, this system can also be applied to virulence studies of
and Technology, Wuxi, China. different Y. pestis mutants.
Maintenance of membrane function is essential for bacterial pathogenesis.
Adaptive mechanisms through which the Francisella membrane is remod-
eled, in response to transmission from the environment to a host requires al-
terations in the structure of the lipid A component of LPS. Francisella
synthesize lipid A with an altered distribution of fatty acids in response to
growth at environmental (21ºC) versus host (37ºC) temperatures. Mass
spectrometric analysis shows that this is due to alterations mainly at the N-
linked hydroxy fatty acids of lipid A by the acyltransferase, LpxD. In this
study, we indentified two N-acyltransferase enzymes, designed as LpxD1
and LpxD2 present in the genomes of all Francisella subspecies. To deter-
mine the role of the individual enzymes in membrane remodeling, LpxD1
and LpxD2 deletion mutants were made by allelic exchange. Complete lipid

BIODEFENSE PROGRAM & ABSTRACTS 79


M O N D A Y O R A L A B S T R A C T S | 3:00–4:00 p.m.

118 119
A Variant of LcrV, the Plague Protective Antigen and Needle Cap Hide and Seek: Changes in Francisella Surface Proteins during
Protein of Yersinia pestis that Blocks Type III Secretion Infection Reveal Ideal Vaccine Targets
K. Given1, A. Mitchell2, N. Miller1, O. Schneewind1; 1Univ. of Chicago, Argonne, J. Huntley; Univ. of Toledo Coll. of Med., Toledo, OH.
IL, 2Univ. of Chicago, Chicago, IL. Francisella tularensis is one of the most dangerous bacterial pathogens
Yersinia pestis, the causative agent of plague, employs a type III secretion known, due to its ease of aerosolization, low infectious dose, and ability to
machine to infect animal and human hosts. A key feature of its pCD1 viru- cause rapid, fatal disease in humans. Identification and characterization of F.
lence plasmid-encoded type III machine is the transport of effector Yops into tularensis virulence factors has been a central focus of many investigators,
immune cells, thereby blocking phagocytosis and with it the ability of infected yet little is actually known about how the bacterium binds to, invades, or ma-
hosts to clear the invading pathogen. Type III needle complexes are com- nipulates the host cell despite years of research. We have focused our efforts
prised of machine components in the bacterial inner and outer membrane on studying F. tularensis outer membrane proteins (OMPs), given their pe-
that enable the transport and assembly of the YscF needle protein. Each hol- ripheral localization and surface-exposed domains. Indeed, OMPs have been
low needle structure is capped by a ring-shaped molecule comprised of five shown to be key virulence factors for many disease-causing bacteria. Con-
subunits of LcrV, the plague protective antigen. LcrV is thought to fulfill sev- versely, OMPs also have been used as potent immunogens to stimulate pro-
eral different dynamic functions, which include the regulation of Yop effector tection against bacterial infections. As proof of principle, we previously
secretion and the translocation of these proteins via the YopB/YopD effector demonstrated that F. tularensis outer membrane vesicles (including OMPs)
pore across the plasma membrane of host immune cells. We discovered that were capable of protecting animal models from Type A F. tularensis pulmonary
the addition of more than three amino acid residues to the C-terminus of LcrV infection. In order to identify the most relevant OMPs to include in a future
abrogates type III secretion in response to calcium depletion even when mu- subunit vaccine, we have performed a series of transcriptional and proteomic
tant lcrV is expressed in Y. pestis strains with wild-type lcrV. Using an LcrV analyses to identify OMPs that are specifically up-regulated during in vivo in-
variant with a C-terminal Strep-tag, which is comprised of eight residues, we fection. Surprisingly, F. tularensis dramatically modifies its OMP profile dur-
observed binding to LcrG, the cytoplasmic chaperone for LcrV. A protocol was ing infection, with different OMP subsets being expressed in particular
developed that allowed the visualization of type III secretion needles from Y. tissues (lung, liver, or spleen) and a large number of OMPs being down-reg-
pestis. Strep-tagged LcrV was deposited at the tip of YscF needles in a man- ulated during mouse infection. Further, our results demonstrated that OMP
ner similar to wild-type LcrV. We propose that the extended variant of LcrV oc- expression in vivo varies dramatically from in vitro macrophage infections,
cludes the hollow needle, possibly by inhibiting the dynamic opening of the highlighting the fact that mammalian infections are complex processes that
LcrV lid, and abolishes its calcium sensing via the YscF protein, a prerequisite cannot be recapitulated in vitro. Finally, we compared immunoproteomic
for the transport of effector Yops into host immune cells. analyses with in vivo OMP expression data and found that the most im-
munogenic OMPs (those which stimulate the strongest antibody responses)
were typically down-regulated immediately following infection. Taken to-
gether, these studies provide novel information about pathogen “decoy” pro-
teins and offer a rationale strategy to develop safe and effective vaccines
against intracellular bacteria.

80 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S D AY O R A L A B S T R A C T S

blood and brain was identified. Similarly, a gene expression signature specific
Highlighted Oral Abstract Presentations to neuroinvasive and the non-neuroinvasive strains of VEEV were identified.
019. Immune Response Computational analysis identified pathways such as Tight junction signaling
and Granzyme-A signaling to be exclusively modulated during infection by
Tuesday, February 28, 2012 | 3:00 PM - 4:00 PM the neuroinvasive strains of VEEV. These gene signatures can be further ex-
Diplomat Ballroom plored to develop a biomarker signature against VEEV infection, help in un-
derstanding the mechanisms involved in VEEV neuroinvasion and identifying
221 host derived drug development targets against VEEV infection. Further stud-
ies correlating the host responses in the brain and blood using the gene ex-
Ornithine Lipids as Therapeutic Targets for Melioidosis pression data are in progress. This work was supported by funding from
M. Gonzalez-Juarrero1, R. Bowen1, W. Mwangi2, T. M. Eckstein1; 1Colorado DTRA. Opinions expressed herewith are those of the authors and do not re-
State Univ., Fort Collins, CO, 2Texas A&M Univ., College Station, TX. flect those of USUHS, BITS or FDA.
Burkholderia pseudomallei (BPS) is the etiological agent of melioidosis.
There is an increased interest in the pathogenesis of BPS following its clas-
sification as category B priority pathogen. Although antibiotics have been 223
proven to be effective tools in treating melioidosis, for unknown reasons they Dynamic Analysis of Respiratory Immune System Provides New
lack efficacy during the chronic and/or recurring stages of melioidosis. In-
Insights on Pulmonary Anthrax
terestingly, BPS does not develop a rapid drug resistance that could explain
D. Fiole1,2, A. Quesnel-Hellmann1, J. D. Mathieu1, J. Douady2, J-N. Tournier1;
the treatment failure and it seems that BPS is capable of modifying the im- 1
Inst. de Recherche Biomédicale des Armées, Grenoble, France, 2Univ. Joseph
mune system to allow its persistence. BPS contains large amounts lipids on
Fourier Grenoble, Saint Martin d’Hères, France.
the surface that could serve as target or tool for additional therapeutic op-
Lung efficiency as gas exchanger organ is based on the delicate balance of its
tions. Lipids were extracted and purified from BPS by standard methods.
associated mucosal immune system between inflammation and sterility. Ex-
Polar lipids were used to stimulate caprine dendritic cells and autologous T
ploration of lung immune system dynamic under infection has not been per-
cells. Intracellular staining for cytokines was analyzed by Flow Cytometry.
formed in detail so far, mainly because ventilation mechanic and chest
Bacteria were also grown in U937 for various times. Lipids of intracellular
movement impair microscope focus. We addressed this issue by synchroniz-
grown bacteria were extracted by standard methods. Lipids were analyzed
ing ventilation and imaging, allowing the collection of important information
by mass spectrometry. We identified several polar lipids involved in the reg-
about the early stages of pulmonary anthrax pathophysiology, a severe dis-
ulation of the immune response. The immune response for the in-vitro polar
ease due to inhalation of Bacillus anthracis spores. Mice are anesthetized
lipids seems to be balanced between stimulating and repressing the immune
and placed under a ventilator linked to the LSM 710 Zeiss™ microscope al-
response. There are four major polar lipid groups and two of them (rhamno-
lowing two-photon imaging. Alexa chemically labelled Sterne’s strain spores
lipids, ornithine lipids) are strong inducer of IFNγ while the other two groups
are instilled in CX3CR1+/gfp mice, which express green fluorescent protein
(phospholipids, unknown group) stimulate IL-10 production. However, this
(GFP) mainly in dendritic cells (DCs) and monocytes. Our study shows for the
balanced immune response dramatically changes when the pathogen enters
first time the infection dynamic in the lungs after inhalation of B. anthracis
cells. During the first hours of infection the lipid profile of BPS changes to a
spores, demonstrating that subepithelia DCs take up intraluminal spores
more IL-10 stimulatory profile with reduced amounts of rhamnolipids and or-
through trans-epithelial extensions. To our knowledge, the use of this novel
nithine lipids. Since ornithine lipids are the strongest inducer of IFNγ among
in vivo lung imaging protocol led to the first visualization of pathogen uptake
polar lipids they would be excellent tools to stimulate the host immune re-
by immune cells in the lungs, demonstrating the key role of dendritic cells
sponse during infection. Further investigation with chemically synthesized
acting as Trojan horses to penetrate the host defence system.
ornithine lipids will allow for additional analyses.

222 224
Visualizing Early Immune Response: Bacterial Specific Reor-
Identification of Host Responses Contributing Towards Neu-
ganization at the Nanoscale
roinvasion and Neurovirulence by Venezuelan Equine En- J. S. Aaron, Q. B. Smith, B. D. Carson, J. A. Timlin; Sandia Natl. Lab., Albu-
cephalitis Virus querque, NM.
1,2 1 1,2 3 3 3
P. Gupta , A. Sharma , M. Bhomia , J. Han , A. Yang , R. K. Puri , R. K. Ma- One of the early events in cellular innate immune response to gram(-) bacte-
heshwari1; 1Uniformed Services Univ. of Hlth.Sci., Bethesda, MD, 2Birla Inst. ria is the spatial reorganization of Toll-like Receptor 4 (TLR4) at the plasma
of Technology and Sci., Pilani, India, 3Food and Drug Admin., Bethesda, MD. membrane following binding of bacteria-specific lipopolysaccharides (LPS)
Venezuelan equine encephalitis virus (VEEV) is an alphavirus which may that coat the bacterial surface. Spatial reorganization of receptors is often a
cause a bi-phasic illness in humans where primary replication in lymphoid necessary component of downstream cell signaling and alhough bulk assays
organs is followed by entry into the central nervous system (CNS). The CNS have indicated that TLR4 receptors cluster upon stimulation by LPS, the
phase of infection causes encephalitis and neuronal death ultimately result- TLR4:LPS distribution has remained largely uncharacterized in intact cells
ing in mortality. Host factors which play an important role in VEEV neuroin- due to the mismatch between the optical diffraction limit and the nanoscale
vasion are poorly understood. Since the host-pathogen interactions are at which reorganizations occur. Over the past decade several optical ap-
reflected as specific changes in the gene-expression pattern of the host, we proaches have been introduced that effectively break the optical diffraction
compared the host responses during pathogenic and non-pathogenic VEEV barrier. Of these, Stochastic Optical Reconstruction Microscopy (STORM) rep-
infection to identify factors involved in neuroinvasion and/or neurovirulence. resents an attractive method for studying early immune response because it
CD-1 mice were infected with either neuroinvasive V3000 or partially neu- can provide nanometer scale resolution with specificity to the plasma mem-
roinvasive V3034 or non-neuroinvasive V3014 strain of VEEV. Whole genome brane. Using a simultaneous dual-color, STORM imaging system we imaged
microarrays were performed to obtain the differential gene expression ki- the organization of TLR4 at <30 nm resolution. We will show that challenge
netics in the blood and brain of infected mice. A signature gene expression with LPS produces a significant increase in TLR4 clusters on the cell mem-
pattern specific to VEEV infection independent of the type of strains in the brane of mouse macrophages, presumably due to recruitment of receptors to

BIODEFENSE PROGRAM & ABSTRACTS 81


T U E S D A Y O R A L A B S T R A C T S | 3:00–4:00 p.m.

lipid rafts. We also exploit dual-color imaging capabilities to visualize both


the receptor and LPS simultaneously permitting TLR4:LPS complexes to be
226
distinguished from TLR4 without LPS. We show for the first time that LPS de- EV-035: A New Compound Series with Broad Spectrum Antibi-
rived from E. coli, an immuno-stimulatory bacteria, result in significantly otic Activity against Burkholderia pseudomallei and Other Se-
higher LPS:TLR4 cluster sizes in the cell membrane, as well as a nearly two- lect Agent Pathogens
fold greater ligand/receptor colocalization as compared to immuno-evading S. Goldman1, D. Milligan1, H. Yao1, G. Liu1, G. Salerno2, P. Roussel2, J. Heim2;
LPS from Y. pestis. Additionally, preliminary results studying the effect of the 1
Evolva, Inc., Palo Alto, CA, 2Evolva SA, Reinach, Switzerland.
actin cytoskeleton has on TLR4 receptor reorganization will be presented. The need for new antimicrobial compounds is increasing due to the devel-
Collectively, these results provide novel insight into the nanoscale spatial dy- opment of resistance in bacterial pathogens and the pathogens used as
namics of TLR4 in response to bacterial-specific LPS and are important to elu- weapons of mass destruction. The compound class described here are novel,
cidate possible routes by which pathogens such as Y. pestis are able to small synthetic molecules with antimicrobial activity against the intracellular
circumvent the innate immune response. pathogen, Burkholderia pseudomallei, as well as several other select agents.
Evolva has developed this antibiotic series which acts against the clinically
validated target, topoisomerase IIa, with broad-spectrum antibacterial ac-
Highlighted Oral Abstract Presentations tivity and favorable drug-like properties. The EV-035 series exhibits antibac-
020. Therapeutics terial activity via specific inhibition of DNA gyrase and topoisomerase IV.
Moreover, the EV-035 series is active against many bacterial pathogen strains
Tuesday, February 28, 2012 | 3:00 PM - 4:00 PM that are resistant to quinolones and other bacterial topoisomerase inhibitors.
Ambassador Ballroom We have tested the EV-035 series for potency against B. pseudomallei in
mammalian cell intracellular infection models using B. pseudomallei and
Burkholderia thailandensis. Along with activity against Bps and Bth, we have
225 determined the activity of selected EV-035 molecules against other select
Novel Strategies to Enhance Current Antibiotic Therapies agents including: B. anthracis, F. tularensis, B. abortus, Y. pestis, F. tularen-
R. V. D’Elia, T. R. Laws, A. Carter, R. A. Lukaszewsk, G. C. Clark; DSTL, Salis- sis and B. mallei. Potency was determined by broth microdilution assays per-
bury, United Kingdom. formed according to CLSI guidelines. Bacterial and human topoisomerase
Current treatment options for personnel exposed to bacterial BW agents pre- inhibition assays were used to quantify on and off target activities and mam-
dominantly involve the use of licensed antibiotics. These treatments work malian cell cytotoxicity assays were used to identify compounds with suit-
very effectively when used shortly after exposure and, ideally, before the able toxicity profiles. Compounds were further characterized for bacterial
symptoms of disease associated with infection begin to emerge. The timing cidality and spontaneous resistance rates and for drug-like properties such
of the decision for military personnel to take these treatments in theatre is as solubility, serum effects and hERG channel inhibition. The potency, selec-
therefore important to the overall effectiveness of these medical counter- tivity and excellent preclinical toxicity profile of the EV-035 series suggest
measures (MedCMs). This decision is primarily informed by a “triggering that further development of these antibiotic molecules could help fill the
event” such as a detector signalling the presence of a BW agent. However, if need for novel, effective drugs for treatment of infections caused by biothreat
this information is not available the first “triggering event” may be the symp- agents as well as drug-resistant pathogens in the public health arena.
toms associated with infection being displayed in personnel exposed to the
BW agent. Initiating treatment at this point (i.e. post-symptomatically) is
likely to be less effective, putting personnel at risk of developing a life-threat- 227
ening disease. We are evaluating a novel therapeutic strategy which aims to A Single PMOplus Oligomer is Effective in a Cynomolgus
make existing MedCMs more effective and which may make it possible to ex- Macaque Marburg Virus Lethal Challenge Model
tend the timeframe (“window of opportunity”) within which antibiotics can be P. Iversen1, T. Warren2, A. Shrutleff2, J. Wells2, N. Garza2, K. Stuthman2, S. Van
administered. This project aims to establish whether the use of specific anti- Tongeren2, L. Dong2, D. Mourich3, L. Hauck1, J. Kugelman2, B. Beitzel2, G. Pala-
inflammatory compounds, that target fundamental immune pathways, can cios2, S. Bavari2; 1AVI BioPharma, Bothell, WV, 2USAMRIID, Fredrick, MD, 3AVI
be used to enhance the overall effectiveness of antibiotics by controlling the BioPharma, Bothell, WA.
unbridled (and ultimately futile) immune response typically found during the Marburgvirus (MARV) is a highly virulent RNA virus of the family Filoviridae
course of infection with the bacterium Francisella tularensis. Anti-HMGB1 an- and a causative agent of viral hemorrhagic fever (VHF). PMOplus oligomers
tibody, which targets the nuclear protein released by cells following injury or targeting NP and VP24 in the Ravn virus mouse lethal challenge model re-
infection, has been assessed during in vitro and in vivo models of infection. sulted in 65% (13/20) and 90% (18/20) survival while the combination of NP
Preliminary data suggests that this compound may be beneficial in enhanc- and VP24 provided 95% (19/20) survival. The MARV Musoke guinea pig lethal
ing the effectiveness of current MedCMs in treating the disease tularaemia challenge model resulted in 55% (6/11) and 0% (0/11) survival for NP and
caused by F. tularensis. Strategies, such as this, will be essential for the de- VP24, but the combination provided for 73% (8/11) survival. The current
velopment of a post-exposure MedCM system. study was to determine the contributions of oligomers targeting NP and VP24
individually versus the combination of the two in a cynomolgus
macaque(NHP) MARV lethal challenge model. Two independent and blinded
studies were conducted in which NHP were infected with a target dose of
1000 pfu of MARV Musoke administered subcutaneously. The first study in-
volved 4 groups; group 1 = 5 NHP with VP24, group 2 = 5 NHP with NP, group
3 = 5 NHP with VP24 and NP with all oligmers dosed at 7.5 mg/kg adminis-
tered daily by the iv route, and group 4 was a single NHP administered saline.
The second study involved 3 groups; group 1 = 10 NHP with NP and VP24,
group 2 = 10 NHP with NP only with all oligomers dosed at 15 mg/kg ad-
ministered daily by the iv route, and group 3 = 5 NHP administered saline.
Survival was determined at 36 (study 1) and 41 (study 2) days post infection.

82 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S D AY O R A L A B S T R A C T S

Survival observations in study 1 included 0/5 (0%) in group 1 targeting VP24, collection of more than 700 CYP51 antifungal inhibitors that show differing
3/5 (60%) in group 2 targeting NP, 3/5 (60%) in group 3 targeting both VP24 potencies against diverse fungi including yeast, dermatophytes, molds, and
and NP, and 0/1 (0%) in group 4, the saline control. Survival observations in plant pathogens. The molecules exemplified in the table below illustrate po-
study 2 included 9/10 (90%) in group 1 targeting NP and VP24, 10/10 (100%) tential new and safer treatments for a wide range of fungal infections.
in group 2, and 0/5 (0%) in group 3, the saline control group. We conclude the Compound C. albicans T. rubrum A. fumigatus CYP3A4 Selectivity
single PMOplus oligomer (AVI-6003) targeting NP provides for dose depend- MIC1 MIC1 MIC1 IC503 Ratio4
ent protection against a MARV lethal challenge in NHP.
VT-1148 ≤0.0001 ≤0.008 0.52 18 160000
VT-1161 ≤0.0001 0.016 8 65 720000
228 VT-1546 0.008 0.031 42 >200 9125
Therapeutic Efficacy of ST-246 in Cynomolgous Macaques Itraconazole 0.016 0.062 0.52 0.07 4.3
Challenged with Monkeypox Virus via Aerosol
Posaconazole ≤0.001 ≤0.001 ≤0.0622 0.05 50
A. T. Russo, K. Agans, T. Brasel, K. Dearen, T. Bailey, S. Storch, R. Baker; LRRI,
Albuquerque, NM. 1. MICs in µg/mL. 2. Complete inhibition of growth. 3. IC50s in µM. 4. CYP3A4
Monkeypox virus (MPXV) is a biological agent that could be used as a IC50 (µM)/C. albicans MIC (µg/mL). VT-1161 is also active against Coccid-
bioweapon. It is also a useful surrogate for smallpox in studies of potential ioides immitis. VT-1161 and related molecules have excellent pharmacoki-
medical countermeasures against this agent, which is regarded as one of the netic and CNS-penetration properties. Theability for these agents to treat
most devastating pathogens of human history. The deliberate release of pathogens in neural tissues provides for a clear path to rationally design ad-
smallpox is regarded as a possible threat by the US government. Smallpox is ditional drug candiates for the treatment of the Biodefense pathogen, Coc-
highly contagious and can cause severe disease and death in humans. cidiodes. Data related to these findings will be presented.
Human trials on candidate antiviral drugs cannot be done for either of these
agents so well characterized animal models of severe orthopoxvirus disease
are needed. NHPs were treated orally once daily for 14 days with ST-246 start-
Highlighted Oral Abstract Presentations
ing 1, 2, 3, or 4 days post MPXV aerosol challenge with 1x105 PFU. Clinical ob- 021. Diagnostics and Vaccines
servations were made twice daily for up to 45 days post challenge.
Pharyngeal swab and blood analysis, clinical pathology, chest x-rays, and le- Tuesday, February 28, 2012 | 3:00 PM - 4:00 PM
sion counts were conducted at routine intervals. Tissues were collected from Palladian Ballroom
each animal at necropsy for determination of viral load and histopathologi-
cal analysis. All treated NHPs (26) survived to the end of the study while only
1 of the 4 controls survived. Severity of illness in the treated NHPs correlated
231
with the delay of treatment post challenge with NHPs starting treatment 1 WITHDRAWN
day post-challenge showing virtually no signs of infection and those starting
treatment 4 days post challenge becoming nearly as sick as controls. These
animals exhibited changes in hematology and clinical chemistry consistent
232
with but less severe than those observed in control NHPs and also showed Construction of a Lateral Flow Immunoassay for the Diagnosis
moderate weight loss and elevated respiration rates. NHPs starting treat- of Melioidosis
ment two or three days post-challenge presented signs of illness intermedi- D. Reed1, N. Chantratita2, S. J. Peacock3, M. N. Burtnick4, P. J. Brett5, S. Ray-
ate between the extremes. Delay of treatment also correlated with increased chaudhuri6, R. Houghton6, T. R. Kozel1, D. P. AuCoin1; 1Univ. of Nevada Sch. of
presence of virus in pharyngeal swabs. Results indicate that significant sur- Med., Reno, NV, 2Mahidol Univ., Bangkok, Thailand, 3Univ. of Oxford, Oxford,
vival efficacy with ST-246 administration has been demonstrated in this United Kingdom, 4Univ. of South Alabama, Reno, NV, 5Univ. of South Alabama,
aerosol MPXV model up to 4 days post challenge and that delay of treatment Mobile, AL, 6InBios Intl., Seattle, WA.
is correlated with an increase in severity and incidence of signs of disease. The goal of our research is to develop a point-of-care (POC) immunoassay
that detects secreted Burkholderia pseudomallei bacterial antigens in pa-
tient samples for early diagnosis of melioidosis. A POC immunoassay could
229 greatly impact patient outcome because a high percentage of melioidosis pa-
Rationally Designed Broad-Spectrum Antifungals Agents for tients with acute septicemia die before culture results are available. Also, the
the Treatment of Biodefense Pathogens antibiotics used for empirical treatment of septicemia are not effective for B.
E. Garvey, S. Brand, W. Hoekstra, C. Yates, W. Moore, R. Schotzinger; Viamet pseudomallei. In order to develop a POC diagnostic our first objective was to
Pharmaceuticals, Morisville, NC. identify B. pseudomallei antigens that are secreted/shed during infection.
There is an urgent unmet biodefense need to develop therapeutic agents for We employed a novel approach termed In vivo Microbial Antigen Discovery
the treatment of the deadly infection coccidioidal meningitis (CM) caused by (InMAD) to identify secreted/shed B. pseudomallei antigens. By using this
Coccidioides, a fungus that can be weaponized and used in a bioterrorist at- method we have identified a candidate polysaccharide and numerous pro-
tack. CM is 100% lethal if untreated and early intervention with current ther- tein antigens that could be targeted for diagnosis. The B. pseudomallei cap-
apy results in a 40% mortality rate. Currently available antifungals agents sular polysaccharide (CPS) was one antigen identified by InMAD and a
also are limited by inconvenient routes of administration, toxicities, and the CPS-specific mAb was produced. The mAb was able to directly detect anti-
potential for drug-drug interactions. Viamet Pharmaceuticals, Inc, discovers gen in urine from melioidosis patients by Western blot and antigen-capture
and develops ‘best-in-class’ metallo-enzyme inhibitor drugs, including those ELISA. A prototype lateral flow immunoassay (LFI) was developed in collab-
that target the validated anti-fungal target, lanosterol demethylase (CYP51). oration with our private-sector partner InBios International (Seattle, WA). The
To our knowledge, the oral clinical candidate VT-1161 is the most potent and LFI has a sensitivity of 0.2 ng/ml when purified CPS is used as an antigen
selective yeast CYP51 antifungal compound described to date. It is currently source. Preliminary studies indicate that the LFI can detect CPS in a variety of
in Phase I clinical studies in the U.S. as a treatment for either invasive can- patient samples including serum, urine, pus and respiratory secretions.
didiasis or oncychomycosis. In addition, Viamet has a proprietary chemical

BIODEFENSE PROGRAM & ABSTRACTS 83


T U E S D A Y O R A L A B S T R A C T S | 3:00–4:00 p.m.

233
Immunization with Burkholderia pseudomallei Outer Mem-
brane Vesicles Protects against Pneumonic and Septicemic Me-
lioidosis
W. Nieves1, B. Judy2, A. Torres2, C. J. Roy3, L. A. Morici1; 1Tulane Univ. Sch. of
Med., New Orleans, LA, 2Univ. of Texas Med. Branch, Galveston, TX, 3Tulane
Natl. Primate Res. Ctr., Covington, LA.
B. pseudomallei (Bps), the causative agent of melioidosis, can be naturally
acquired through inhalation, ingestion, or inoculation. Due to its threat as a
biological warfare agent, vaccine development against Bps remains a high
priority. We previously demonstrated that immunization of mice with natu-
rally-derived outer membrane vesicles (OMVs) provided significant protec-
tion against lethal aerosol challenge with Bps strain 1026b. OMVs are
constitutively shed from the surface of Gram-negative bacteria and are en-
riched with lipids, polysaccharides, and proteins. We postulated that the
multi-antigen nature of OMVs could provide protection against additional
Bps strains and routes of infection. In this work, we examined the protective
efficacy of OMVs against systemic challenge with a heterologous Bps strain.
BALB/c mice (n=10) were immunized subcutaneously with 5 μg of Bps OMVs
+/- 10 μg CpG ODN 2395 on day 0 and boosted on days 21 and 42. Control
groups received CpG only or were untreated. Serial bleeds were performed
over the course of immunization and at the time of challenge for measure-
ment of serum antibody. One month after the last immunization, mice were
challenged intraperitoneally with 40 LD50 Bps strain K96243. Survival was
monitored for 3 weeks. OMV immunization provided significant protection
against high dose systemic challenge with Bps K96243 and protection was
significantly enhanced by the addition of CpG. Control groups displayed
100% mortality by day 3, while mice that received OMV/CpG demonstrated
80% survival over the 3 week study. OMV immunization induced high titers
of OMV-specific serum IgG1 and IgG3. Addition of CpG stimulated production
of IgG2a, indicative of Th1-type immune response which could account for
the increased protective efficacy of the OMV/CpG formula. In conclusion,
OMVs represent a promising vaccine against pneumonic and septicemic Bps
infection.

234
Sortase-Conjugation Generates a Capsule Vaccine That Pro-
tects Guinea Pigs against Bacillus anthracis
O. Schneewind, G. Garufi, Y-T. Wang, S-Y. Oh, H. Maier, D. Missiakas; Univ. of
Chicago, Chicago, IL.
Capsules protect bacteria against phagocytic clearance. Capsular polysac-
charides or polyglutamates have evolved also to resist antigen presentation
by immune cells, thereby interfering with the production of opsonophago-
cytic antibodies. Linking capsular material to a carrier protein stimulates its
presentation to the immune system. For many conjugate vaccines this is
achieved by a process of random chemical cross-linking. Here we describe a
new technology, designated sortase-conjugation, which generates a single
amide bond between the C-terminal end of a carrier protein and the capsu-
lar material. Sortase-conjugation was used to link the poly-D-γ-glutamic acid
(PDGA) capsule of Bacillus anthracis to the receptor binding domain (D4) of
protective antigen (PagA). When used as a vaccine, PDGA-D4 conjugate
elicited robust antibody responses against both capsule and D4. Immuniza-
tion with PDGA-D4 afforded guinea pigs complete protection against anthrax
challenge with wild-type B. anthracis Ames.

84 10th ASM Biodefense and Emerging Diseases Research Meeting


S U N D AY I N V I T E D S P E A K E R A B S T R A C T S

conducted under the auspices of the Animal Rule would require animals to
Focus Session reach a moribund state or die as a result of the agent or toxin, with no inter-
001. Animal Models vention. In the 2009 nonbinding FDA document “Guidance for Industry: Ani-
mal Models - Essential Elements to Address Efficacy Under the Animal Rule,”
Sunday, February 26, 2012 | 12:00 PM - 2:00 PM some consideration was given to a refinement in this approach where it states
Diplomat Ballroom that in some instances, supportive care should be administered to animals as
part of the study design. More recent discussions in regulatory circles have
001 Introduction and Setting the Stage
begun to acknowledge that animal efficacy studies are clinical trials in which
Louise Pitt; Center for Aerobiological Sciences, USAMRIID, Frederick, MD.
the animal patients should be provided some basic level of supportive care
Animal studies are the cornerstone of obtaining regulatory approval for prod-
that would be expected for a human patient. While this issue can be become
ucts to protect against biodefense agents and emerging threats. To date,
quite complicated based on the type of countermeasure being developed
guidance from the FDA regarding these studies has been minimal and some-
(e.g., vaccine versus therapeutic) and the model being used, this presenta-
times contradictory. To bring a single product to the point where it can move
tion will explore how we might consider and develop approaches that pro-
forward for regulatory approval requires the cooperation of a large commu-
vide consistent veterinary supportive care and early endpoint (i.e.,
nity, to include government agencies, contract research organizations and
euthanasia) criteria under different scenarios.
the biodefense pharmaceutical industry. Animal studies have to be designed
and executed in a manner that minimizes animal use and distress, while al- 006 Animal Rule Requirements vs. Expectations: How Can the
lowing for studies to be scientifically valid and comparable across research
Developers of Medical Countermeasures Succeed
sites. In order for data to be comparable between laboratories, clear well-de-
Elizabeth Leffel; PharmAthene, Inc., Annapolis, MD.
fined ethical study endpoints have to be established for each infectious agent
In 2002 the FDA finalized the “Animal Rule” which allowed for the approval
in each appropriate animal model. This requires an early understanding, dur-
of medical products whose efficacy cannot be demonstrated in humans for
ing initial model development, of what requirements will be needed to eval-
ethical reasons. To date, no new molecular entity has been licensed under
uate efficacy of medical countermeasures.
the Animal Rule despite large investments by the US government and indus-
002 Process and Implications of Animal Model Qualification try. Central to product approval via the Animal Rule is the development of re-
Rosemary Roberts; CDER, FDA, Silver Spring, MD. liable animal models that can be predictive of human benefit. Issues
Background: The final rule, “New Drug and Biological Drug Products; Evi- regarding the development of animal models to support product develop-
dence needed to demonstrate effectiveness of new drugs when human effi- ment will be discussed including the need for realistic understanding of the
cacy studies are not ethical or feasible” (21 CFR 314.600 for drugs; 21 CFR challenges associated with heavy reliance upon government and private lab-
601.90 for biological products), was published in May 2002. This regulation oratories (i.e. inter-lab variability with the models, methods, and challenge
is referred to as the “Animal Rule,” and provides a regulatory pathway for agents) and conducting trials in high containment facilities (i.e. animal mon-
the development of products for which human efficacy studies cannot be con- itoring and observation). Also discussed will be the need for a compelling ra-
ducted for reasons of ethics or feasibility. In these situations, the Food and tionale from the FDA regarding topics like added benefit and supportive care
Drug Administration (FDA) will rely on evidence from studies in animals to of animals in light of these challenges; the rationale for conducting studies
provide substantial evidence of effectiveness of these products. Methods: of these products in special populations that will derive no benefit also needs
Under the Animal Rule, FDA may grant marketing approval based on ade- clarification. Finally, an attempt to identify some steps needed for the indus-
quate and well-controlled animal efficacy studies when the results of those try to have a better understanding of the regulatory strategy to licensure
studies establish that the product is reasonably likely to produce clinical ben- using the Animal Rule, or other avenues making these products available for
efit in humans. Developing animal models that will yield efficacy results that use will be considered (e.g. EUA).
can be expected to be predictive for human response is challenging. To help
animal model developers with this challenge, the Agency is developing an Focus Session
Animal Model Qualification Program. Results: Product-independent animal
models can be evaluated and qualified as part of the qualification program 002. What’s Next? Pathogen Discovery From the
outlined for Drug Development Tools ( Draft Guidance for Industry: Qualifi-
cation Process for Drug Development Tools, October 2010). The initial ani-
Field to the Laboratory
mal model to be qualified should demonstrate the natural history of the Sunday, February 26, 2012 | 12:00 PM - 2:00 PM
disease caused by the threat agent and will have a defined context of use. Palladian Ballroom
After the natural history model is qualified, the model may be used for drug
development for efficacy indications. Qualified animal models will be pub- 007 Early Detection of Novel Biothreats Through International
lished in the public domain. Conclusion: The sponsor of an investigational Scientific Engagement and Medical Diplomacy
product can use a qualified animal model within its defined context of use Joseph Fair; Global Viral Forecasting Initiative, San Francisco, CA.
and the model will not require another review by the FDA. The use of quali- Imperative to the future of global health is the early detection, identification,
fied models will provide economies of time, animals, and resources in a drug and characterization of novel biological threats, followed by the implemen-
development program under the Animal Rule. tation of public health measures to mitigate the pandemic potential of such
events. Historically, the global health community has distinctly segregated
005 Exploring Approaches to Consistent Veterinary Care and human, animal and plant disease surveillance systems; however, given the
Euthanasia Criteria current state of their interconnectedness it is imperative that those systems
Jim Swearengen; NBACC, Fort Detrick, MD. be linked. We face a unique moment in history in that recent advances in tech-
With the initial implementation of the Animal Rule in 2002, the language re- nology, including molecular diagnostics, rapid sequencing, smartphone tech-
garding marketing approval criteria for a product developed and submitted nology, and the internet, has made it possible to create a virtual immune
under the Animal rule was quite stark with regard to study endpoints. The system that detects, identifies, and responds to biological challenges in all
guidance stated that animal study endpoints must be clearly related to the sectors. For such a system to function, there must be international buy-in
desired benefit in humans, generally the enhancement of survival or preven- from wealthy and impoverished nations and their populations, as well as non-
tion of morbidity. Many considered the guidance to imply that most studies governmental stakeholders - all with varying agendas, societal challenges,

BIODEFENSE PROGRAM & ABSTRACTS 85


S U N DAY I N V I T E D S P E A K E R A B ST R AC TS

and differences in health laws, use of biological materials and national de- 012 Population Genetics and Evolution of Burkholderia
fense policies. While the challenges are great, they can be overcome through pseudomallei
human relationships and understanding that is developed by international David Wagner; Northern Arizona Univ., Flagstaff, AZ.
scientific engagement and medical diplomacy. Burkholderia pseudomallei is a significant cause of mortality and morbidity
in endemic regions where it is widespread in the environment. We have found
008 Viral Discovery Using Pan-Pathogen Microarrays and Deep that recombination rates in B. pseudomallei, are very high, with a recombi-
Sequencing nation to mutation ratio that is higher than any other bacterial species yet
Charles Chiu; Univ. of California Sch. of Med., San Francisco, CA. reported. The high rates of recombination (i.e. genetic plasticity) observed
Traditional approaches to identify novel pathogens are limited in sensitivity within B. pseudomallei is a consequence of its lifestyle. It is a saprophytic
and scope. We still fail to diagnose approximately 25% of cases of respiratory organism that lives in soil where it comes into contact with numerous other
infections and more than 50% of diarrheal disease and encephalitis, and species, as well as conspecifics. As a result, it exhibits incredibly high-levels
threats from novel viruses such as the SARS coronavirus and 2009 pandemic of genetic/genomic diversity at even very small spatial scales. For example,
influenza H1N1 are continually on the horizon. New strategies are urgently several recent studies using multi-locus sequence typing have demonstrated
needed to increase the breadth, speed, and “throughput” of pathogen de- that multiple B. pseudomallei genotypes are present in individual soil sam-
tection. Here we will describe the utility of the ViroChip, a pan-viral microar- ples from sites in Northeast Thailand; this high genetic diversity is known to
ray, in combination with deep sequencing, in rapidly screening for the causes be associated with lateral gene transfers. This is in contrast to B. mallei, which
of unknown outbreaks in the field. We will highlight two recent applications is actually a clone of B. pseudomallei that exhibits a clonal evolutionary struc-
of the technology— (1) identification of a novel adenovirus responsible for a ture because it has evolved to live within equine hosts and therefore rarely
deadly pneumonia outbreak in a New World monkey colony with cross- comes into contact with conspecifics with which it could exchange genomic
species transmission to a human researcher, and (2) discovery of a novel material. Due to its high recombination rates, B. pseudomallei has what is
human hemorrhagic fever virus from the Congo, Africa. referred to as an “open genome”—it readily incorporates genomic material
from conspecifics and other species. As we recently reported, 10-15% of the
genome of a given strain of B. pseudomallei can be novel genomic compo-
Focus Session nents not previously observed in other whole genome sequences from this
003. Burkholderia pseudomallei: Understanding species. These novel components, found in some B. pseudomallei strains but
not others, comprise the accessory genome of this species and can be con-
Pathogenesis and Immunity to Guide the Next trasted with the core genome, which is comprised of genomic components
Generation of Medical Countermeasures found in all B. pseudomallei strains. Together, the accessory genome and the
core genome make up the pan genome— the full complement of genomic
Sunday, February 26, 2012 | 2:15 PM - 4:15 PM
material within this species.
Palladian Ballroom
013 Elucidation of Type 6 Secretion System as a Critical Viru-
011 Melioidosis: Clinical and Therapeutic Issues
Bart J. Currie; Menzies Sch. of Hlth. Res. and Dept. of Infectious Diseases,
lence Determinant of Burkholderia pseudomallei
David Deshazer; USAMRIID, Fort Detrick, MD.
Royal Darwin Hosp., Casuarina, Australia.
The Burkholderia pseudomallei K96243 genome encodes six type VI secre-
The clinical course of melioidosis following infection is likely to be determined
tion systems (T6SSs), but little is known about the role of these systems in
by a combination of: 1. Host risk factors for disease; 2. Mode of infection; 3.
the biology of B. pseudomallei. We constructed six hcp deletion mutants
Infecting bacterial load and 4. Putative Burkholderia pseudomallei strain dif-
(Δhcp1 through Δhcp6) and tested them for virulence in the Syrian hamster
ferences in virulence. Diabetes is the most important host risk factor and se-
model of infection. The 50% lethal doses (LD50s) for the Δhcp2 through Δhcp6
vere disease and death are very uncommon in a normal host if there is timely
mutants were indistinguishable from K96243 (<10 bacteria), but the LD50
diagnosis with appropriate treatment. Therefore B. pseudomallei should be
for the Δhcp1 mutant was >103 bacteria. Interestingly, similar results were
seen more as an opportunistic pathogen. It has been proposed that
obtained using the Madagascar Hissing Cockroach as a novel alternative
aerosolization of B. pseudomallei during severe weather events results in a
host. The hcp1 deletion mutant also exhibited a growth defect in RAW 264.7
shift from percutaneous inoculation to inhalation, thus accounting for more
macrophages and was unable to form multinucleated giant cells in this cell
severe disease and higher mortality. Nevertheless the occurrence, frequency
line. Unlike K96243, the Δhcp1 mutant was only weakly cytotoxic to RAW
and magnitude of such aerosolization in endemic locations remain entirely
264.7 macrophages 18 h after infection. The results suggest that the cluster
undetermined. Despite the recognition that melioidosis can be such a dra-
1 T6SS is essential for virulence and plays an important role in the intracel-
matically overwhelming infection, the specific virulence factors responsible
lular lifestyle of B. pseudomallei.
for severe disease remain surprisingly poorly elucidated, as does whether all
or only a subset of environmental B. pseudomallei are capable of causing
melioidosis. In addition to early diagnosis, the use of IV ceftazidime or car-
bapenems is the most important therapeutic factor, with longer durations of
IV therapy potentially curative without the need for a prolonged eradication
course of oral antibiotics.

86 10th ASM Biodefense and Emerging Diseases Research Meeting


S U N D AY I N V I T E D S P E A K E R A B S T R A C T S

014 Burkholderia pseudomallei Antibiotic Resistance Mecha- overexpression of both sRNAs result in a decrease in RNA levels of the 3’seg-
nisms: Impact on Therapeutic Strategies ment of both transcripts while the first gene is unaffected. FAS revealed that
Herbert P. Schweizer; Colorado State Univ., Fort Collins, CO. knockouts of these RNAs attach to and from pedestals on HeLa cells at lev-
Burkholderia pseudomallei is the etiologic agent of melioidosis. The bac- els far higher than the wild type strain. rtPCR also showed that these sRNAs
terium is an emerging pathogen and potential biothreat agent. B. pseudo- have no effect on the transcript levels of ler, the master regulator of the LEE
mallei infections are difficult to treat because of the bacterium’s intrinsic as well as shiga toxin. Conclusion: These two sRNAs play a vital role in the
resistance and biofilm and intracellular lifestyle. Antimicrobial therapy is post-transcriptional regulation of LEE4 and LEE5. This level of regulation may
biphasic and prolonged to address life-threatening acute infections, as well be important for the timing of infection.
as the organism’s propensity to enter into prolonged latency. This presenta-
tion will address the major antibiotic resistance mechanisms impacting ther- 017 Regulation of Francisella tularensis Virulence by Small
apeutic strategies for the treatment of B. pseudomallei infections. Special Non-Coding RNAs
emphasis will be on beta-lactam resistance mechanisms and efflux pump- John Gunn; Ohio State Univ., Columbus, OH.
mediated multidrug resistance. The discussion will address how knowledge Background: Francisella tularensis is a gram-negative non-motile intracellu-
gained from studies of clinical isolates and laboratory-generated mutants lar pathogen and the causative agent of the zoonotic human disease tu-
can be exploited to rapidly detect resistance and to redirect (clinical settings) laremia. It can survive in the environment and in a wide range of hosts,
or initiate (intentional release) proper melioidosis therapies. making the ability of this pathogen to sense its environment and respond ac-
cordingly key to pathogenesis. Most of the virulence genes of F. tularensis
identified localize to the Francisella Pathogenicity Island (FPI). FPI gene ex-
Focus Session pression is affected by various environmental cues and transcriptional viru-
lence regulators. The published literature suggests that MglA, SspA, PmrA,
004. Non-Coding RNAs and the Regulation of MigR, Hfq and FevR/PigR co-operate to regulate the Francisella virulence net-
Virulence work. Methods: Because of limited protein transcriptional regulatory factors
in Francisella, we have focused on sRNA where a genome wide in silico search
Sunday, February 26, 2012 | 2:15 PM - 4:15 PM identified 23 intergenic loci that encode putative sRNAs. Results: Two of the
Diplomat Ballroom sRNAs were predicted to target Francisella virulence regulators or FPI genes.
rprA is 55 nucleotides, located between FTT1564 and FTT1565 and predicted
015 Small RNAs and the Evolution of Yersinia Species
to bind to 42 targeted mRNAs including pmrA and FPI genes. rfrA is 88 nt, lo-
Wyndham W. Lathem; Northwestern Univ., Chicago, IL.
cated between FTT0618 and FTT0619 and predicted to bind to 51 targeted
Small, noncoding RNAs (sRNAs) are major regulators of gene expression in
mRNAs including FevR/PigR. Over expression of these sRNAs in the human
bacteria, affecting protein synthesis at the post-transcriptional level. In this
virulent F. tularensis subsp. tularensis demonstrated a 400-500 fold growth
session we will discuss the discovery and contribution to virulence of sRNAs
defect in J774.1, THP-1 and human monocyte-derived macrophages com-
in Yersinia pseudotuberculosis, a mild gastrointestinal pathogen, and Yersinia
pared to the wild type strain. The quantitative RT-PCR results demonstrated
pestis, the causative agent of the devastating disease plague. By taking a
that over expression of rprA down regulates several target genes including
deep sequencing approach, we have identified 216 previously unannotated
pmrA, fevR and FPI genes while over expression of rfrA down regulated tar-
sRNAs expressed in vitro by either Y. pseudotuberculosis or Y. pestis. While
get genes including fevR. rfrA and rprA regulation of target genes is primarily
most sRNAs are conserved between these two closely related species, the
independent of the RNA chaperone, Hfq. Deletion of the sRNAs confirm the
plague pathogen lacks 6 sRNAs that are present in Y. pseudotuberculosis and
regulation and phenotypes observed in overexpression experiments. Con-
expresses 5 sRNAs that are absent from Y. pseudotuberculosis. In addition,
clusion: These data suggest that rprA and rfrA regulate Francisella virulence
the expression of many sRNAs conserved between Y. pseudotuberculosis and
factors, though the exact mechanism behind this regulation is still unknown.
Y. pestis differs in both timing and dependence on Hfq, suggesting evolu-
Future studies will focus on all 23 sRNAs and their importance in Francisella
tionary changes in post-transcriptional regulation. We will discuss how these
pathogenesis.
changes may have contributed to the dramatic difference in virulence po-
tential between the species. 018 Legionella pneumophila Small Regulatory RNAs and Intra-
016 sRNA Regulation of EHEC Virulence cellular Multiplication
Vanessa Sperandio; Univ. of Texas Southwestern Med. Ctr., Dallas, TX. Sebastien P. Faucher; McGill Univ., Ste-Anne-de-Bellevue, Canada.
Background: Enterohemorrhagic Escherichia coli O157:H7 is a major cause of Background: Legionella pneumophila is a gram-negative human oppor-
hemorrhagic colitis and hemorrhagic uremic syndrome throughout the world. tunistic pathogen and the causative agent of Legionnaires’ disease, a pul-
One major virulence factor of this organism is a type three secretion system monary infection acquired by inhaling aerosols of contaminated water. It is
(TTSS) encoded by the five operons of the locus of enterocyte effacement able to infect and multiply in a broad range of protozoan and macrophages.
(LEE). Two of these operons, LEE4 and LEE5, are known to be post-transcrip- Several regulatory proteins are known to be involved in the regulation of its
tional processed. The first gene in these operons, sepL and tir respectively, virulence factors. However, as it has become evident that small regulatory
are cleaved off from the rest of the operon. The regulation of this post-tran- RNAs (sRNA) play an important role in the regulation of genes involved in
scriptional modification is largely unknown. The QseCE system is intimately pathogenesis, we wanted to test the hypothesis that sRNA also control viru-
involved in the regulation of virulence in EHEC and this system regulates two lence-related functions in L. pneumophila. Methods: Potential sRNA genes
sRNAs. The role these sRNAs play in the post-transcriptional regulation of were predicted by searching intergenic regions for Rho-independent termi-
the LEE will be investigated. Methods: Overexpression constructs for the nators and requiring evolutionary conservation of the sequences upstream
sRNAs were constructed. Knockouts were constructed through the use of them. A microarray was then constructed for the detection of 101 predicted
lambda red. Bacterial strains were grown in DMEM to an OD of 1.0 and RNA sRNAs from bacteria grown under a variety of conditions. Results: Twenty-
was extracted. Northern blots were probed with RNA probes against sepL two predicted sRNAs were shown to be actively transcribed in at least one
and espA or tir and eae. rtPCR was done to measure the expression levels of condition. Two of those sRNAs, called RsmY and RsmZ, are homologs of the
ler and stx2a. FAS was performed by infected HeLa cells with EHEC. Cells were CsrB sRNA that inhibits the action of the CsrA RNA-binding protein in other
infected for 6hrs and stained with FITC-phalloidin and propidium iodide and bacterial species. By using complementation assays we were able to show
visualized with fluorescent microscopy. Results: Northern blots show that that RsmY and RsmZ functionally link two major response regulators of L.

BIODEFENSE PROGRAM & ABSTRACTS 87


S U N DAY I N V I T E D S P E A K E R A B ST R AC TS

pneumophila virulence factors, the sigma factor RpoS and the RNA-binding
protein CsrA. Another identified sRNA is a homolog of 6S RNA, a widely dis-
tributed prokaryotic sRNA. Deletion of 6S RNA significantly reduced L. pneu-
mophila intracellular multiplication but had no effect on growth in rich media.
By using a whole genome microarray, 6S RNA was shown to positively regu-
late expression, in stationary phase, of Icm-Dot effectors, stress response
genes such as groES as well as many genes with unknown functions. Con-
clusion: Computational prediction and microarrays were successfully used
to identify several sRNAs. RsmY and RsmZ were shown to be the missing link
between major regulators of virulence in L. pneumophila. In addition, the L.
pneumophila 6S RNA homolog was identified and shown to be involved in
the regulation of genes related to intracellular growth.

019 A Unique Intergenic RNA Thermosensor Controls Yersinia


Virulence
Petra Dersch; Helmholtz Ctr. for Infection Res., Braunschweig, Germany.
Background: Expression of all Yersinia pathogenicity factors encoded on the
virulence plasmid, including the yop effector and the ysc type III secretion
genes, is controlled by the transcriptional activator LcrF in response to tem-
perature. Methods: Gene expression analysis, structure probing and toeprint-
ing analysis were performed to characterize temperature regulation of lcrF
on the transcriptional and post-transcriptional level. Results: We found that
a protein- and RNA-dependent hierarchy of thermosensors induce LcrF syn-
thesis at body temperature. Thermally regulated transcription of lcrF is mod-
est and mediated by the thermo-sensitive modulator YmoA, which represses
transcription from a single promoter located far upstream of the yscW-lcrF
operon at moderate temperatures. The transcriptional response is comple-
mented by a second layer of temperature-control induced by a unique cis-
acting RNA element located within the intergenic region of the yscW-lcrF
transcript. This region forms a secondary structure composed of two stem-
loops at 25°C of which the second hairpin sequesters the lcrF ribosomal bind-
ing site by a stretch of four uracils. Opening of this structure was favored at
37°C and permitted ribosome binding at host body temperature. Following
oral infections in mice, we found that Y. pseudotuberculosis patient isolates
expressing stabilized thermometer variants were strongly reduced in their
ability to disseminate into the Peyer’s patches, liver and spleen and have fully
lost their lethality. Intriguingly, Yersinia strains with a destabilized version of
the thermosensor were attenuated or exhibited a similar, but not a higher
mortality. Conclusion: The LcrF RNA thermometer is relevant during infection
and is the decisive control element providing just the appropriate amounts of
LcrF protein for optimal infection efficiency.

88 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N D AY I N V I T E D S P E A K E R A B S T R A C T S

fection with STEC serotypes other than O157, now reported more frequently
Plenary Session than O157, but about which less is known. Such culture independent tests
007. Outbreaks, Response, and Hindsight need to be coupled to reflex culture and subtyping in PulseNet labs, or ulti-
mately to advanced methods that provide the same subtyping and virulence
Monday, February 27, 2012 | 8:30 AM - 12:00 PM information critical to public health, or our ability to detect, investigate and
Regency Ballroom ultimately to prevent these outbreaks and infections will rapidly erode.
022 Swine Flu Emergence in Mexico 025 E.coli O104:H4: The Outbreak Experience from Germany
Edgar E. Sevilla-Reyes; Natl. Inst. of Respiratory Diseases, Mexico City, Mex- Reinhard F. Burger; Robert Koch Inst., Berlin, Germany.
ico. Germany experienced in 2011 one of the largest reported EHEC/HUS out-
Background: Infectious diseases have always been a threat to human popu- breaks worldwide with almost 3,000 EHEC and 855 HUS cases and 53 deaths.
lations. Followed by evidence of a number of influenza pandemics in recent Evidence for the role of raw vegetables as vehicle of the infection was rapidly
centuries, the threat caused by the emergence of the highly pathogenic H5N1 obtained. The pathogen was identified as EHEC O104:H4. Over 30 cohorts
viruses generated a number of actions around the world: development of pre- including a recipe-based restaurant cohort study revealed sprouts as vehicle.
vention, control and recovery programs. Situation: The surprising 2009 out- The food safety agencies (BfR, BVL) applied forward- and backward-tracing
break in Mexico and the pandemic that followed was closely followed and strategies. 40 outbreak clusters were linked to a farm producing sprouts in
documented by the media, the scientific community and health systems. In Lower Saxony using fenugreek seed from Egypt. The rare serotype O104:H4
this presentation I will review a number of actions and programs already in produced Shigatoxin 2 and had virulence characteristics of enteroaggregative
place by the time of the H1N1 outbreak and the difficulties found during im- E.coli indicating a virulence combination of two different pathogens. Sec-
plementation at multiple levels: in hospitals and small clinics, education cen- ondary infections in household partners, hospitals, and via food distribution
ters and public places, but also at the epidemiological surveillance level. The chain occurred, emphasising the importance of hygiene.
Mexican population was receptive and responded positively to most of these
actions. Outcome: The H1N1 health response in Mexico followed Interna- 026 Molecular Epidemiology and Characterization of the E.coli
tional Health Regulations along other preparedness and response programs. O104:H4 Outbreak Strain
A number of useful lessons were learned during the pandemic, and further- Alexander Mellmann; Univ. Hosp. Muenster, Muenster, Germany.
more, multiple economic, human and material resources were put in place Background: In the past early summer, we were confronted with the largest
all around the country that can be reapplied in the future. One of the main outbreak of hemolytic uremic syndrome ever caused by an exceptionally vir-
challenges today is not to forget those lessons and to continue building ulent Shiga toxin (Stx)-producing Escherichia coli O104:H4. Previously, this
stronger prevention, detection and control networks despite economic and serotype has rarely been associated with HUS. Methods: This presentation
political swings. will focus on the unique characteristics of this strain by comparing the out-
break isolates with other O104:H4 strains on the genome level. Results: In-
023 Swine Flu in the United Kingdom terestingly, the current outbreak strain and the historical isolates associated
Gail Thomson; Hlth. Protection Agency, Salisbury, United Kingdom.
with HUS carried genes typically found in two types of pathogenic E. coli, en-
During the 2009 pandemic the clinical response took on an additional mode
teroaggregative E. coli (EAEC) and enterohemorrhagic E. coli (EHEC). Phylo-
of communication and coordination - the virtual network. This framework
genetic analyses of the core genome genes indicated that the HUS-causing
been utilised during the SARS outbreak and had served the frontline well in
O104:H4 strains and the previously published sequence of the EAEC strain
2003. The Health Protection Agency’s (HPA) network of Intensivists caring for
55989 showed a close relationship but were only distantly related to com-
severe cases of H1N1 ran for six months and then again in winter 2010. With
mon EHEC serotypes like O157:H7 or O26:H11. Though closely related, the
weekly calls the UK intensive care community had an open line of communi-
outbreak strain differed from the historical strains in plasmid content and
cation and a forum to discuss with world influenza experts, public health and
fimbrial genes. Conclusion: The data determined enabled an evolutionary
to discuss challenging case management issues. Clinical colleagues from
model in which EAEC 55989 and EHEC O104:H4 strains evolved from a com-
across the globe contributed to the discussions. The outputs included guid-
mon EHEC O104:H4 progenitor, and suggest that by stepwise gain and loss
ance notes for the care of critically ill adults and children. During the pres-
of chromosomal and plasmid-encoded virulence factors, a highly pathogenic
entation consideration will be given to how this model could be built upon for
hybrid of EAEC and EHEC emerged as the current outbreak clone.
future events.

024 Overview of E. coli Outbreaks in the USA Symposium


Robert Tauxe; CDC, Atlanta, GA.
Since 1982, when Escherichia coli O157:H7 first emerged as a foodborne 012. Biodefense at the Dinner Table: Emerging
pathogen, the Shiga toxin producing E. coli (STEC) have driven major changes
in public health surveillance and food safety. Pulsenet, the national molecu-
Foodborne Illnesses
lar subtyping network housed at CDC, includes public health and food regu- Monday, February 27, 2012 | 4:15 PM - 6:15 PM
latory laboratories across the country, and has greatly improved detection, Diplomat Ballroom
investigation and control of bacterial enteric outbreaks by CDC, state and
local public health authorities. They have been linked to contaminated foods, 120 Mechanisms of Murine Norovirus Interaction with the In-
and to water, contact with animals and spread in daycare-centers. The dom- testinal Epithelium
inant pathway from the ruminant reservoir has been through beef. While the Christiane Wobus; Univ. of Michigan Med. Sch., Ann Arbor, MI.
number of outbreaks reported has increased, their size has dropped, and the Murine norovirus (MNV) is an enteric virus that replicates in the intestine of
incidence of STEC O157 infections had dropped by 44%, as prevention meas- its host. It has a tropism for murine macrophages and dendritic cells both in
ures driven by outbreak findings have been implemented, particularly for culture and in mice. However, it does not infect intestinal epithelial cells, rais-
ground beef. In recent years, outbreaks related to produce (including apple ing the question how MNV crosses the epithelial barrier to reach its target
cider, sprouts, and leafy greens) have also driven prevention efforts; further cells. To investigate the initial steps of MNV penetration of the intestinal mu-
measures are needed to reach the national target of 0.6 per 100,000 for STEC cosa, we are using two approaches; an in vitro M cell model of differentiated
O157. Use of Shiga toxin-based screening tests facilitates the diagnosis of in- epithelial monolayers and in vivo studies in mice. For in vitro studies, mICcl2

BIODEFENSE PROGRAM & ABSTRACTS 89


MO N DAY I N V I T E D S P E A K E R A B ST R AC TS

cells are plated into transwells for differentiation and some are also co-cul- for proposing novel strategies towards prevention and treatment of diarrheal
tured with murine B cells to promote M cell development. MNV was able to illnesses that overwhelmingly inflict the health of the world’s population.
translocate across the epithelial layer without disrupting tight junctions or
virus replication. Consistent with a role for M cells in viral transcytosis, signif-
icantly more MNV reached the basolateral compartment under co-culture con- Symposium
ditions, and MNV-1 was localized in M-like cells by confocal microscopy. To
study early time points during MNV pathogenesis and determine whether de-
013. Persistence and Inactivation of Biothreat
tected virus is replicating or input virus, we have adapted the use of light-sen- Agents and Emerging Pathogens
sitive, neutral red (NR)-containing virus for in vivo studies. The basic premise Monday, February 27, 2012 | 4:15 PM - 6:15 PM
of this technology is that NR-labeled virions are inactivated upon exposure to
light, whereas virions that have undergone replication are no longer labeled Palladian Ballroom
with NR and remain viable. Following oral infection of Balb/c mice with either 124 Persistence of Category A Select Agents
MNV-1 or CR3, only input virus was detected in the small or large intestine at Chuck Gerba; Univ. of Arizona, Tucson, AZ.
3 hrs postinfection (hpi), while by 24 hpi only replicated virus was detected. Fomites (intimate objects) contaminated with biothreat agents can play an
To determine whether M cells are critical for MNV infection in vivo, we condi- important role in the transmission of respiratory, enteric and dermal infec-
tionally depleted mice of M cells using an antibody against RANKL (receptor tions that is often not recognized. Biothreat agents can survive for days to
activator of NF-kB ligand), which is essential for the production of M cells, and months on common fomites depending on the type of organism (bacteria,
orally infected mice with neutral red containing MNV-1 and CR3. MNV-1 and virus, spore), fomite (stainless steel, paper, ceramic, glass, cloth), suspend-
CR-3 replication in the intestine was significantly reduced in RANKL-depleted ing media (water, mucus, feces, soluble organics) and relative humidity. Die-
compared to isotype-depleted or untreated mice. Taken together our data off is usually biphasic with greater die-off during drying of suspending media.
demonstrate that M cells mediate MNV transport across the intestinal ep- Information on inactivation of biothreat agents is essential in the develop-
ithelium and are required for MNV pathogenesis in mice. ment of models for predicting exposure and the potential success of inter-
ventions in indoor environments.
121 Picking the Lock: Stealth Invasion of Epithelial Barriers by
Enteric Pathogens 125 Persistence and Inactivation of Vaccinia Virus
Manuel R. Amieva; Stanford Univ. Sch. of Med., Stanford, CA. Joseph Wood; EPA, Research Triangle Park, NC.
A number of bacterial pathogens can cross the gastrointestinal epithelium Background: Variola major is a CDC Category A biological agent, and is the
to cause inflammatory gastroenteritis, enteric fever or sepsis. The receptors etiological microorganism for smallpox disease. Since the variola virus is un-
for epithelial cell invasion have been defined for several of these pathogens, available for research, we used the vaccinia virus (lyophilized) as a surrogate.
and include adhesion molecules like E-cadherin for Listeria monocytogenes, Methods: Tests were conducted to determine how long the vaccinia virus
integrins for Yersinia enterocolitica and Shigella sp. Of note is that many of would remain infectious in the environment, as a function of material, tem-
these receptors for entry are not found on the luminal side of the intestine, perature, and relative humidity. Tests were also conducted to assess the ef-
but are instead abundant components of the basolateral membranes of ep- ficacy of two common disinfectant chemicals (1% citric acid and quaternary
ithelial cells. This implies that pathogens first breach the epithelial tight junc- ammonium) in inactivating the virus. Approximately 107 plaque forming units
tions, or find alternative mechanisms to cross the epithelial barrier before (using Vero cells) per coupon material were used in the persistence and dis-
reaching their receptors for epithelial invasion. We will discuss a novel model infection tests. Results: The virus generally remained viable longer under low
of intestinal invasion by bacterial pathogens. We discovered that the tips of RH conditions, and after 56 days at low temperature and humidity, more than
intestinal villi are a specialized site of microbial invasion where the normal 104 PFU were detected on 3 of the 4 materials tested. In the inactivation tests,
process of cell senescence and cell extrusion allows basolateral receptors to neither of the disinfectants reduced the virus to non-detectable levels. Con-
be exposed at the luminal surface. clusion: The data presented elucidates some conditions when natural atten-
uation or common disinfectants may inactivate vaccinia and related viral
122 Multi-Dimensional Analysis of E. coli Colonization of Mam- agents.
malian Cells and Green Leafy Vegetables
Jorge A. Giron; Univ. of Florida, Gainesville, FL. 126 Inactivation of Bacterial Biothreat Agents in Water
The several classes of diarrheagenic Escherichia coli continue to represent a Laura Rose; CDC, Atlanta, GA.
significant health burden in developed and developing countries, particularly Concern for the security of drinking water supplies has prompted questions
in the pediatric population. In the United States and other industrialized about the survival of biothreat bacteria in water and the efficacy of current
countries, Enterohemorrhagic or Shiga-toxigenic E. coli (EHEC or STEC) disinfection practices to control contamination. We investigated the survival
strains of several serotypes are associated with serious intestinal and kid- of six species of non-spore forming biothreat bacteria (Burkholderia pseudo-
ney illness due to the consumption of tainted beef and agricultural products. mallei, Burkholderia mallei, Brucella suis, Brucella melitensis, Yersinia pestis,
The alarming outbreaks of bloody diarrhea and hemolytic uremic syndrome and Francisella tularensis) in water without disinfectants. B. suis and Y. pestis
caused in the Summer of 2011 in Northern Europe by a new enteroaggrega- were culturable for up to 21 days, and B. pseudomallei was culturable up to
tive E. coli (EAEC) strain of O104:H4 serotype producing the Shiga toxin high- 30 days. All were metabolically active, as determined by esterase activity,
light, undoubtedly, the significance of pathogenic E. coli and the plasticity of after 30 days. The non-spore forming bacteria and Bacillus anthracis spores
their genomes that allows the emergence of variants with novel and high in- were tested for susceptibility to free available chlorine (FAC, ≤ 1 - 4.8 mg/L),
fective and disease causing capabilities, as well as the need to better un- monochloramine (1 or 2 mg/L), chlorine dioxide (0.25 or 2.0 mg/L) and ul-
derstand the function of putative virulence factors present in the E. coli traviolet light (254 nm). The non-spore forming bacteria were susceptible to
pangenome against the possibility that similar outbreaks may occur anytime all disinfectants tested within reasonable contact times, with some disinfec-
elsewhere in the world. The strategies that EHEC O157:H7 utilizes to colo- tants more effective than others. B. anthracis spores were more resistant than
nize human, animal and epithelial will be discussed. The dissection of the the non-spore forming bacteria to all disinfectants tested, with chlorine diox-
mechanisms that these bacteria employ to interact with their host cells will ide more efficacious for biofilm associated spores and FAC more efficacious
help better understand the role of the several pili and other products in the for planktonic spores.
different E. coli lifestyles, outside and within the host, and establish a ground

90 10th ASM Biodefense and Emerging Diseases Research Meeting


M O N D AY I N V I T E D S P E A K E R A B S T R A C T S

127 Decontamination of Bacillus Spores from Water 130 Characterizing 24-48h Tick Saliva Proteins to Understand
Using Germinant Early Stage Tick Feeding Biology
Jeff Szabo; USEPA/NHSRC, Cincinnati, OH. Albert Mulenga; Texas A&M Univ., College Station, TX.
Background: Bacterial spores are persistent on drinking water infrastructure Through out the feeding process ticks differentially inject proteins to regulate
and common decontamination methods such as flushing and chlorination feeding, acquisition and transmission of disease agents. In our research we
have had limited success. Germination was evaluated as an enhancement to are interested in discovery and characterization of tick saliva that are injected
disinfection of Bacillus spores from drinking water infrastructure with free into the host during the first 24-48h of the tick feeding process. To do this
chlorine and flushing. Methods: A pilot scale pipe loop was outfitted with we have generated antibodies to 24-48h tick saliva proteins. We are using
iron (corroded) and cement-mortar coupons, which were conditioned in tap these antibodies to mine for cDNAs that encode tick saliva proteins. Here we
water for one month. Bacillus globigii spores were injected into the loop and have described characterization of an apparent universally conserved tick
allowed to adhere for 2 hours. Germinant was added after the adhesion anticoagulant protein present in Amblyomma americanum and Ixodes scapu-
phase, and allowed to contact the spores for an additional 2 hours. Germinant laris. The role of these proteins and others in tick feeding regulation and con-
was flushed out of the loop, and chlorination followed by flushing was per- sequently tick borne disease agent transmission is discussed
formed. Experiments only using chlorination and flushing were also per-
formed to determine the effectiveness of the germinant. Results: 131 Tweets from Ticks: Modulation of Vector-Specific Gene Ex-
Decontamination with free chlorine at 5 mg/L was ineffective (~0.2 log re- pression by BadR in the Agent of Lyme Disease
moval) on iron and achieved a 1.8-log reduction on cement mortar. Increas- Janakiram Seshu; Univ. of Texas, San Antonio, TX.
ing free chlorine concentration to 25 mg/L resulted in 1.2 and 2.2-log Borrelia burgdorferi, the agent of Lyme disease, is intimately dependent on
reductions of spores on iron and cement-mortar, respectively. Flushing after its arthropod and vertebrate hosts for a variety of nutrients as it lacks several
disinfection provided additional reduction, but spores persisted in each case metabolic pathways. While there is a large body of information on how B.
except cement-mortar decontaminated with 25 mg/L, where they dropped burgdorferi adapts to vertebrate host-specific conditions following the bite of
to undetectable levels. Adding a germinant (trypic soy broth) alone decreased an infected tick, there is a dearth of information on how it adapts to arthro-
the number of spores adhered cement-mortar and iron by 1.1 and 1.4 log, pod host when uninfected ticks feed on infected vertebrate hosts. Since reg-
respectively. Chlorination after germination at 5 mg/L further reduced spores ulators of carbon and nutrient utilization facilitate virulence and adaptive
attached to cement-mortar to undetectable levels. Spores were reduced to gene expression, we analyzed the role of BB0693, annotated as xylose
undetectable levels on iron coupons by chlorinating at 5 mg/L and then flush- operon regulatory protein (XylR1) in the patho-physiology of B. burgdorferi.
ing (increasing shear) after germination. Application of chlorine dioxide may XylR1 shares homology with two other ROK family repressors, NagC involved
also be an effective method of decontamiantion. Conclusion: This study in regulating N-acetylglucosamine (GlcNAc) utilization and Mlc, a global reg-
shows that germinating spores before application of disinfectant or flushing ulator of glucose metabolism. Immunoblot analysis indicated increased ex-
is an effective way to decontaminate drinking water infrastructure. pression of XylR1 in B. burgdorferi lysates grown under conditions mimicking
arthropod hosts compared to that of vertebrate hosts. A xylR1 deficient mu-
tant generated in the infectious B31A3 strain of B. burgdorferi exhibited in-
Symposium creased levels of pathogenesis related proteins synthesized under vertebrae
host-specific conditions, such as RpoS and OspC, suggesting that increased
014. Taking the Bite Out of Insect Bites: Biodefense levels of this regulator under tick-specific conditions could repress vertebrate
Implications of Vector-Borne Infections host-specific gene expression. Microarray analysis and quantitative real-time
reverse transcription-PCR analysis further confirmed drastic increases in the
Monday, February 27, 2012 | 4:15 PM - 6:15 PM
expression of these vertebrate host-specific determinants in the xylR1-defi-
Ambassador Ballroom cient strain. Based on these findings, we have designated this regulator as
129 Dissecting the Mechanisms of Tick Transmission from Borrelia host-adaptation Regulator (BadR). Collectively, the badR mutant has
Anaplasma to Francisella uncovered another layer of regulation in this spirochetal pathogen, and has
demonstrated BadR’s role as a putative carbon regulatory protein involved in
Susan M. Noh; USDA-ARS Animal Disease Res. Unit, Pullman, WA.
sensing the host-specific environment as well as modulating gene expres-
For ongoing transmission, vector-borne pathogens must make the remark-
sion necessary for survival in the ticks or vertebrate hosts. Further analysis
able transition from the mammalian host to the vector. In the case of intra-
of BadR mutant strain will facilitate identification potential targets to reduce
cellular pathogens, little is known about the molecular requirements of this
the incidence of Lyme disease.
transition. To identify these mechanisms, the transcriptional and proteomic
profiles of Ehrlichia spp. and Anaplasma spp. have been studied and shown
to be host cell-type dependent. For example, in A. marginale many surface
proteins are down-regulated in tick cells, while approximately 15 proteins,
including one surface protein are specifically up-regulated. In A. marginale
and A. phagocytophilum a majority of genes or gene products which are up-
regulated in the tick are hypothetical, thus function cannot be inferred. Ad-
ditionally genetic manipulation of these pathogens is difficult, further limiting
functional characterization. Because Francisella sp. are tick transmissible,
we used Francisella novicida, which is amenable to genetic manipulation, to
develop a tick transmission model to directly interrogate gene function at the
vector-pathogen interface with the long-term goal of identifying convergent
and divergent mechanisms of tick transmission.

BIODEFENSE PROGRAM & ABSTRACTS 91


T U E S DAY I N V I T E D S P E A K E R A B ST R AC TS

body (mAb) intended for treatment of inhalation anthrax. The mAb is highly
Plenary Session efficacious and resulted in no adverse effects in aerosol challenge or toxi-
017. Targeting Virulence Factors in Acute Infection cology studies. The ability to correlate the predictive value of nonclinical mod-
els to help ascertain potential safety risks in clinical research will be
Tuesday, February 28, 2012 | 8:30 AM - 12:00 PM discussed. It is anticipated that because the ECL-PA assay has been devel-
Regency Ballroom oped and tested in numerous nonclinical studies, it could be appropriately
applied in a clinical setting in the event of an ‘anthrax attack’ and this theory
135 Antimicrobial Peptide Modulation of Chemokine and Pro-
will be discussed.
Inflammatory Cytokine Responses: It’s All in the Timing
Kim A. Brogden; Univ. of Iowa, Iowa City, IA. 138 Targeting Cyclic Di-GMP Signaling to Prevent Biofilm
Background: B. anthracis produces lethal toxin (LeTx) composed of lethal fac- Formation
tor, a metalloprotease that cleaves mitogen activated protein kinase (MAPK) Chris Waters; Michigan State Univ., East Lansing, MI.
kinases (MKKs) and protective antigen, a carrier. Through cleavage of MKKs, Background: Biofilms are multicellular communities of bacteria encased in a
LeTx suppresses early cytokine responses by dendritic cells. However, recent network of protective extracellular polymers. In this state, bacteria are re-
studies show that suppression of cytokine production is not uniformly ob- sistant to environmental stress, predation, immune clearance, and anti-mi-
served suggesting that methods to enhance cytokine production may reverse crobial treatment. Cyclic di-GMP is a nearly ubiquitous second messenger in
the early events of immune suppression. Methods: Recently, we assessed bacteria that induces biofilm formation and represents an attractive target
the chemokine and cytokine response of human myeloid dendritic cells to for the development of new anti-biofilm strategies. Methods: A high-through-
Porphyromonas gingivalis hemagglutinin B (HagB) and human beta defensin put small molecule screen utilizing a cyclic di-GMP inducible luciferase tran-
3 (HBD3) mixtures. Results: HagB administered alone induces a robust scriptional reporter construct in Vibrio cholerae was developed. 66,000
chemokine and cytokine response. HagB administered with HBD3 does not. compounds/extracts from the University of Michigan Center for Chemical Ge-
Interestingly, HBD3 administered 1 hour prior or 1 hour after HagB, results in nomics chemical library were screened, and 184 hits possessing IC50 values
an early, robust, and sustained chemokine and cytokine response to HagB less than 10 micromolar were identified. The ability of these compounds to
(Figure 1 example). Conclusion: It is tempting to speculate that a mechanism inhibit biofilm formation and c-di-GMP synthesis both in vivo and in vitro was
involving HBD3 could ‘arrest’ the early events of LeTx immune suppression determined. Results: Two classes of compounds that possess broad-spec-
and ‘normalize’ the host immune response to the presence of B. anthracis trum anti-biofilm activity were identified. One class of compounds, repre-
toxins thus facilitating normal clearance and subsequent adaptive immune sented by the molecule we have designated anti-biofilm compound 1 (ABC-1)
responses to B. anthracis components. inhibits biofilms of multiple bacterial pathogens including, but not limited to,
Vibrio cholerae, a cystic fibrosis isolate of Pseudomonas aeruginosa, and
MRSA Staphylococcus aureus. The mechanism of action of ABC-1 is currently
under investigation. Moreover, we have identified 7 lead compounds that di-
rectly inhibit in vitro c-di-GMP synthesis by GGDEF enzymes. These com-
pounds also inhibit biofilm formation of V. cholerae. Two of these compounds
reduce total intracellular c-di-GMP. Conclusions: This research has identified
multiple small molecules that disrupt biofilm formation in a broad spectrum
manner, including the molecules that directly inhibit c-di-GMP synthesis by
GGDEF enzymes. These molecules have potential for the treatment of biofilm
based infections, and we are currently examining their in vivo efficacy in rel-
evant biofilm based disease models.

139 Identification of Inhibitors of DNA Adenine Methylation as


Novel Therapeutics
Petra Oyston; DSTL, Salisbury, United Kingdom.
Multiply antibiotic resistant strains of plague are emerging, driving a need
for the development of novel antibiotics effective against Yersinia pestis. DNA
adenine methylation (Dam) regulates numerous fundamental processes in
bacterial cells and alteration of Dam expression is attenuating for several
136 Anthrax Diagnostic Tools (Nonclinical and Clinical) for pathogens, including Y. pestis. The lack of a functionally similar enzyme in
Medical Countermeasure Development Under the Animal Rule humans makes Dam a suitable target for development of novel therapeutics
Elizabeth Leffel; PharmAthene, Inc., Annapolis, MD. for plague. We have identified a novel class of potent Dam inhibitors. The
The process of identifying a virulence factor or simply a marker of infection, most active compound was shown to have a competitive mode of inhibition
followed by developing an assay to detect such a biomarker can be difficult. with respect to DNA and a Ki of 6.46 nM. Moving from activity in vitro against
When developing a therapeutic drug under the FDA Animal Rule, efficacy has purified protein to activity against the bacterium requires inhibitors capable
to be tested in animals infected with the pathogen of interest. Care must be of penetrating into cells, accumulating to a sufficient level intracellularly, and
taken to ensure that animals have active infection so that treatment is not interacting with the target proteins to achieve an effect on the physiology of
prematurely initiated. For this reason, the successful identification of bio- the cell. One compound was found to meet these criteria, as indicated by in-
markers and the ability to test for them becomes critical. This discussion will hibition of DNA methylation in Yersinia cultures. The effects of this inhibition
focus on an assay to detect a biomarker of Bacillus anthracis. A historical on the physiology of the cell were widespread, and included altered expres-
overview of development of the electrochemiluminesence (ECL) assay to de- sion of known virulence traits, including iron acquisition and Type III secre-
tect protective antigen (PA) and how it was transitioned from the bench to tion. suggesting these inhibitors may attenuate bacterial virulence and
use as a diagnostic assay in regulated (21CFR58) nonclinical studies will be function as antibiotics.
presented. The ECL-PA was used to confirm infection in appropriate animal
models designed to evaluate the efficacy of an anti-toxin monoclonal anti-

92 10th ASM Biodefense and Emerging Diseases Research Meeting


T U E S D AY I N V I T E D S P E A K E R A B S T R A C T S

Symposium 237 Host Genetic Resistance to Gram-Negative Bacterial Infec-


tions: The Case of Yersinia pestis
022. Host Genetic Resistance to Infections Christian E. Demeure; Inst. Pasteur, Paris, France.
While laboratory mice are usually highly susceptible to Yersinia pestis, the
Tuesday, February 28, 2012 | 4:15 PM - 6:15 PM
SEG mouse exhibit an exceptional capacity to resist bubonic plague. Genetic
Diplomat Ballroom and physiopathologic comparisons with C57BL/6 mice were performed to
235 Host Genetic Resistance to Parasitic Infections: The Case identify genetic loci and immune mechanisms associated with resistance.
Strikingly, bacterial invasion in SEG occurred earlier and transiently compared
of Malaria
to C57BL/6 mice, which bacterial load was delayed but continuous until
Simon J. Foote; Macquarie Univ., Sydney, Australia.
death. Characterisation of cellular populations and cytokine production dy-
Malaria is a parasitic disease that kills around 1 million women and children
namics in various tissues revealed that the production of five cytokines (G-
annually. There is increasing resistance to current antimalarials with the time
CSF, KC, MCP-1, IL-1α and IL-6) involved in monocyte and neutrophil
between introduction of a new drug and the development of resistance de-
recruitment was significantly correlated with the early bacterial load in all or-
clining with each new addition. However there are natural, genetic resistance
gans. Indeed, an early massive macrophage recruitment in SEG spleen was
mechanisms to malaria. These usually focus on the red cell and are ubiqui-
observed. SEG and B6 macrophages were further compared to define the role
tous in areas of malaria endemicity. We are studying the host response to
of such cells in the early and effective innate immune response necessary for
disease in order to identify host targets for therapeutic intervention. We an-
surviving plague.
ticipate that host targets will provide antimalarials that are steadfast against
the development of resistance. We use several approaches to identify host 238 Host Genetic Resistance to Viral Infections: The Case of HIV
targets, these include a bioinformatics approach, an ENU mutagenesis screen Florencia Pereyra; Brigham and Women’s Hosp. and the Ragon Inst. of MGH,
and the development of a program looking at the role platelets play in a Boston, MA.
malarial infection. The results of some of these programs will be presented Genetic studies of infectious and inflammatory diseases have repeatedly
in this talk. Our ENU mutagenesis program screens offspring of ENU-treated shown strong associations within the MHC region of human chromosome 6.
malaria-susceptible mice. Mice surviving a murine model infection are prog- However, the basis for these associations remains elusive, in part because ex-
eny-tested and exons sequenced looking for segregating mutations. We have tensive linkage disequilibrium precludes assignment of causal variants. To
identified a number of mutations and several mutation affecting red cell cy- further define host genetic effects on the outcome of a chronic viral infec-
toskeletal proteins will be discussed. Bioinformatic analysis has identified tion, we performed genome-wide association analysis in a multiethnic co-
the heme-synthetic pathway as providing potential targets. We will present hort of HIV-1 infected persons, and coupled this to a novel imputation
data examining the effect of inhibition on two enzymes within this pathway. method to analyze the effects of individual amino acids within the classical
HLA proteins. Comparing 974 HIV-1 controllers, who maintain low virus loads
236 Host Genetic Resistance to Mycobacterial Infections: The
in the absence of therapy, to 2,648 persons with high virus loads and pro-
Case of Mycobacterium tuberculosis
gressive disease, we identified &gt;300 variants within the HLA region of
Philippe Gros; McGill Univ., Montreal, Canada.
chromosome 6 that reach genome-wide significance (P &lt; 5 x 10-8), and
Early response to infectious agents involves the concerted action of various
none elsewhere. Subsequent association analysis of amino acids within the
cell surface receptors and associated signaling pathways, and the rapid re-
HLA proteins pinpointed six independently significant residues in class I, five
sponse of different cell types such as NK cells, macrophages, dendritic cells,
lining the peptide binding groove, that explain the SNP associations in the
and T lymphocytes. These cells secrete regulatory cytokines and chemokines
GWAS and reconcile both protective and risk HLA associations with control of
that initiate a very rapid “inflammatory” response, direct early Th1 polariza-
HIV-1. These results implicate the nature of the peptide-HLA class I interac-
tion of the immune response, ultimately leading to destruction of the invad-
tion as the major genetic factor modulating control of HIV infection.
ing agent and establishment of protective immunity. IRF8 is a transcription
factor that plays a key role in all aspects of this response, both with respect
to ontogeny of different myeloid lineages, activation of host defenses in these
cells, including production of IL-12. Mice bearing mutations in IRF8 or its het-
erodimerization partner IRF1 are extremely susceptible to mycobacterial in-
fections, including pulmonary tuberculosis. Mutation in human IRF8 cause a
novel type of immunodeficiency associated with partial or complete absence
of dendritic cells, and susceptibility to mycobacterial infections. On the other
hand, IRF8 mutant mice are completely protected against cerebral malaria, a
disease in which excessive host-driven inflammatory response has lethal
pathological consequences. A genome-wide ENU mutagenesis screen has re-
cently been implemented to identify mutations that systematically protect
against cerebral malaria in vivo. The corresponding genes may play a critical
role in early host defenses against mycobacterial infections, including the
ontogeny and function of myeloid cells.

BIODEFENSE PROGRAM & ABSTRACTS 93


T U E S DAY I N V I T E D S P E A K E R A B ST R AC TS

Symposium 241 On the Molecular Evolution of Influenza A Surface Proteins


Joshua B. Plotkin; Univ. of Pennsylvania, Philadelphia, PA.
023. Dynamic Pathogens Make Difficult Targets Influenza viruses evolve rapidly. Their evolution reflects not only high muta-
tion rates, but also strong pressures for mutations that evade host immunity
Tuesday, February 28, 2012 | 4:15 PM - 6:15 PM or anti-viral drugs. A detailed understanding of which mutations are of adap-
Ambassador Ballroom tive value to the virus, and in what combinations, can improve our ability to
make rational decisions about control measures, including the composition
239 Diversity and Evolution of Pathogenic Escherichia coli:
and deployment of seasonal vaccines and drugs. I will discuss several recent
Lessons Learned from High-Throughput Genome Sequencing
developments, arising from computational studies, in our understanding of
David A. Rasko; Univ. of Maryland Sch. of Med., Baltimore, MD.
how hemagglutinin and neuraminidase evolve. I will also discuss ongoing
Background: The identification of the E. coli O104:H4 outbreak in Germany
studies into the relationship between the structure and stability of influenza
in the summer of 2011 was a tragic reminder that bacterial pathogens are
surface proteins, and their molecular evolution.
dynamic entities. High-throughput genome sequencing has become a feasi-
ble option for outbreak investigation at the diagnostic level as well as having 242 Burkholderia pseudomallei: A Bacterial Pathogen with a
potential in the routine diagnostic laboratory. Methods: Through high- Highly Adaptable Genome
throughput genome sequencing and comparative genomic analyses we have Paul Keim; Northern Arizona Univ., Flagstaff, AZ.
been able to identify regions of genomes that are shared or are unique in iso- The great biological and genomic diversity of the B. pseudomallei are
lates or groups of isolates that could be used as diagnostic markers. In the doubtlessly related, as this organism occupies multiple niches that require a
case of the German E. coli O104:H4 outbreak we examined the phylogeny of wide repertoire of genetic options. Environmentally, B. pseudomallei can be
the organism at the whole genome level as well as identified unique viru- commonly found in water and soil of certain geographic regions, with some
lence factor profiles among the outbreak isolates. Results: The isolates from evidence of plant associations. Its human pathogenic niche seems to be only
the E. coli O104:H4 outbreak in Germany were analyzed using a whole opportunistic and infrequent, but related to particular strain genotype.
genome phylogeny and demonstrated that these isolates were most similar Among strain genetic variation includes single nucleotide polymorphisms
to other enteroaggregative E. coli (EAEC), but had also acquired a phage con- (SNPs), variable number tandem repeats (VNTR) and genomic islands (GI).
taining the Shiga toxin genes. The phage was inserted into a genomic loca- Genomic islands are by definition due to horizontal gene transfer from some-
tion that is common in O157:H7 E. coli isolates. Additionally, we what distantly related bacteria. But the core genome is also affected by this
demonstrated that subclinical doses of ciprofloxacin activated the expres- process; we estimate that recombination is 20 to 30 times more important
sion of the Shiga toxin genes by greater than 80 fold. Virulence factor pro- than mutation in generating new MLST alleles. VNTR loci are numbers and
files of the outbreak isolates also revealed the carriage of an increased novel allele generation here is probably due to slip strain mismatch repair.
number of serine protease autotransporters (SPATEs) compared to other Most genomic diversify is generated in the environment prior to infection,
EAEC isolates. Conclusion: High-throughput rapid genomics was a central but the processes continue in vivo and change the phenotype during a single
tool utilized in the identification of the phylogeny and virulence factors as- host event. Some of these events are clearly adaptive in providing antibiotic
sociated with the isolates responsible for the E. coli outbreak in Germany in resistance or evasion of the immune responsible. Others such as VNTR mu-
2011. This methodology also provided information of a unique assemblage tations may be merely stochastic events and observable due to extraordi-
of virulence factors in the EAEC background of the isolate. With the expansion narily high rates.
of genomic sequencing projects the variants that do not fit our current typing
schema will be identified more often and we must aware of these rapidly
evolving pathogens. Symposium
240 Adaptive Arbovirus Landscapes and Implications for Dis- 024. Visual Discovery of Microbe-Host Interactions
ease Emergence Tuesday, February 28, 2012 | 4:15 PM - 6:15 PM
Scott C. Weaver; Univ. of Texas Med. Branch, Galveston, TX.
Arboviruses, like other RNA viruses, exhibit high rates of mutation and there- Palladian Ballroom
fore exist as genetically diverse populations within their hosts and vectors. 243 Viral Nesting: Rewiring the Host to Generate Organelle
Many infect people via direct spillover from enzootic cycles, but some can Platforms for Replication
also undergo secondary transmission cycles in domesticated animals to in- Nihal Altan-Bonnet; Rutgers Univ., Newark, NJ.
crease human exposure in agricultural settings. Moreover, several of the most Many RNA viruses upon infecting the host, generate membrane-bound repli-
important arboviral pathogens such as dengue and chikungunya viruses can cation organelles derived from the host’s intracellular membranes. The mem-
emerge from primate-mosquito enzootic cycles to circulate exclusively among brane surface of these organelles are then used as a platform on which viral
humans, transmitted by peridomestic mosquito vectors in urban habitats. replication machinery is assembled and viral RNA is synthesized.
These animal- or human-amplified secondary transmission cycles are some- We have been investigating the lipid microenvironment of these replication
times but not always associated with adaptation to the amplification hosts or platforms to identify panviral features that facilitate viral RNA synthesis. We
bridge vectors, and single amino acid substitutions often mediate these host have recently discovered that diverse set of RNA viruses all rely on host phos-
range changes. Using the alphaviruses chikungunya and Venezuelan equine phatidylinositol 4- kinases for replication. Specific viral proteins selectively hi-
encephalitis as examples, I will review several examples where host range jack these host enzymes, recruiting them to the replication membranes, in
changes, with or without adaptive mutations, have led to major epidemics order to generate a PI4P lipid enriched microenvironment which is critical for
and epizootics. I will also present evidence that, despite some having dra- viral RNA synthesis. In addition we have recently discovered a role for cho-
matic affects on fitness for infection and transmission, these host- and vec- lesterol in organizing the replication complexes and a pathway by which RNA
tor-adaptive mutations appear to be selected very inefficiently in nature, even viruses route host cholesterol lipids to these replication platforms. We will
during major epidemics with large amounts of virus circulation. Finally, I will discuss roles for PI4P lipids and cholesterol in regulating viral replication; de-
review experimental studies designed to test mechanistic evolutionary hy- signing antiviral therapeutics targeting the production of PI4P lipids; and how
potheses to explain restrictions on the Darwinian selection of adaptive al- studying viral biology can provide insight into cellular biology.
phavirus mutations in nature.

94 10th ASM Biodefense and Emerging Diseases Research Meeting


W E D N E S D AY I N V I T E D S P E A K E R A B S T R A C T S

245 Lighting Up Paramyxovirus Pathogenesis: Is Virus Eradica- for lung-specific delivery of organisms resulting in a disease model that more
tion a Reality, a Redundant Concept, or a Risk? closely resembles the clinically observed duration of disease culminating in
Paul Duprex; Boston Univ. Sch. of Med., Boston, MA. a systemic disease. The ability to monitor pulmonary-specific disease dis-
Understanding the myriad of interactions which occur when a pathogen in- semination in real time will support future studies investigating therapeutic
fects a primary target cell, spreads within an organism, establishes a sys- interventions of melioidosis.
temic infection and positions itself for transmission to the next susceptible
host is critical for the development of novel therapeutics. Such studies de- 247 Computer-Assisted Quantitative Analysis of Infectious
mand access to unpassaged “real world” clinical isolates which are grown in Disease Imaging
disease-relevant cells. This ensures that no untoward laboratory-adapted Daniel J. Mollura; NIH, Bethesda, MD.
mutations find their way into the genome as such changes can lead to erro- Background: Quantitative and molecular methods can greatly enhance de-
neous conclusions when in vivo pathogenesis experiments are performed, tection and study of preclinical and clinical infectious disease models. Meth-
particularly if the model of disease uses an outbred species where animal to ods: Computer-based quantitative measurements of CT and PET were used in
animal variations are greater. Recognizing the importance of these issues we animal models and human patients to measure pulmonary infectious dis-
developed a range of reverse genetics systems based on unpassaged ease. Results: FDG-PET provided in vivo measurement of pulmonary infec-
paramyxovirus isolates which can be used to produce recombinant viruses tion in Ferret models with strong correlations between histology and FDG
that illuminate pathogenesis both macroscopically and microscopically. uptake in H1N1 infected subjects. Computer-based measurement of respira-
These viruses express fluorescent proteins from an additional transcription tory infection accurately detected and quantified small airway inflammation
unit which allows infected cells to be detected with unprecedented levels of on computed tomography (CT). Computer-assisted detection (CAD) models
sensitivity. Measles virus (MV) is currently being considered for global erad- achieved higher diagnostic accuracy than comparison models for character-
ication, although there is no real consensus as to whether this is a reality. izing pulmonary disease. Conclusion: Molecular imaging using FDG-PET and
Following the elimination of smallpox, MV is one of the most infectious quantitative computer-based assessments of CT can be effective tools for
human pathogens with an R0 of between 12 and 18. This high level of trans- studying infectious pulmonary disease.
missibility is evident by the frequent importations into the US from endemic
regions and although vaccine uptake remains high the virus targets suscep-
tible hosts who have either not been vaccinated or have not generated a pro-
Plenary Session
tective immune response post-vaccination. If MV were to be eradicated 027. Immunity to Pulmonary Pathogens
globally discussions would ensue as to whether or not to terminate vaccina-
tion and destroy all laboratory stocks. Two critical factors would need to be Wednesday, February 29, 2012 | 8:30 AM - 12:00 PM
considered. First, how straightforward would it be to use synthetic biology Regency Ballroom
to recreate a fully pathogenic, highly transmissible biosafety level 2 agent?
252 The Host Immune Response to Pulmonary Virus Infection:
If that is the case then eradication is a redundant concept. Second, what is
the potential for closely related animal viruses to jump the species barrier It’s About Self Control
and fill the empty post-eradication niche? If this happened one might predict Thomas J. Braciale; Univ. of Virginia, Charlottesville, VA.
it would be associated with greater levels of morbidity and mortality and The lower respiratory tract is the site of oxygen uptake for the body. The com-
therefore be a risk. promise of this process resulting from severe respiratory virus infection is in-
compatible with life. Effective recovery from respiratory virus infection
246 Insights into Respiratory Melioidosis through Optical Di- necessitates that the infecting virus be eliminated from the respiratory tract
agnostic Imaging with minimal inflammation and injury to the surrounding tissue. During ex-
Jonathan M. Warawa; Univ. of Louisville, Louisville, KY. perimental murine infection with type A influenza virus (IAV), CD8+ CTL infil-
Background: Diagnostic imaging approaches have the potential to facilitate trating the IAV infected lungs produce high levels of both proinflammatory
a better basic understanding of the disease process in real time as well as the mediators (cytokines/chemokines) and the anti-inflammatory/regulatory cy-
potential to translate into rapid clinical diagnostics. In these studies, we char- tokine IL-10. We report on recent studies examining the interaction of CD8+
acterize the respiratory disease progression of Burkholderia pseudomallei in CTL with viral antigen expressing target cells in the IAV infected lungs, and the
several mouse models using optical imaging technologies to evaluate the re- consequences of these interactions for virus clearance, the regulation of IL-
latedness to descriptions of clinical melioidosis. Methods: B. pseudomallei 10 production and the control of excess inflammation during IAV infection
was engineered to constitutively express light from the chromosome and the (supported by USPHS grants AI-15608, HL-33391, and U19-AI-083024).
resultant lux+ strain was used to study respiratory melioidosis in a variety of
253 Requirements for Pulmonary Immunity Against Virulent
mouse models including BALB/c, SKH1, and C57BL/6J mice. Results: In-
tranasal infections of mice with B. pseudomallei result in significant colo- Francisella tularensis
nization of both the upper and lower respiratory tracts (URT/LRT), where Catharine Bosio; NIAID/Rocky Mountain Lab., Hamilton, MT.
colonization of the lung can be detected by imaging at 24 h post infection Background: Francisella tularensis is a Gram negative, facultative intracellu-
and earlier. In BALB/c and SKH1 mice, the lung represents the primary site of lar bacterial pathogen. Infections mediated by F. tularensis, i.e. tularemia,
infection. Interestingly, a capsule mutant was found to colonize the URT at can result in lethal disease within 5-7 days following inhalation of as few as
higher levels than the parent strain. In contrast, a novel intratracheal delivery 10 bacteria. Although several animal models are available to study infection
of bacteria results in LRT infection without involvement of the URT, with a mediated by virulent F. tularensis, identification of the specific immune com-
lower infectious dose than intranasal infections. Additionally, we observe an ponents that contribute to pathogenesis or resolution of infection have been
extended course of disease which progresses into a systemic disease more hampered due to the rapid mean time to death. Methods: To resolve this
closely resembling clinical melioidosis. Conclusion: B. pseudomallei is in- problem we developed a convalescent model of tularemia in C57BL/6 mice
fectious by several routes of entry, including the biodefense-related respira- that relies upon the host response for clearance and survival of infection. Re-
tory tract infection. Mice are excellent surrogate models for the study of sults: Using this model we found that, consistent with reports using more at-
melioidosis, however infections involving the URT do not accurately model tenuated strains of Francisella, alpha beta TCR+ cells and B cells were
clinical melioidosis given the non-clinical propensity of B. pseudomallei to required for survival. Although gamma delta TCR+ cells contributed to sur-
colonize the URT in mice leading to rapid death. Intratracheal infections allow vival they were not as critical as alpha beta TCR+ cells for resolution of F. tu-

BIODEFENSE PROGRAM & ABSTRACTS 95


T U E S DAY I N V I T E D S P E A K E R A B ST R AC TS

larensis SchuS4 infection. We also found that both IL-12p40 and IL-12p35
were essential for survival of SchuS4. Similarly, IFN-gamma was also required
since animals lacking IFN-gamma receptors failed to survive antibiotic ther-
apy following SchuS4 infection. Interestingly, unlike infections mediated by
attenuated strains of F. tularensis, SchuS4 provoked both CD4+ and CD8+
IFN-gamma producing cells in the lungs, whereas CD8+ cells dominated the
IFN-gamma response in the spleen. NK and gamma delta TCR+ cells made lit-
tle contribution toward IFN-gamma production after day 7 of infection. Con-
clusion: Together this model provides a means to identify key components of
the host response required for survival or exacerbation of tularemia and will
be useful in development of novel therapeutics and vaccines.

254 Infection of Human Alveolar Macrophages by the Q Fever


Agent, Coxiella burnetii
Daniel Voth; Univ. of Arkansas for Med. Sci., Little Rock, AR.
The intracellular bacterial pathogen Coxiella burnetii is a category B select
agent that causes human Q fever. In vivo, C. burnetii targets alveolar
macrophages in which the pathogen generates a lysosome-like para-
sitophorous vacuole (PV) for replication. C. burnetii infects a variety of cul-
tured cell lines that have collectively been used to model host cell
interactions. To obtain a clearer picture of an in vivo scenario, we are estab-
lishing primary human alveolar macrophages (hAMs) as an in vitro model of
C. burnetii-host cell interactions. Virulent C. burnetii pathotypes infect and
replicate in hAMs, albeit with unique PV formation properties. The pathogen
also modulates hAM signaling required for PV formation, including the pro-
tein kinase C and cAMP-dependent protein kinase cascades. We are currently
assessing the hAM cytokine response to infection and the role of the C. bur-
netii type IV secretion system to better define the host response to this highly
infectious pathogen. The hAM model represents a disease relevant platform
for identifying potential therapeutic targets to combat Q fever.

255 The Protective Role of Surfactant Protein-A During My-


coplasma pneumoniae Infection in Allergic and Non-Allergic
Airways
Julie G. Ledford; Duke Univ., Durham, NC.
Mycoplasma pneumoniae (Mp) frequently colonizes the airways of chronic
asthmatics and is thought to contribute to exacerbations of asthma. Surfac-
tant protein A (SP-A) has well-established functions in reducing bacterial in-
fections but its role in chronic lung diseases, such as asthma, is less well
defined. Our previous work shows that mice lacking SP-A have increased air-
way constriction during Mp infection compared to WT mice and that inhibition
of TNF-α transcription reduces their responses. Additionally, mice deficient in
SP-A have enhanced inflammation and airway constriction in an allergic/in-
fection model (Ova+Mp) and inhibition of TNF-α transcription prior to Mp in-
fection can also attenuate airway reactivity in SP-A-/- allergic mice to levels
measured in WT allergic mice. Our current research provides evidence that
Mp interacts with pulmonary mast cells in non-allergic airways and causes
activation/degranulation and that SP-A plays a role in protecting from Mp-
stimulation, thereby protecting the airways from damage due to the poten-
tial release of harmful products. Additionally, in Mp-infected allergic airways
SP-A interacts with eosinophils and mediates EPO-driven clearance mecha-
nisms and protects against Mp-induced exacerbations. Therefore, SP-A plays
a protective role in non-allergic and allergic airways via regulation of mast
cell and eosinophil cellular activation during Mp infection.

96 10th ASM Biodefense and Emerging Diseases Research Meeting


KEYWORD I N DE X

Keyword........................................Presentation Number Keyword........................................Presentation Number Keyword........................................Presentation Number

16S/18S PCR ...................................176 (D)


B C
A Bacillus anthracis toxins.........................042 (B) capillary electrophoresis .......................216 (J)

abrin ..................................................181 (E) Bacillus anthracis capsule .........042 (B), 151 (B) capsule ........................................156 (B), 234

Achromobacter xylosoxidans....................165 (B) Bacillus anthracis .......034 (B), 039 (B), 045 (B), capture...............................................069 (D)
.......059 (B), 75 (E), 081 (G), 095 (I), 153 (B), cat fleas ..............................................152 (B)
Acinetobacter baumannii.............101 (J), 158 (B) .................162 (B), 168 (B), 171 (B), 179 (D),
acute respiratory infection ..................027 (A) cell division ........................................051 (B)
...............................................189 (F), 214 (I)
adeno-associated virus ........................143 (A) cell envelope ......................................039 (B)
Bacillus cereus G9241 ...........................155 (B)
centrifugal microfluidic platform........065 (D)
Adenylate cyclase ..................................180 (D) Bacillus thuringiensis .............................164 (B)
challenge ............................................059 (B)
adjuvant................................204 (K), 208 (K) bacteria.................................147 (B), 177 (D)
chemical library .................................194 (G)
adoptive transfer .................................198 (K) bacterial agents ..................................178 (D)
chemokine receptor CCR7 .................214 (I)
aerosol.......029 (A), 030 (A), 038 (B), 146 (A), bacterial pathogenesis .........................049 (B)
.............................................149 (B), 161 (B) climate condition .....................................108
bacteriophage-based qPCR................066 (D)
African green monkeys ........................097 (I) clonality .............................................060 (B)
bacteriophages....................................050 (B)
alhydrogel ..........................................202 (K) Coccidioides ............................................229
baculovirus expression system.............142 (A)
allergen ..............................................182 (E) complex matrices ...............................055 (B)
Bangladesh .........................................074 (E)
alphavirus...........................................208 (K) compound screen ...............................031 (A)
beef chicken .......................................172 (B)
aminoglycosides..................................037 (B) computational fluid dynamics ..............102 (J)
benefits and risks.................................077 (F)
animal model development.................034 (B) coronavirus ........................................033 (A)
bioaerosol...........................................061 (B)
animal model...........029 (A), 030 (A), 038 (B) correlates of protection.......................200 (K)
biochip.....................................................110
Anopheles mosquito ............................099 (J) Coxiella burnetii ...................................190 (F)
biodefense concentration....................103 (K)
anthracis ..................046 (B), 159 (B), 160 (B) Coxiella ..............................................149 (B)
biodefense pathogen.................................229
anthrax lethal toxin..............................212 (I) Crimean-Congo Haemorrhagic
biofilm...............................................068 (D) Fever Virus .......................................141 (A)
anthrax toxin .......................................210 (I)
bioinformatics resource .......................187 (F) csaB ...................................................155 (B)
anthrax...................064 (D), 067 (D), 071 (E),
.................075 (E), 156 (B), 162 (B), 180 (D), biological Security..............................072 (E) cytopathic-effect ................................031 (A)
........................197 (K), 198 (K), 204 (K), 223 biological toxins .......................................110
antibacterial activity ...........................106 (K)
D
biosecurity ..............107 (K), 177 (D), 219 (K)
antibiotic resistance..035 (B), 172 (B), 195 (G) bioterror..............................................215 (J) decontamination..................................102 (J)
antibiotic .......................045 (B), 147 (B), 225 bioterrorism .......................................164 (B) dendritic cells...........................................223
antibody ............................................080 (G) biothreat pathogens............................065 (D)
antifungal .................................................229 Topic Categories:
botulinum neurotoxins .............................111
Below is a list of descriptions of topic cate-
antimicrobial agents............................165 (B) botulism ............................................091 (H) gories (letters in parentheses by each pres-
entation number refer to this chart).
antimicrobial peptide ...........................101 (J) broad-spectrum..................................192 (G)
anti-ricin ...........................................080 (G) A. Viral Agents
Brucella melitensis..................170 (B), 193 (G)
B. Bacterial Agents
antisense .....................................195 (G), 227 Brucella ..................................040 (B), 049 (B) C. Fungal Agents
antiviral ...........082 (G), 143 (A), 192 (G), 228 D. Diagnostics
BSL-2, BSL-3 ....................................220 (K) E. Environmental Detection
arenavirus...........................................144 (A) Burkholderia cepacia ..............................174 (B) F. Informatics and Genomics
assay...................................................064 (D) G. Therapeutics
Burkholderia mallei ..................038 (B), 173 (B) H. Vaccines
automation.........................................150 (B) Burkholderia pseudomallei.........043 (B), 096 (I), I. Immune Responses
autonomous .......................................073 (E) .................174 (B), 211 (I), 221, 226, 232, 233 J. Decontamination, Biosafety,
and Containment
autotransporter ...................................154 (B) Burkholderia thailandensis......................043 (B) K. Other and Information Only
avian influenza, inactivated vaccine.....084 (H) Burkholderia ........................................154 (B)

BIODEFENSE PROGRAM & ABSTRACTS 97


KEYWORD I NDE X

Keyword........................................Presentation Number Keyword........................................Presentation Number Keyword........................................Presentation Number

dengue ...............................................031 (A) Francisella ...............052 (B), 088 (H), 116, 119 invasion .............................................086 (H)
dentistry.............................................104 (K) Ionic propulsion .................................182 (E)
G
detection ..............................073 (E), 177 (D)
gamma irradiation ..............................199 (K) L
diagnosis ............................................178 (D)
diagnostic and therapeutic potential ....050 (B) genetic sequence analysis.....................076 (F) lab-on-paper .........................184 (E), 186 (E)
diagnostics ..................................067 (D), 232 GeneXpert ........................................063 (D) larvicidal agent.....................................099 (J)
DNA extraction.................................176 (D) genomics ............................................078 (F) LC16m8 ............................................090 (H)
DNA..................................................150 (B) genotyping .........................................190 (F) LcrV ........................................................118
doxycycline........................................193 (G) geographical topography ..........................108 lipid A......................................................116
drinking water quality ........................074 (E) Georgia .....046 (B), 047 (B), 048 (B), 159 (B), lipopolysaccharide ..............................034 (B)
.............................................160 (B), 168 (B)
DTRA....................046 (B), 047 (B), 048 (B), lipoteichoic acid ...................051 (B), 081 (G)
.............................................159 (B), 160 (B) growth temperature............................166 (B)
LtaS....................................................051 (B)
guinea pig.............................091 (H), 149 (B)
E lung dendritic cells ..............................214 (I)
gyrA...................................................044 (B)
lung .........................................................223
Ebola Virus ...........................029 (A), 146 (A)
H lymphocytic choriomeningitis virus ...144 (A)
Ebola .....................................098 (I), 207 (K)
hantavirus.................................................112 lyophilisation .....................................063 (D)
Edema Factor.....................................180 (D)
education .............................104 (K), 175 (D) hemorrhagic fever virus...............076 (F), 227
M
elderly ...............................................089 (H) hepatic disease..........................................113
macaque .....................................201 (K), 228
electrochemiluminescence........................111 high-throughput screen......................082 (G)
macrophages........................................212 (I)
Electrospray-Neutralization Method ..083 (G) HMGB-1.................................................225
Madagascar...............................................112
emerging pathogen.............................165 (B) horizontal gene transfer ......................056 (B)
MAPK and STAT3..............................096 (I)
endospore............................................216 (J) host responses .....................................188 (F)
humanization .....................................080 (G) Marburg Virus.............................030 (A), 227
enteritidis ...........................................157 (B)
master’s degree...................................103 (K)
environmental samples ........................071 (E)
I medicinal plant ....................................099 (J)
epidemiology study ............................060 (B)
identification......................................068 (D) melioidosis........057 (B), 108, 161 (B), 211 (I),
epidemiology .....................................032 (A)
.........................................................226, 232
IL-1beta and IL-6................................096 (I)
Escherichia coli O157:H7.........079 (F), 172 (B)
microbial infectious diseases ................079 (F)
immobilized ........................................101 (J)
evolution............................................033 (A)
iImmune response ........................094 (I), 221 microfluidics........................................216 (J)
export control ....................................218 (K)
immunity ...........................................207 (K) microRNA expression profiling................231

F immunoassay...............................182 (E), 111 microRNA ..............................................115

immunochromatographic test 184 (E), 186 (E) Mn-DP-Pi Complexes .......................199 (K)
F1 and LcrV antigens..........................166 (B)
Immunocompromised model.............090 (H) mobile genetic element ......................056 (B)
F1 and V Antiboides ...........................200 (K)
imvamune..........................................089 (H) modifiable risk factors ........................105 (K)
field and clinical applications ..............066 (D)
in vivo imaging ........................................117 monkeypox.................................201 (K), 228
filovirus .............098 (I), 142 (A), 146 (A), 231
inactivated..........................................206 (K) monoclonal antibodies ..........142 (A), 151 (B)
first responders ...................................104 (K)
indirect Yersinia pestis detection...........066 (D) morbidity...........................................105 (K)
forensic awareness ................................215 (J)
inflammation ......................................157 (B) mosquito............................................167 (B)
forensic entomology ............................215 (J)
influenza-A virus................................027 (A) MRSA ...............................................036 (B)
formaldehyde.......................................100 (J)
influenza ............................................032 (A) multiplex...............................073 (E), 184 (E)
fosmidomycin.....................................052 (B)
Francisella tularensis ................049 (B), 054 (B), inhibitors .................................................114 murine..................................045 (B), 161 (B)
...................................................167 (B), 225 insect..................................................148 (B) MVA..................................................205 (K)

98 10th ASM Biodefense and Emerging Diseases Research Meeting


KEYWORD I N DE X

Keyword........................................Presentation Number Keyword........................................Presentation Number Keyword........................................Presentation Number

MVLA................................................190 (F) personal safety.....................................077 (F) replicon platform ...............................208 (K)


MVLA-8............................................170 (B) phosphorodiamidate morpholion oligomers resistance .................044 (B), 052 (B), 068 (D)
195 (G).......................................................... respiratory viral pathogens ..................028 (A)
N plague vaccine ......................087 (H), 200 (K) reverse genetics ..................................144 (A)
nano-aerosol ......................................083 (G) plague ........061 (B), 070 (E), 092 (H), 097 (I), rhesus macaque ..................................193 (G)
........................117, 148 (B), 152 (B), 205 (K)
national security studies......................103 (K) ribonuclease mutants ..........................037 (B)
plant-produced vaccine ......................197 (K)
natural foci .........................................070 (E) ribosome assembly..............................037 (B)
neuroinvasion...........................................222 preparedness .......................................028 (A)
ribosome ...........................................086 (H)
neurovirulence .........................................222 priorization........................................219 (K)
ricin ...................................................181 (E)
neutrophil ...............................095 (I), 211 (I) protective antigen.........................210 (I), 234
Ricinus communis .................................183 (E)
new antibiotic ..........................................226 protective immunity ...........................169 (B)
Rickettsia ...........................................058 (B)
new....................................................145 (A) protein therapeutic.............................083 (G)
Rickettsiae.........................................093 (H)
next generation sequencing....189 (F), 191 (F) Pseudomallei ........................................154 (B)
Rift Valley Fever Virus, aerosol,
NF-kappB signaling cascade .....................114 Pseudomonas aeruginosa ...........053 (B), 054 (B)
enchephalitis ..........................................113
non-human primate model ................203 (K) pyrosequencing ..................................171 (B)
Rift Valley Fever Virus ..............................114
nonhuman primate .............088 (H), 092 (H), risk factors..........................................060 (B)
............................................196 (G), 209 (K) Q
rodent................................................196 (G)
nosocomial infection ..........................106 (K) Q fever ....................................................109
rOmpB..............................................093 (H)
qPCR .................................................213 (I)
O ROS....................................................212 (I)
quality assurance ................................175 (D)
rPA vaccine........................................197 (K)
on site collection and detection ..........072 (E) quasispecies ...........................033 (A), 076 (F)
One Health........................................107 (K) RV-PCR ............................................071 (E)
quinolone...........................................035 (B)
on-site detection ......................................110 quorum sensing..................................054 (B) S
ornithine lipids.........................................221
R salmonella .............................044 (B), 157 (B)
orthobunyavirus..................................078 (F)
sample preparation ................055 (B), 150 (B)
orthopox virus ...................................203 (K) rabbit genome array ............................188 (F)
sampling.............................................217 (K)
outbreaks ...........................................067 (D) rabbitpox ............................................188 (F)
scenario .............................................219 (K)
outer membrane protein...........................119 rabbits ................................................059 (B)
outer membrane vesicle............................233 Schistosom mansoni ................................094 (I)
rabies virus .........................................140 (A)
oxyR..................................................173 (B) school absenteeism .............................032 (A)
rapid detection ...................................185 (E)
rapid diagnostic platform....................065 (D)
P Topic Categories:
rare pathogens....................................176 (D) Below is a list of descriptions of topic cate-
Pakistan..............................................141 (A) gories (letters in parentheses by each pres-
rare variant .........................................189 (F) entation number refer to this chart).
pandemic ...........................................028 (A)
reaerosolization...................................162 (B)
parvovirus............................................100 (J) A. Viral Agents
reagent...............................................064 (D) B. Bacterial Agents
passive transfer ...................................091 (H) C. Fungal Agents
real-time PCR........055 (B), 141 (A), 181 (E),
pathogen detection ............................069 (D) D. Diagnostics
..........................................................185 (E) E. Environmental Detection
pathogen ............................................218 (K) reassortment .......................................078 (F) F. Informatics and Genomics
pathology ............................................094 (I) G. Therapeutics
receptor cluster formation ........................224 H. Vaccines
PCR platform comparison.................062 (D) regulation .............................145 (A), 218 (K) I. Immune Responses
peptide mapping..................................210 (I) J. Decontamination, Biosafety,
regulatory compliance, safety and and Containment
periplasm............................................041 (B) maintenance.....................................220 (K) K. Other and Information Only
persistence..........................................043 (B) respiratory viruses...............................027 (A)

BIODEFENSE PROGRAM & ABSTRACTS 99


KEYWORD I NDE X

Keyword........................................Presentation Number Keyword........................................Presentation Number

second generation sequencing ............069 (D) Tularemia............................................088 (H)


secondary cell wall polysaccharide ......153 (B) Tularensis ............................................061 (B)
select agent...........................145 (A), 175 (D) tungiasis .............................................105 (K)
sGP .....................................................098 (I) type III secretion041 (B), 118, 163 (B), 169
shRNA..............................................194 (G) (B) .................................................................

single domain antibodies ....................179 (D) typing.................................................170 (B)

single nucleotide repeats .....................171 (B)


U
siRNA ...............................................143 (A)
ultra-rare variant detection..................191 (F)
site specific recombination..................035 (B)
S-layer...................................153 (B), 155 (B) V
small mammals .........................................112
vaccine efficacy......................085 (H), 097 (I)
smallpox vaccine ....................090 (H), 213 (I)
vaccine...........092 (H), 093 (H), 119, 201 (K),
smallpox.............................................196 (G) .................202 (K), 204 (K), 205 (K), 206 (K),
SNP, MLVA........................................168 (B) ...................................................207 (K), 233
sortase ......................................................234 vaccinia neutralization assay .................213 (I)
special unit .........................................072 (E) vaccinia virus .....................................203 (K)
spores ..................................................102 (J) vaccinia-experienced .........................089 (H)
stabilization ........................................202 (K) validated PCR system.........................075 (E)
Staphylocidal ......................................036 (B) vector....................................148 (B), 152 (B)
Staphylococcus aureus ...............053 (B), 081 (G) VEEV ......................................................222
Staphyloxanthin..................................053 (B) Venezuelan Equine Encephalitis virus
........................082 (G), 115, 199 (K), 206 (K)
super resolution microscopy .....................224
vibrio cholerae ...................................056 (B)
surface chemistry ................................186 (E)
ViPR..................................................187 (F)
surveillance.........................................070 (E)
virulence analysis................................042 (B)
Swiss Webster mice ............................085 (H)
virulence ............................................173 (B)
synergistic ..........................................036 (B)
virus pathogen resource ......................187 (F)
synthetic biology (SB).........................077 (F)

W
T
water quality sensors...........................164 (B)
temperature regulation .............................116
weapons mass destruction...................217 (K)
therapeutic.........................................192 (G)
World Health Organization ................074 (E)
therapeutics ........................................156 (B)
third world.........................................107 (K) X
Thymus munbyanus ..............................106 (K)
XF Drugs ...........................................158 (B)
tick born diseases ...............................178 (D)
ticks ...................................................058 (B) Y
toll-like receptor ......................................224
Yersinia pestis, Clostridium botulinum ......185 (E)
Toxoplasma gondii................................086 (H)
Yersinia pestis,Y. pseudotuberculosis ..........050 (B)
training courses ..................................217 (K)
Yersinia pestis...........085 (H), 079 (F), 087 (H),
transformation....................................058 (B) ..............................117, 118, 166 (B), 209 (K)
transitional operations ........................220 (K) Yersinia .....................041 (B), 047 (B), 169 (B)
transmission........................................167 (B)

100 10th ASM Biodefense and Emerging Diseases Research Meeting


AUTHOR INDEX

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Aaron, J. S. ...............................................224 Axthelm, M. .....................................088 (H) Blaney, J. ...........................................207 (K)


Abazashvili, N. ...................................046 (B) Baccetty, A. .......................................062 (D) Blaylock, B. .........................041 (B), 163 (B)
Abbott, E. .........................................202 (K) Backman, S. ......................................167 (B) Blazevic, E. ........................................073 (E)
Abdukhalilova, G. ..............................062 (D) Bae, E. ..............................................068 (D) Bloomfield, H. .....................029 (A), 030 (A)
Achazi, K. .........................................065 (D) Baer, A. ....................................................114 Bloomfield, H. A. ..............................146 (A)
Adamia, R. ........................................160 (B) Bagarozzi, D. .....................................180 (D) Boboye, B. E. ......................................099 (J)
Adams, C. .........................................165 (B) Bailey, C. .................................................114 Bonvini, E. ........................................196 (G)
Agans, K. ...................................201 (K), 228 Bailey, T. .....................................201 (K), 228 Booth, J. .............................................214 (I)
Agrawal, S. .........................................079 (F) Baillie, L. ............................................095 (I) Borel, A. ............................................204 (K)
Ågren, P. ............................................055 (B) Bakanidze, L. .....................................047 (B) Borja, N. A. ........................................060 (B)
Alavi, M. R. ......................................053 (B) Bakanidze, L. G. ................................070 (E) Borucki, M. ......................................033 (A)
Albornoz, E. ......................................035 (B) Baker, R. ..............................029 (A), 030 (A) Bosio, C. .................................................253
Alfaro, T. ............................................071 (E) Baker, R. .................................................228 Bowe, M. ..........................................196 (G)
Allen, J. .............................................033 (A) Bakken, R. ........................................206 (K) Bowen, L. E. ......................................200 (K)
Almuzara, M. .....................................165 (B) Bakken, R. R. ...................................208 (K) Bowen, R. ...............................................221
Altan-Bonnet, N. ....................................243 Balabaev, N. K. ..................................083 (G) Bower, K. ..........................................043 (B)
Alterson, K. .......................................208 (K) Bales, J. M. ..............................................113 Boydston, J. A. ...................................200 (K)
Aly, K. ...............................................163 (B) Balinandi, S. ......................................107 (K) Boyer, A. ...........................................180 (D)
Amieva, M. R. .........................................121 Baron, C.. ................................................137 Boyer, A. E. .........................059 (B), 067 (D)
Amoako, K. .......................................171 (B) Barr, J. R. ...............059 (B), 067 (D), 180 (D) Boylan, J. ...........................................173 (B)
Amosun, A. E. ....................................172 (B) Barry, E. ............................................049 (B) Boylan, R. J. .....................................104 (K)
Anderson, A. D. .......................................109 Bate, S. R. ..........................................045 (B) Braciale, T. J. ............................................252
Anderson, B. ......................................028 (A) Bates, S. .............................................161 (B) Brand, S. ..................................................229
Anderson, G. .....................................179 (D) Bavari, S. .................................................227 Branda, S. ..........................................069 (D)
Anderson, M. ....................................193 (G) Bearden, S. W. ....................................148 (B) Brasel, T. ....................................201 (K), 228
Andres, P. ...........................................035 (B) Beckstrom-Sternberg, J. .....................168 (B) Braun, A. ............................................214 (I)
Andrews, C. ......................................202 (K) Beckstrom-Sternberg, S. ....................168 (B) Brett, P. J. .................................................232
Andrianaivo, T. ........................................112 Bedi, K. .............................................180 (D) Brewoo, J. .........................................205 (K)
Ansari, U. ..........................................180 (D) Beeri, K. ............................................051 (B) Brezillon, C. ......................................042 (B)
Antonic,V. .........................................053 (B) Beitzel, B. ..................................082 (G), 227
Aranio, D. .........................................067 (D) Bell, T. ..............................................032 (A) Topic Categories:
Below is a list of descriptions of topic cate-
Arcidiacono, S. ...................................101 (J) Belle, A. ............................................059 (B) gories (letters in parentheses by each pres-
entation number refer to this chart).
Arterburn, J. ......................................082 (G) Bengtsson, U. A. ..................................100 (J)
Arvans, D. .........................................194 (G) Bent, Z. ...........................................069 (D) A. Viral Agents
B. Bacterial Agents
Astwood, M. ......................................183 (E) Berthelot, T. ..........................184 (E), 186 (E) C. Fungal Agents
Atimnedi, P. .......................................107 (K) Bethel, L. M. ...........................................113 D. Diagnostics
E. Environmental Detection
Atkins, T. P. ........................................154 (B) Bewley, K. R. ....................................149 (B) F. Informatics and Genomics
Atkinson, B. ......................................141 (A) Bhomia, M. .............................................222 G. Therapeutics
H. Vaccines
Atkinson, D. J. ....................................149 (B) Birdsell, D. .........................................168 (B) I. Immune Responses
AuCoin, D. ........................................156 (B) Birkenholz, J. .....................................150 (B) J. Decontamination, Biosafety,
and Containment
AuCoin, D. P. ..........................................232 Bjork, A. ..................................................109 K. Other and Information Only
Avaliani, N. ........................................046 (B) Blaney, D. ..........................................067 (D)

BIODEFENSE PROGRAM & ABSTRACTS 101


AUTHOR I N DE X

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Briscoe, C. ........................................193 (G) Chen, Z. ...........................................180 (D) Das, R. ....................................................114


Brittingham, K. T. ..............................057 (B) Cheng, B.Y. H. ..................................144 (A) DaSilva, L. .........................................188 (F)
Brogden, K. A. .........................................135 Cheng, L. W. ............................................111 Daugherty, S. ......................................079 (F)
Broladze, M. ......................................046 (B) Cheong, H. .........................................216 (J) Dauphin, L. A. ...................................183 (E)
Brooks, K. ..........................................097 (I) Cherwonogrodzky, J. .........................080 (G) de la Garza, M. ..................................059 (B)
Brown, P. B. ......................................217 (K) Chichester, J. .....................................197 (K) de la Torre, J. C. .................................144 (A)
Bruce, C. ............................141 (A), 142 (A) Chinglai,Y. .........................................098 (I) Dean, R. E. ........................................154 (B)
Brunelle, S. ........................................075 (E) Chiu, C. ..................................................008 Dean, S. N. .......................................054 (B)
Buller, M. ..........................................196 (G) Cho, M.-H. .......................................096 (I) Dearen, K. .................................201 (K), 228
Burger, R. F. ............................................025 Cho, M. .............................................151 (B) Dekonenko, A. ..................................196 (G)
Burke, S. ...........................................196 (G) Choi, S.Y. ..........................................056 (B) Delarosa, P. ........................................107 (K)
Burke, W. ...........................................150 (B) Christman, J. W. ..................................212 (I) Delp, K. .............................................043 (B)
Burkhardt, N.Y. .................................058 (B) Chun, J. .............................................056 (B) Dembele, M. ....................................178 (D)
Burtnick, M. N. .......................................232 Chun, J.-H. ...........................096 (I), 151 (B) Demeure, C. E. ........................................237
Busuulwa, M. ....................................107 (K) Chung, D. ........................................082 (G) Dergel, I. ............................................100 (J)
Byfors, S. ..........................................055 (B) Ciccarone,V. ......................................196 (G) Dersch, P. ................................................019
Callahan, J. ........................................062 (D) Ciletti, N. ..........................................169 (B) DeShazer, D. ............................................013
Cann, J. .............................................207 (K) Ciletti, N. A. .....................................087 (H) Dieringer-Boyer, S. ............................043 (B)
Cardwell, M. M. ................................093 (H) Clark, G. C. .............................................225 Dietrich, J. .........................................187 (F)
Carlos, M. P. ......................................028 (A) Clemens, K. M. ..................................075 (E) Disqué, C. .........................................176 (D)
Carlos, M. P. ......................................178 (D) Clement, K. H. .................................064 (D) Doherty, L. .........................................101 (J)
Carrion, R. .......................................059 (B) Clohessey, A. ......................................102 (J) Dong, L. ..................................................227
Carson, B. D. ...........................................224 Coffin, J. ............................................158 (B) Dorsey, B. ..........................................043 (B)
Carson, H. .........................................145 (A) Coggeshall, K. .....................................214 (I) Douady, J. ................................................223
Carter, A. .................................................225 Cohen, J. ...........................................206 (K) Dowall, S. ..........................................141 (A)
Cebula, T. A. ......................................056 (B) Colman, R. E. ...................................189 (F) Drew, J. ............................................202 (K)
Centrón, D. .......................................165 (B) Colwell, R. R. ......................056 (B), 185 (E) Drexler, J. ..........................................065 (D)
Chamberlain, J. ..................................141 (A) Compans, R. ......................................098 (I) Duggan, E. ..........................................214 (I)
Champney, W. S. ................................037 (B) Connor, M. G. .........................................117 Duggan, M. .......................................150 (B)
Chan, W. .............................................216 (J) Cook, J. L. ..........................................212 (I) Dupke, S. ..........................................042 (B)
Chance, T. B. .....................................146 (A) Cook, N. ...........................................141 (A) Duprex, P. ...............................................245
Chantratita, N. .........................................232 Corso, A. ...........................................035 (B) Dworkin, J. ........................................153 (B)
Chanturia, G. ......................047 (B), 159 (B) Credou, J. ............................184 (E), 186 (E) Dyer, D. N. ........................................034 (B)
Chanturia, G. .....................................168 (B) Cronin, L. X. .....................................067 (D) Easterbrook, L. ..................................142 (A)
Chaplin, P. ........................................089 (H) Cross, A. .............................................095 (I) Eberhard, M. .....................................065 (D)
Chapman, J. .......................................150 (B) Crosson, S. ........................................194 (G) Eckstein, T. M. ............................174 (B), 221
Chappie, D. J. .....................................162 (B) Cummings, P. J. .................................103 (K) Eduok, N. N. ....................................147 (B)
Charlwood, M. ..................................149 (B) Currie, B. J. .............................................011 Egan, C. ............................................040 (B)
Chau, D. ...........................................080 (G) Curtis, D. ..........................................069 (D) Ehrs, S. ..............................................055 (B)
Chen, A. ............................................185 (E) Custer, M. .........................................208 (K) EL-BARRAWY, M. .........................027 (A)
Chen,Y.-L. ..............................................108 D’Elia, R.V. .............................................225 Eleazar, C. I. ......................................147 (B)
Chen,Y.-S. ..............................................108 Daly, M. J. ..........................................199 (K) Ellerbrok, H. ....................................203 (K)

102 10th ASM Biodefense and Emerging Diseases Research Meeting


AUTHOR INDEX

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Elli, D. .................................087 (H), 169 (B) Galindo, C. L. .....................................188 (F) Sites, R. N. ....................................089 (H)
Elssner, T. ................................................110 Gallegos, K. ..........................092 (H), 097 (I) Gros, P. ....................................................236
Emmoth, E. .......................................100 (J) Gallegos, M. ......................................180 (D) Grunow, R. .......................................042 (B)
Engelthaler, D. M. ..............................189 (F) Gallegos-Candela, M. ...........059 (B), 067 (D) Gu, Z. ................................................187 (F)
Ennis, J. .............................................208 (K) Galyov, E. E. ......................................154 (B) Gubeladze, L. .....................................160 (B)
Eppinger, M. .....................................079 (F) Gandhi, P. ..........................................182 (E) Guendel, I. .......................................114, 115
Epstein, G. L. ...................................134, 251 Garcia, A. D. .......................................213 (I) Gunn, J. ...................................................017
Epstein, J. H. ............................................010 Garcia de la Guardia, R. .....................060 (B) Gupta, P. .......................199 (K), 206 (K), 222
Ernst, R. K. .............................................116 Garib,V. ............................................062 (D) Hadfield, T. ........................................075 (E)
Facemire, P. .......................................038 (B) Garrett, J. ..........................................145 (A) Hahn, K. R. ......................................171 (B)
Fair, J. ......................................................007 Garufi, G. ..................................051 (B), 234 Hakim, S. T. .......................................077 (F)
Farb, D. .............................................196 (G) Garvey, E. ................................................229 Haley, B. J. .........................................185 (E)
Fatima zohra, M. ..............................106 (K) Garza, N. .................................................227 Hallis, B. ............................................198 (K)
Faucher, S. P. ............................................018 Gauthier,Y. .......................................204 (K) Han, J. .....................................................222
Felder, E. ..............................063 (D), 181 (E) Gayen, M. ........................................199 (K) Hardt, W.-D. ......................................157 (B)
Fellows, P. ...........................085 (H), 209 (K) Gelhaus, H. C. ...................................057 (B) Harman-Richardson, N. ....................200 (K)
Felsheim, R. F. ...................................058 (B) Geller, B. ...........................................195 (G) Harris-Chen, H. ................................033 (A)
Feodorova,V. A. .................................166 (B) Gene Olinger, G. ...............................082 (G) Hartman, A. L. .........................................113
Filippov, A. A. ......................050 (B), 066 (D) Gerba, C. ................................................124 Hasan, N. A. ......................................056 (B)
Fiole, D. ...................................................223 Gherardini, F. .....................................173 (B) Hasan, Z. ...........................................141 (A)
Firmani, M. A. ...................................043 (B) Gillece, J. ............................................189 (F) Hashizume, S. ...................................090 (H)
Fisher, I. ................................092 (H), 097 (I) Gingras, B. .........................................061 (B) Hatch, G. .............................149 (B), 161 (B)
Fitzpatrick, K. A. ......................................109 Giordano, A. ......................................068 (D) Hatch, G. J. .......................................045 (B)
Fofanov,V. ..........................................191 (F) Giron, J. A. ..............................................122 Hauck, L. ................................................227
Foote, S. J. ...............................................235 Given, K. .................................................118 Hayter, I. ...........................................158 (B)
Foster, J. .............................................189 (F) Glass, P. ............................................206 (K) He,Y. .................................................050 (B)
Francesconi, S. ...................................047 (B) Glass, P. J. ............................199 (K), 208 (K) Healey, B. ..........................................028 (A)
Francesconi, S. ..................................062 (D) Glotzer, D. L. .....................................104 (K) Heaton, B. .........................................194 (G)
Francesconi, S. ...................................159 (B) Glyakina, A.V. ....................................083 (G) Heckert, R. .......................................107 (K)
Franchini, G. .....................................090 (H) Goji, N. .............................................171 (B)
Franz, T. .............................................042 (B) Goldman, E. ......................................179 (D) Topic Categories:
Below is a list of descriptions of topic cate-
Fraser-Liggett, C. ...............................079 (F) Goldman, S. ............................................226 gories (letters in parentheses by each pres-
entation number refer to this chart).
Frazier, A. D. .....................................037 (B) Goldstein, J. .......................................180 (D)
Frederick, S. .........................029 (A), 030 (A) Gonzalez-Juarrero, M. .............................221 A. Viral Agents
B. Bacterial Agents
Freiberg, A. ........................................207 (K) Goodlett, D. R. ........................................116 C. Fungal Agents
Fretwell, J. ............................045 (B), 161 (B) Goodman, S. M. ......................................112 D. Diagnostics
E. Environmental Detection
Funatsu, A. ........................................090 (H) Goossens, P. L. ...................................042 (B) F. Informatics and Genomics
Funnell, S. G. P. .....045 (B), 149 (B), 161 (B) Gordon, J. .........................................182 (E) G. Therapeutics
H. Vaccines
Gabay, J. E. ........................................194 (G) Gordon, M. .............................................123 I. Immune Responses
Gage, K. L. ........................................148 (B) Gordon, S. ........................................090 (H) J. Decontamination, Biosafety,
and Containment
Gaidamakova, E. K. ...........................199 (K) Gray, J. ..............................................175 (D) K. Other and Information Only
Galens, K. ..........................................079 (F) Greenberg for the POX-MVA-024

BIODEFENSE PROGRAM & ABSTRACTS 103


AUTHOR I N DE X

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Heffron, F. .........................................088 (H) Inamine, J. M. ...................................174 (B) Klint, M. ...........................................055 (B)


Heim, J. ...................................................226 Isbell, K. ..............................067 (D), 180 (D) Knight, I. ...........................................185 (E)
Hendrickx, A. ....................................051 (B) Iversen, P. ................................................227 Koblentz, G. ............................................133
Hendrickx, A. P. .................................155 (B) Iversen, P. L. .....................................195 (G) Koenig, S. ..........................................196 (G)
Hendrickx, A. P. A. ............................081 (G) Izadjoo, M. ........................................053 (B) Koide, F. ..............................196 (G), 200 (K)
Henning, L. ......................................193 (G) Jahrling, P. .........................................207 (K) Kondas, A.V. ............................................109
Heraud, J.-M. ..........................................112 Jamil, B. .............................................141 (A) Kopaskie, K. .....................................163 (B)
Hermanas, T. M. ..................087 (H), 152 (B) Jang, J. ...............................................151 (B) Koroleva, G. ......................................148 (B)
Herron, M. J. .....................................058 (B) Janzen, T. ...........................................171 (B) Koroleva, G. I. ....................................078 (F)
Herzog, C. .........................................072 (E) Jefferson, C. .............................................250 Koshinsky, H. ....................................191 (F)
Hewitt, J. .................................................003 Johnson, S. ........................................196 (G) Kozel, T. ............................................156 (B)
Hewson, R. ........................141 (A), 142 (A) Jones, E. ..................................................004 Kozel, T. R. .............................................232
Heymann, D. L. .......................................021 Jones, M. ...........................................197 (K) Kramski, M. ......................................203 (K)
Hill, L. .....................................................114 Jonsson, C. B. ....................................082 (G) Krile, R. ............................................209 (K)
Hillman, R. .......................................093 (H) Judy, B. ....................................................233 Kugelman, J. ............................................227
Hoekstra, W. ............................................229 Kaboyo, R. W. ....................................107 (K) Kugelman, J. R. .....................078 (F), 148 (B)
Hoffmaster, A. R. ..............................067 (D) Kadan, M. .........................................196 (G) Kuiken, C. ........................................076 (F)
Holbrook, M. ....................................207 (K) Kamrud, K. I. ....................................208 (K) Kuklenyik, Z. ....................................180 (D)
Holland, L. ........................................061 (B) Kane, J. A. ..........................................161 (B) Kumar, S. ...........................................187 (F)
Holland, L. ........................................085 (H) Kane, S. .............................................071 (E) Kurtti, T. J. .........................................058 (B)
Hong, K.-J. .........................................096 (I) Kanehara, T. ......................................090 (H) Kutateladze, M. ..................................160 (B)
Hong, K. ...........................................151 (B) Kanev, I. L. ........................................083 (G) Kvachadze, L. ....................................160 (B)
Honnold, S. .......................................206 (K) Karaolis, D. K. R. ..............................043 (B) Lander, A. ..........................................042 (B)
Hoq, M. ............................................056 (B) Kashanchi, F. ....................................114, 115 Lane, P. ..............................................069 (D)
Hoque, S. ............................................102 (J) Kaup, F.-J. ..........................................203 (K) Lane, T. .............................................069 (D)
Houghton, R. .........................................232 Kehn-Hall, K. ..................................114, 115 Langer, M. .........................................214 (I)
Hu, S.-C. ...........................................061 (B) Keim, P. .........................168 (B), 189 (F), 242 Langevin, S. .......................................069 (D)
Hu, W. ..............................................080 (G) Keim, P. S. ..........................................057 (B) Larson, C. ..........................................187 (F)
Huang, L. ..........................................196 (G) Kekeklidze, M. ...................................047 (B) Latham, J. ...........................................190 (F)
Huang, X.-Z. ...................................170 (B) Kekelidze, M. ....................................159 (B) Lathem, W. W. ..........................................015
Hubbard, M. A. .................................156 (B) Kekelidze, M. ....................................168 (B) Lavander, M. ......................................055 (B)
Huber, N. T. ......................................064 (D) Kelly-Cirino, C. .................................040 (B) Lawrenz, M. B. ........................................117
Hudson, L. ........................................142 (A) Kern, J. ..............................................051 (B) Laws, T. R. ...............................................225
Hultén, C. ...........................................100 (J) Kersh, G. J. ..............................................109 Lawson, T. .........................................178 (D)
Hunt,V. ..............................................187 (F) Khizhnyakova, M. A. ..........................166 (B) Lazar Adler, N. R. .............................154 (B)
Huntley, J. ...............................................119 Khmaladze, E. ....................................159 (B) LeClaire, R. ..........................092 (H), 097 (I)
Huq, A. ................................056 (B), 185 (E) Khmaladze, E. ...................................168 (B) Ledford, J. G. ...........................................255
Hutchins, A. S. ..................................183 (E) Khodiev, A. .......................................062 (D) Lee, M.-H. ........................................072 (E)
Ibadova, G. ........................................062 (D) Kidney, A. ..........................................040 (B) Lee, P. ...............................................179 (D)
Imnadze, P. ...........................046 (B), 159 (B) King, C. A. ..............................................115 Leffel, E. ..........................................006, 136
Imnadze, P. ........................................168 (B) Klee, S. R. .........................................042 (B) Legasse, A. .........................................088 (H)
Imnadze, P. G. ....................................070 (E) Klem, E. ............................................187 (F) Lehman, M. ......................................067 (D)

104 10th ASM Biodefense and Emerging Diseases Research Meeting


AUTHOR INDEX

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Lehman, S. .........................................043 (B) Mackie, R. S. ....................................052 (B) Mitchell, A. .............................................118


Lemoine, S. ...........................092 (H), 097 (I) Maheshwari, R. .................................206 (K) Moatter, T. .........................................141 (A)
Leppla, S. H. ......................................180 (D) Maheshwari, R. K. .....................199 (K), 222 Mohan, G. S. ......................................098 (I)
Letant, S. ............................................071 (E) Maier, H. .................................................234 Mollik, M. ........................................074 (E)
Levi, A. .............................................194 (G) Malania, L. .............046 (B), 159 (B), 168 (B) Mollura, D. J. ...........................................247
Levitan, I. ...........................................212 (I) Mamisashvili1, E. ...............................048 (B) Monier, A. ............................092 (H), 097 (I)
Levy, S. ....................................................248 Manceva, S. .......................................197 (K) Montague, J. ......................................217 (K)
Leysath, C. E. ....................................180 (D) Maniatis, P. ........................................067 (D) Montgomery, S. .................................178 (D)
Li, H. ................................................067 (D) Mark, D. ...........................................065 (D) Moore, W. ................................................229
Li, H. ................................................196 (G) Marohn, M. ......................................049 (B) Morici, L. A. ............................................233
Li, L. ...................................................216 (J) Maroo, A. ...........................................079 (F) Morozov,V. N. ..................................083 (G)
Li,Y. ........................................................116 Marshall, S. .......................................085 (H) Mossbrugger, I. ..................................181 (E)
Lin, H.-S. ................................................108 Marston, C. .......................................067 (D) Motin,V. L. ........................................166 (B)
Lin, S. ................................................200 (K) Martin, S. .........................................091 (H) Motlagh, H. ......................................068 (D)
Lin, W. ..............................................085 (H) Martinez, J. .......................................194 (G) Mourich, D. ............................................227
Lin, W. C. ..........................................061 (B) Martinez, J. J. ....................................093 (H) Mourich, D.V. ...................................195 (G)
Lind, C. .............................................206 (K) Martinez-Sobrido, L. .........................144 (A) Mukhopadhyay, K. .............................036 (B)
Lindström, A. .....................................215 (J) Maruno, S. ........................................090 (H) Mulenga, A. .............................................130
Linke, S. ............................................065 (D) Mathieu, J. .........................................204 (K) Muller, W. ...........................................101 (J)
Linow, M. .........................................176 (D) Mathieu, J. D. ..........................................223 Munderloh, U. G. ..............................058 (B)
Lins, R. C. ..........................067 (D), 180 (D) Matsui, H. .........................................090 (H) Murphy, G. ........................................071 (E)
Liu, D. ...............................................175 (D) Mätz-Rensing, K. ..............................203 (K) Murphy, N. ......................................176 (D)
Liu, G. .....................................................226 McCool, J. .........................................163 (B) Murtaugh, W. A. ................................028 (A)
Liu, J. ................................................179 (D) McCormick, K. D. ............................031 (A) Musiychuk, K. ...................................197 (K)
Liu, J. .................................................191 (F) McGuinness, E. .................................193 (G) Mwangi, W. .............................................221
Liu, M. ..............................................187 (F) McIver, L. ..........................................188 (F) N. Ivanov, O. Deryabin,V. Skripnik, ....140 (A)
Liu, P.-J. ..................................................108 McKenney, E. S. .................................052 (B) Na, M. P. M. N. ..................................094 (I)

Llewellyn, Z. N. ................................200 (K) McLuckie, G. .....................................149 (B) Nadeem, S. G. ....................................077 (F)

Lockman, H. .....................................209 (K) McMahon, K. ....................................188 (F) Nakao, B. ...........................................073 (E)

Lofts, L. L. .........................................078 (F) Mellmann, A. ..........................................026


Mello, C. ............................................101 (J) Topic Categories:
Logue, C. ..........................................141 (A) Below is a list of descriptions of topic cate-
Lohman, K. L. ....................................158 (B) Meseda, C. A. ......................................213 (I) gories (letters in parentheses by each pres-
entation number refer to this chart).
Lordo, R. A. .......................................162 (B) Metcalf, J. ...........................................214 (I)
Metcalfe, K. ......................................091 (H) A. Viral Agents
Love, H. ............................................142 (A) B. Bacterial Agents
Love, W. G. .......................................158 (B) Migliozzi, R. H. ................................064 (D) C. Fungal Agents
Mikeladze, E. .....................................047 (B) D. Diagnostics
Lukaszewsk, R. A. ....................................225 E. Environmental Detection
Lunderberg, J. M. ..............................153 (B) Miller, N. ................................................118 F. Informatics and Genomics
Miller, N. C. .....................................169 (B) G. Therapeutics
Lundin Zumpe, A. .............................055 (B) H. Vaccines
Lyapina, A. M. ....................................166 (B) Miller, S. ...........................................193 (G) I. Immune Responses
Milligan, D. .............................................226 J. Decontamination, Biosafety,
Lyons, A. ...........................................180 (D) and Containment
M. Nikolaishvili, M. Zakareishvili, Missiakas, D. ....039 (B), 051 (B), 081 (G), 234 K. Other and Information Only
I. Beradz, e. ......................................048 (B) Missiakas, D. M. .................................153 (B)

BIODEFENSE PROGRAM & ABSTRACTS 105


AUTHOR I N DE X

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Nalca, A. ..029 (A), 030 (A), 146 (A), 188 (F) Oyston, P. ................................................139 Powell, D. ................................................113
Narayanan, A. ..........................................114 Pak, D. ..............................................193 (G) Powell, D. A. ............................................116
Natidze, M. .......................................160 (B) Pal, U. .....................................................128 Powell, T. ...........................................205 (K)
Ndumu, D. B. ...................................107 (K) Palacios, G. ..............................................227 Power, A. .................................................231
Negrych, L. .......................................080 (G) Palacios, G. F. ......................................078 (F) Prenzler, F. ..........................................214 (I)
Newman, K. A. .................................064 (D) Palacios, G. F. .....................................148 (B) Price, J. ..............................085 (H), 209 (K)
Ngomi, N. N. N. N. ..........................105 (K) Pandrea, I. ...............................................113 Priestley, R. A. .........................................109
Nguyen-Mau, S.-M. ............039 (B), 153 (B) Papaneri, A. .......................................207 (K) Prior, J. L. ..........................................154 (B)
Nichols, D. K. ....................................146 (A) Park, C. ...................................................132 Probst, L. ..........................................073 (E)
Nichols, T. L. .....................................162 (B) Park, J. ...............................................151 (B) Puri, R. K. ..............................................222
Niedrig, M. .......................................065 (D) Parrish, T. ..........................................150 (B) Quenee, L. ........................................093 (H)
Niemuth, N. .....................................091 (H) Partidos, C. ........................................205 (K) Quenee, L. ........................................169 (B)
Nieves, W. ................................................233 Patel, P. ............................................065 (D) Quenee, L. E. ....................................152 (B)
Nikolich, M. ......................................168 (B) Patel,V. ...............................................214 (I) Quenee, L. E. F. ................................087 (H)
Nikolich, M. P. .....................050 (B), 066 (D) Pati, N. B. ..........................................157 (B) Quesnel-Hellmann, A. .............................223
Nikolich, M. P. ..................................170 (B) Pauli, G. ............................................203 (K) Quinn, C. ...........................................210 (I)
Nilsson, C. .........................................055 (B) Peacock, S. J. ............................................232 Quinn, C. P. ........................067 (D), 180 (D)
Noh, S. M. ..............................................129 Pearson, T. .........................................168 (B) Raetz, C. R. H. .......................................116
Nordstrom, J. L. ................................196 (G) Peck, M. ............................................040 (B) Rainey, G. J. ......................................196 (G)
Noronha, J. ........................................187 (F) Pennington, J. ..........................................117 Rajagopal, S. ....................................086 (H)
Northurp, M. A. .................................073 (E) Pepler, A. ...........................................190 (F) Ramachandran, G. .............................095 (I)
Nozadze1, M. ....................................047 (B) Pepper, I. ...........................................164 (B) Ramirez, M. ......................................165 (B)
Nunneley, J. .......................................059 (B) Pereyra, F. ................................................238 Ramishvili, M. ...................................046 (B)
Nyakiti, D. .........................................038 (B) Periaswamy, B. ...................................157 (B) Rasko, D. A. .....................................116, 239
Obiso, R. ..........................................062 (D) Perry, M. ..........................................040 (B) Ravel, J. .............................................079 (F)
Obiso, R. J. ............046 (B), 047 (B), 048 (B), Pesik, N. ............................................067 (D) Rawat, A. ...........................................189 (F)
.............................................159 (B), 160 (B) Peters, H. ...........................................028 (A) Rawlinson, W. ...................................175 (D)
Obom, K. M. ....................................103 (K) Petroni, A. .........................................035 (B) Raychaudhuri, S. .....................................232
Offit, P. A. ................................................020 Phillippy, A. .......................................185 (E) Razafindralambo, N. ................................112
Ogeneh, B. O. ....................................147 (B) Pickett, B. ..........................................187 (F) Reed, D. ..................................................113
Oh, S.-Y. ....................................155 (B), 234 Pitman, J. K. ......................................149 (B) Reed, D. ..................................................232
Ohkuma, K. ......................................090 (H) Pitt, L. .......................................001, 038 (B) Revelli, D. A. ....................................173 (B)
Olatoye, O. .......................................172 (B) Pitt, M. L. M. .....................................034 (B) Reyes-Lamothe, R. ...........................035 (B)
Olive, M.-M. ...........................................112 Pitts, R. .............................................180 (D) Reynes, J.-M. ..........................................112
Oliver, J. D. .......................................058 (B) Plank, J. .............................................142 (A) Rhee, A. ............................................197 (K)
Omoya, F. O. ......................................099 (J) Plotkin, J. B. ............................................241 Rhie, G.-E. ...........................096 (I), 151 (B)
Onashvili, T. .....................................048 (B) Pohl, J. ................................................210 (I) Rhys-Williams, W. .............................158 (B)
Ongarbaev, A. ...................................062 (D) Pöhlmann, C. ..........................................110 Richards, K. ......................................142 (A)
Ortiz Riano, E. .................................144 (A) Polupan, I. ........................................140 (A) Richardson, M. .................................032 (A)
Osorio, J. ...........................................205 (K) Polyanina, T. I. ...................................166 (B) Richter, G. S. .....................................153 (B)
Ota, I. M. ...........................................075 (E) Porter, A. ............................................188 (F) Richter, M. H. ..................................072 (E)
Overheim, K. A. ..................092 (H), 097 (I) Potluri, L. P. .......................................194 (G) Richter, S. ........................................081 (G)

106 10th ASM Biodefense and Emerging Diseases Research Meeting


AUTHOR INDEX

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Richter, S. G. .....................................051 (B) ...........153 (B), 155 (B), 163 (B), 169 (B), 234 Silvera, P. M. .....................................200 (K)
Rider, T. H. ......................................192 (G) Schneider, C. ........................092 (H), 097 (I) Silvestri, E. .........................................162 (B)
Rigvava, S. ........................................160 (B) Schnell, F. ..........................................195 (G) Simpson, A. J. ....................................198 (K)
Riley, S. .............................................194 (G) Schnell, M. ........................................207 (K) Singh, M. ..........................................036 (B)
Riley, S. P. .........................................093 (H) Schotzinger, R. ........................................229 Smagin, A. .........................................062 (D)
Robers, A. .........................................142 (A) Schriewer, J. ......................................196 (G) Smith, J. ............................................082 (G)
Roberts, A. ...........................149 (B), 161 (B) Schupp, J. ...........................................189 (F) Smith, Q. B. .............................................224
Roberts, A. D. G. ...............................045 (B) Schweizer, H. P. .......................................014 Smith, T. L. ........................................067 (D)
Roberts, R. .............................................002 Scully, C. ...........................................082 (G) Soarimalala,V. L. ......................................112
Robins, P. ..........................................194 (G) Self, J. S. ..................................................109 Soler-Bistué, A. ..................................035 (B)
Robinson, J. ......................................068 (D) Semenova,V. ......................................067 (D) Solomonia, R. ..................................047 (B)
Roche, A. J. .............................................109 Semenova,V. A. ..................................210 (I) Sorenson, J. ........................................217 (K)
Roffey, P. ...........................................175 (D) Senina, S. .................................................114 Soroka, S. ............................................210 (I)
Rom, A. ............................................085 (H) Sergueev, K.V. ......................050 (B), 066 (D) Spaulding, U. K. ................................075 (E)
Romero, F. ........................................092 (H) Seshu, J. ...................................................131 Sperandio,V. ............................................016
Rosati Rowe, J. A. ..............................162 (B) Settlage, R. ........................................188 (F) Spurgers, K. ......................................206 (K)
Rose, L. ...................................................126 Sevilla-Reyes, E. E. ..................................022 Spurgers, K. B. ...................................208 (K)
Rosen, G. ...........................................095 (I) Sewald, K. ...........................................214 (I) Squires, R. B. .....................................187 (F)
Roth, A. I. .........................................103 (K) Shadomy, S. .......................................067 (D) Stahl-Hennig, C. ...............................203 (K)
Roussel, P. ...............................................226 Shafagati, N. ............................................115 Stanker, L. H. ...........................................111
Rowan, L. .........................................206 (K) Shaffer, S. A. .............................................116 Stein, K. ............................................196 (G)
Roy, C. J. .................................................233 Shah, K. ............................................196 (G) Stephansson, O. .................................055 (B)
Rudge, T. L. ......................................064 (D) Shah, S. R. ........................................071 (E) Stevens, M. P. .....................................154 (B)
Russell, R. ............................092 (H), 097 (I) Shamloul, M. .....................................197 (K) Steward-Clark, E. ..............................067 (D)
Russo, A. ...........................................201 (K) Sharma, A. ....................199 (K), 206 (K), 222 Stinchcomb, D. ..................................205 (K)
Russo, A. T. ..............................................228 Sharma, S. .........................................086 (H) Stoddard, R. A. ..................................067 (D)
Rutebarika, C. S. ...............................107 (K) Sharpe, D. C. ....................................220 (K) Stojadinovic, A. ..................................053 (B)

Ryan,V. .............................................075 (E) Shearer, J. ..........................................091 (H) Stokes, M. G. .....................................198 (K)

Saah, J. R. ..........................................183 (E) Sherchan, S. P. ...................................164 (B) Storch, S. ....................................201 (K), 228

Sabourin, C. L. ..................................064 (D) Sherratt, D. ........................................035 (B)


Sherwood, R. ....................................201 (K) Topic Categories:
Sadat, E. .............................................187 (F) Below is a list of descriptions of topic cate-
Salerno, G. ...............................................226 Sherwood, R. L. ....................092 (H), 097 (I) gories (letters in parentheses by each pres-
entation number refer to this chart).
Samreen, A. .......................................141 (A) Shields, M. .........................................171 (B)
Shin,Y.-W. ..........................................096 (I) A. Viral Agents
Sanchez, M. .......................................073 (E) B. Bacterial Agents
Sankovich, K. A. ................................064 (D) Shinmura,Y. .....................................090 (H) C. Fungal Agents
Shirey, K. ...........................................049 (B) D. Diagnostics
Santiago, A. ........................................049 (B) E. Environmental Detection
Sarrazine, S. A. ...................................057 (B) Shirley, R. .........................................088 (H) F. Informatics and Genomics
Shrutleff, A. .............................................227 G. Therapeutics
Schaefer, L. ........................................059 (B) H. Vaccines
Scheuermann, R. H. ..........................187 (F) Shuman, H. A. ..................................194 (G) I. Immune Responses
Shuttleworth, H. ................................161 (B) J. Decontamination, Biosafety,
Schmaljohn, C. ..................................082 (G) and Containment
Schneewind, O. ...................039 (B), 041 (B), Siefkas, K. ..........................................061 (B) K. Other and Information Only
.........051 (B), 081 (G), 087 (H), 118, 152 (B), Silvera, P. ...........................................196 (G)

BIODEFENSE PROGRAM & ABSTRACTS 107


AUTHOR I N DE X

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Stratton, D. ........................................043 (B) Tsai, P. ................................................095 (I) Wang, D. .................................................009


Strohmeier, O. ...................................065 (D) Tsanava, S. ............................046 (B), 159 (B) Wang, X. .................................................116
Stuthman, K. ...........................................227 Tsanava, S. .........................................168 (B) Wang,Y.-T. ..................153 (B), 155 (B), 234
Suar, M. .............................................157 (B) Tsanava, S. A. .....................................070 (E) Warawa, J. M. ...................................117, 246
Sullivan, D. ........................................061 (B) Tsang, C. H. ......................................161 (B) Ward, S. .............................................202 (K)
Sun,Y. .....................................................117 Tsertsvadze, N. .....................046 (B), 159 (B) Warren, R. ........................................193 (G)
Surnev, D. ........................................084 (H) Tsertsvadze, N. ..................................168 (B) Warren, R. L. .....................................057 (B)
Sutton,V. ...........................................145 (A) Tsertsvadze, N. S. ...............................070 (E) Warren, T. ................................................227
Svoboda, P. .........................................210 (I) Tuanyok, A. .......................................057 (B) Waters, C. .........................................068 (D)
Swearengen, J. .........................................005 Turell, M. J. ........................................078 (F) Waters, C. ................................................138
Swiderski, W. .....................................091 (H) Turner, J. ...........................................208 (K) Way, M. ...................................................244
Szabo, J. ...................................................127 Twenhafel, N. A. ................................034 (B) Weaver, S. C. ...........................................240
Taft, S. C. ..........................................162 (B) Udoh, I. P. .........................................147 (B) Weidmann, M. ..................................065 (D)
Tallon, L. ...........................................079 (F) Uemura, C. .......................................090 (H) Weir, J. ................................................213 (I)
Tan, J. ................................................028 (A) Uhlenhaut, C. ...................................219 (K) Weiss, M. L. .......................................220 (K)
Tauxe, R. ................................................024 Ulianova, O.V. ...................................166 (B) Welch, W. ..........................................156 (B)
Taviani, E. ..........................................185 (E) Uzarski, J. ...........................................101 (J) Wells, J. ...................................................227
Tayyab, S. M. H. .................................077 (F) Valderas, M. W. ......................092 (H), 097 (I) Whitehouse, C. A. ................078 (F), 148 (B)
Tello, C. A. ........................................060 (B) Valle, E. .............................................171 (B) Wilder, J. ...............................092 (H), 097 (I)
Tempel, R. .......................................088 (H) van der Bruggen, K. ................................249 William, P. .........................................200 (K)
Tettelin, H. ........................................079 (F) van Hoek, M. L. ......052 (B), 054 (B), 083 (G) Williams, D. ......................................062 (D)
Theis, T. ...........................................175 (D) van Oordt, T. .....................................065 (D) Williams, P. .......................................150 (B)
Thomas, J. ..........................................212 (I) Van Tongeren, S. ......................................227 Williams, R. .........................029 (A), 030 (A)
Thomas, J. G. ....................................068 (D) Van Zandt, K. E. ...............................057 (B) Williams, S. K. ...................................148 (B)
Thomas, M. .......................................171 (B) VanderNoot,V. ..................................069 (D) Williamson, E. D. ...............................198 (K)
Thompson, I. J. .................................198 (K) VanDuyne, R. .........................................114 Wirblich, C. ......................................207 (K)
Thomson, G. ...........................................023 Vassar, M. ..........................................091 (H) Wobus, C. ...............................................120
Thorkildson, P. ..................................156 (B) Vay, C. ...............................................165 (B) Wölfel, R. .........................063 (D), 181 (E)
Timlin, J. A. .............................................224 Veres, T. ..............................................214 (I) Wolfraim, L. ......................................085 (H)
Tolley, H. ...........................................142 (A) Verrier, B. ..........................................204 (K) Wolfraim, L. ......................................209 (K)
Tolmasky, M. ....................................035 (B) Vincent, G. ........................................190 (F) Wong, K. K. .....................................143 (A)
Tonks, K. M. ......................................161 (B) Vipond, J. ................045 (B), 149 (B), 161 (B) Wood, J. ..................................................125
Torres, A. .................................................233 Vipond, R. ........................................190 (F) Woodman, M. E. ................................211 (I)
Tournier, J.-N. ...........................204 (K), 223 Vishwakarma,V. ................................157 (B) Wooten, R. M. ...................................211 (I)
Traglia, G. ..........................................165 (B) Vogel, S. ............................................049 (B) Worth, R. G. .......................................211 (I)
Tran, M. ............................................028 (A) Volland, H. .......................................184 (E) Wu, W. ................................................216 (J)
Tran, T. ..............................................035 (B) Volland, H. ........................................186 (E) Wycoff, K. .........................................059 (B)
Tran-Gyamfi, M. ...............................069 (D) von Stetten, F. ...................................065 (D) Xiao, L. ...............................................212 (I)
Trefethen, J. .......................................197 (K) Voth, D. ...................................................254 Yang, A. ...................................................222
Trescos,Y. ..........................................204 (K) Wagner, D. ..............................................012 Yao, H. ....................................................226
Tret, J. O. ...........................................082 (G) Wagner, D. .........................................168 (B) Yates, C. ..................................................229
Trivedi, L. .........................................178 (D) Walper, S. A. .....................................179 (D) Ye, L. ..................................................098 (I)

108 10th ASM Biodefense and Emerging Diseases Research Meeting


AUTHOR INDEX

Listed by Author Name, Presentation Number, Abstract Category. *Presenting author is in Bold Face Type.

Yeager, J. ................029 (A), 030 (A), 038 (B)


Yee, S. B. ...........................................034 (B)
Yin, J. ................................................080 (G)
Yokoi, K. ..........................................090 (H)
Yokote, H. ........................................090 (H)
yole, D.Y. S. NA. .................................094 (I)
Young, G. ..........................................205 (K)
Yu, Kuznetsov ...................................084 (H)
Yue, C. ..............................................203 (K)
Yung, P. ..............................................216 (J)
Yusibov,V. .........................................197 (K)
Zakalashvili, M. .................................159 (B)
Zaremba, S. .......................................187 (F)
Zelazowska, E. B. ...............................170 (B)
Zhang,Y. ............................................187 (F)
Zhgenti, E. ...........................047 (B), 159 (B)
Zhghenti, E. ......................................168 (B)
Zhou, L. ............................................187 (F)
Ziese, M. ...........................................028 (A)
Zorreguieta, A. ..................................035 (B)
Zumbrun, E. ........................029 (A), 030 (A)
Zumbrun, E. E. ................................146 (A)

Topic Categories:
Below is a list of descriptions of topic cate-
gories (letters in parentheses by each pres-
entation number refer to this chart).

A. Viral Agents
B. Bacterial Agents
C. Fungal Agents
D. Diagnostics
E. Environmental Detection
F. Informatics and Genomics
G. Therapeutics
H. Vaccines
I. Immune Responses
J. Decontamination, Biosafety,
and Containment
K. Other and Information Only

BIODEFENSE PROGRAM & ABSTRACTS 109


NOTE S

110 10th ASM Biodefense and Emerging Diseases Research Meeting


NOTE S

BIODEFENSE PROGRAM & ABSTRACTS 111


NOTE S

112 10th ASM Biodefense and Emerging Diseases Research Meeting


A M E R I C A N S O C I E T Y F O R M I C R O B I O L O G Y

ASM TELECONFERENCES
March 7, 2012
Optimizing Specimen Management
J. Michael Miller

March 28, 2012


Medical Entomology: Arthropod Pests of Public
Health Importance
Chris Evans

April 11, 2012


CLIA: Past, Present, and a Glimpse of the Future ASM CONFERENCES
Nancy Anderson

May 16, 2012


March 29–April 2, 2012
What Does A Culture of Safety Mean in a Clinical
11th ASM Conference on Candida and Candidiasis
Microbiology Lab?
San Francisco, California
Rebecca T. Horvat
May 6–10, 2012
August 29, 2012
4th ASM Conference on Prokaryotic Cell Biology
Tools for Identification of Mycobacteria in 2012
and Development
Susan M. Harrington
Montreal, Canada
September 26, 2012
June 26–29, 2012
HIV Laboratory Diagnosis: Current and
3rd ASM Conference on Antimicrobial Resistance in
Future Methodology
Zoonotic Bacteria and Foodborne Pathogens
Michele Owen
Aix–en–Provence, France
October 24, 2012
September 29–October 4, 2012
Emergency Preparedness
6th ASM Conference on Biofilms
Thomas Williams
(ASM Biofilms 2012)
Miami, Florida
www.asmaudio.org
October 22–26, 2012
4th ASM Conference on Beneficial Microbes
San Antonio, Texas

www.asm.org/conferences

You might also like