On-Site Progr

100
1 Table of Contents Conference Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .2 Conference Objectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .2 Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .3 Conference Co-Chairs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .4 Conference Organizing Committee . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .4 Scientific Program Committee . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .5 Executive Staff . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .5 Invited Presenters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .6 General Information . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9 American with Disabilities Act . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9 Conference Information Desk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9 Conference Location . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9 Continuing Medical Education (CME) Accreditation . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9 Exhibit Hall . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10 Messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10 No Smoking Policy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10 Poster Sessions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10 Press Room . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10 Program and Abstracts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10 Registration Fees and Hours . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11 Speaker Ready Room and Audiovisual Equipment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11 Verification of Attendance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11 Affiliated Events and Other Meetings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11 Hotel Floorplan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .12 Program At-A-Glance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .13 Final Program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .14 Meet the Experts Presenters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .39 Abstracts Abstracts of Invited Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .45 Abstracts of Oral Submitted Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .57 Abstracts of Submitted Poster Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .67 Author Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .94 Disclosure Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .99

Transcript of On-Site Progr

Page 1: On-Site Progr

1

Table of Contents

Conference Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .2

Conference Objectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .2

Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .3

Conference Co-Chairs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .4

Conference Organizing Committee . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .4

Scientific Program Committee . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .5

Executive Staff . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .5

Invited Presenters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .6

General Information . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9

American with Disabilities Act . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9

Conference Information Desk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9

Conference Location . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9

Continuing Medical Education (CME) Accreditation . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9

Exhibit Hall . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10

Messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10

No Smoking Policy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10

Poster Sessions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10

Press Room . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10

Program and Abstracts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .10

Registration Fees and Hours . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11

Speaker Ready Room and Audiovisual Equipment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11

Verification of Attendance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11

Affiliated Events and Other Meetings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .11

Hotel Floorplan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .12

Program At-A-Glance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .13

Final Program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .14

Meet the Experts Presenters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .39

Abstracts

Abstracts of Invited Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .45

Abstracts of Oral Submitted Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .57

Abstracts of Submitted Poster Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .67

Author Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .94

Disclosure Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .99

Page 2: On-Site Progr

2

Eighth Annual Conference

Overall Conference ObjectivesAt the conclusion of this conference, participants should be able tomeet the following objectives: ■ Discuss recent scientific advances that are contributing to progress in

the development of vaccines■ Identify research opportunities and scientific challenges associated

with vaccine development, production, and distribution

Session Specific ObjectivesKeynote Address■ Identify needs and challenges associated with vaccine use, and to

demonstrate the reasons that research and development of newvaccines must be intensified

Mary Lou Clements-Mann Memorial Lecture in Vaccine Sciences■ Define the burden of bacterial pneumonia using conjugate vaccines

as probes

Conjugate Vaccine Issues■ Understand the methods used for measuring vaccine efficacy under

condition of routine use in the UK; how the efficacy estimates havebeen used to help our understanding of the correlates of protection;and how transmission models of diseases such as meningococcalserogroup C can assist in understanding the impact of a vaccinationprogramme at the population level

■ Understand the role of functional antibodies in protection of humans againstdisease caused by encapsulated bacteria and how conjugate vaccines, incontrast to polysaccharide vaccines elicit antibody responses in young infants

■ Know the role of T cells in providing immunological help to B cells stimulatedby conjugate vaccines

■ Discuss target antigens of Bacillus anthracis that can serve as the basis fornew and improved anthrax vaccines and the general advantages ofconjugate vaccines as a vaccinology strategy

■ Understand the efficacy of conjugate pneumococcal vaccines in rural Africaand consider the implications for vaccination policies

Update on Vaccines Against Enteric and Oral Infections■ Provide an update on the current status of knowledge of rotavirus vaccine

development and clinical trials, and know the challenges which remain forthe future introduction of rotavirus vaccines

■ Understand the impact of norovirus infection on different populations,barriers to the development of vaccines to prevent norovirus infection, andidentify potential norovirus vaccine candidates

■ Know the rationale for developing a vaccine to prevent clostridium difficileassociated diarrhea; the approach to vaccine development; and the resultsof early clinical trials

■ Understand the basic microbial pathogenesis of dental caries, the basicapproaches to prevent dental caries, and the statues of preclinical andclinical studies aimed at developing immunologic means of preventingdental caries, especially regarding the development of mucosal anti-cariesvaccines

Conference OverviewThe remarkable pace of biotechnology discovery is continuing unabated. New cytokines are identified, immune regulatorypathways unraveled, promising adjuvants reported, and investigational products revealed to have high degrees of protectionfor humans against viral diseases not yet vaccine preventable, such as human papilloma virus and herpes simplex. The toolsof vaccination are being applied therapeutically for various cancers and chronic conditions.

The Annual Conference on Vaccine Research provides high-quality, current reports of scientific progress featured in bothinvited presentations and submitted abstracts. The disparate fields covered in both human and veterinary vaccinologyencourage valuable cross-fertilization of ideas and approaches among researchers otherwise focused on specific diseases ormethods.

The Conference has become the largest scientific meeting devoted exclusively to research on vaccines and associatedtechnologies for disease prevention and treatment through immunization. The Eighth Annual Conference promises tomaintain this tradition as the premier venue for cutting edge topics and issues. International experts will lead seminars andpanel discussions on topical areas of basic immunology, product development, clinical testing, regulation, and other aspectsof vaccine research. Opportunities for networking and scientific collaboration critical to advancing vaccine science anddevelopment will be available through audience discussions, meet the experts breakfast sessions, poster presentations,sponsored exhibits, evening ceremonies, and receptions

Conference Objectives

Page 3: On-Site Progr

on Vaccine Research

3

Influenza and Vaccines for Emergency Pandemics■ Understand how vaccine use in poultry will aid in control of the

H5N1 avian influenza outbreak in Asia and reduce risk for humaninfections

■ Review the epidemiology of West Nile virus in North America■ Understand the regulatory considerations for the emergency use of

veterinary vaccines (biologics) in the United States

Concrete Proposals for Ensuring a Secure Supply of Vaccines in the U.S.■ Describe the issues and possible solutions needed to limit shortages

and strengthen vaccine supply in the United States; report onprogress made in the last three years; and to address currentchallenges

■ List six consensus policy recommendations to strengthen adultimmunization in the U.S. and how, after years of discussionwithout agreement, a consensus policy agenda was achieved

Vaccinology of Neglected Diseases: Malaria■ Understand the rationale, background and recent progress with

the RTS,S malaria vaccine and to provide an overview of the nextsteps in its clinical development

■ Acknowledge that vaccines bearing single pre-erythrocyticantigens are capable of impacting parasitologic as well as clinicaloutcomes under conditions of natural malaria exposure.

■ Describe the safety and immunologic profiles of recent adjuvantsthat have been clinically evaluated in the context of malariavaccines

Therapeutic Vaccines■ Explain the status of the development of preventive and

therapeutic vaccines for HPV induced cervical cancer and thebasic immunology that is needed to guide these vaccinedevelopments

■ Identify a therapeutic vaccine approach for treatment ofAlzheimer's disease

■ Summarize the current status of and challenges to thedevelopment of vaccines for Type 1 diabetes

■ Describe the huge disease burden associated with HCV infectionin the US and globally; and vaccine formulations designed toboost cross-neutralizing antibody titers and broadly reactivecellular immune responses

■ Discuss the design of clinical trials underway and planned fornovel immunotherapy strategies

Vaccinology Impact of Recent Advances in Immunology■ Understand the process of innate imprinting and its potential

application to the treatment of inflammatory disease■ Identify the main limitations which limit neonatal and infant IgG

responses to immunization and gain understanding in theexperimental approaches that may be used to study thedeterminants of such limitations

Acknowledgments (as of April 20, 2005)

This conference is supported, in part, through unrestricted educationalgrants from:

■ Acambis■ Aeras Global TB Vaccine Foundation■ Antigenics, Inc.■ Baxter Healthcare■ Becton Dickinson■ Bill & Melinda Gates Foundation■ Chiron Vaccines■ Coley Pharmaceutical Group■ Dynport Vaccine Company■ EpiVax, Inc.■ GlaxoSmithKline■ Globe Immune, Inc.■ Iomai Corporation■ MedImmune, Inc.■ Merck & Co., Inc.■ Protein Sciences Corporation■ sanofi pasteur■ U.S. Food and Drug Administration■ VaxGen, Inc■ Vical, Inc.■ Wyeth Pharmaceuticals

■ Determine mechanisms which are responsible for the weaker andshorter duration of B cell responses in early life

■ Learn the current state of knowledge about the molecular basis for thepoor quality of antiviral antibodies made in response to viral infection

Multi-Agent and Chimeric Vaccines■ Provide an update on the progress towards an HIV/AIDS vaccine■ Utlilize new understandings of immunology to develop vaccines based

on novel technology■ Demonstrate that directed molecular evolution creates better vaccines by

recombining genes and selecting chimeric molecules for improvedvaccine efficacy

■ Explain the principles underlying use of yellow fever 17D as a live vectorfor foreign genes, and discuss the status of development and clinicaltesting of new vaccines against dengue, West Nile and Japaneseencephalitis.

Page 4: On-Site Progr

4

Eighth Annual Conference

John D. Clemens, M.D.International Vaccine InstituteSeoul, Korea

Cyril Gerard Gay, D.V.M., Ph.D.U.S. Department of AgricultureBeltsville, Maryland

Myron M. Levine, M.D., D.T.P.H.Center for Vaccine DevelopmentBaltimore, Maryland

Dean D. MasonAlbert B. Sabin Vaccine Institute Washington, DC

Pamela M. McInnes, D.D.S., M.Sc.National Institute of Allergy and Infectious DiseasesBethesda, Maryland

Karen Midthun, M.D.Center for Biologics Evaluation and ResesarchBethesda, Maryland

Peter L. Nara, D.V.M., Ph.D.Biological Mimetics, Inc., Frederick, Maryland

N. Regina Rabinovich, M.D.Bill and Melinda Gates Foundation, Seattle, Washington

Susan J. Rehm, M.D.National Foundation for Infectious Diseases, Bethesda, Maryland

Bruce G. Weniger, M.D.Centers for Disease Control and Prevention, Atlanta, Georgia

Peter L. Nara, D.V.M., Ph.D.Conference Co-Chair

Gregory A. Poland, M.D.International Society for VaccinesRochester, Minnesota

N. Regina Rabinovich, M.D.Conference Co-Chair

Susan J. Rehm, M.D.Conference Co-Chair

Darshna TannaFondation MérieuxLyon Cedex, France

Bernard A.M. Van der Zeijst, Ph.D.Netherlands Vaccine InstituteBilthoven, Netherlands

Bruce G. Weniger, M.D.Conference Co-Chair

Conference Co-Chairs

Conference Organizing Committee

Page 5: On-Site Progr

on Vaccine Research

5

Scientific Program Committee

Lorne A. Babiuk, Ph.D.Vaccine and Infectious Disease OrganizationSaskatoon, Canada

Carol J. Baker, M.D.Baylor College of MedicineHouston, Texas

Richard J. Duma, M.D., Ph.D.Halifax Medical CenterDaytona Beach, Florida

Hana Golding, Ph.D.Center for Biologics Evaluation and ResearchBethesda, Maryland

Diane E. Griffin, M.D., Ph.D.Johns Hopkins University Bloomberg School of Public HealthBaltimore, Maryland

Paul-Henri Lambert, M.D.Centre Medical Universitaire de GenéveGeneva, Switzerland

Myron M. Levine, M.D., D.T.P.H.Member, Conference Organizing Committee

Pamela M. McInnes, D.D.S.Member, Conference Organizing Committee

Peter L. Nara, D.V.M., Ph.D.Conference Co-Chair

Stanley A. Plotkin, M.D.University of PennsylvaniaDoylestown, Pennsylvania

N. Regina Rabinovich, M.D.Conference Co-Chair

Rino Rappuoli, Ph.D.IRIS, Chiron, SpASienna, Italy

Susan J. Rehm, M.D.Conference Co-Chair

Harriet L. Robinson, Ph.D.Emory University School of MedicineAtlanta, Georgia

Connie Schmaljohn, Ph.D.U.S. Army Medical Research Institute of Infectious DiseasesFrederick, Maryland

Alan R. Shaw, Ph.D.Merck & Company, Inc.West Point, Pennsylvania

George R. Siber, M.D.Wyeth-Lederle VaccinesPearl River, New York

Bruce G. Weniger, M.D.Conference Co-Chair

NFID Executive StaffSharon Cooper-KerrDirector, Events PlanningNational Foundation for Infectious DiseasesBethesda, Maryland

Sheena L. MajetteDirector, Continuing Medical EducationNational Foundation for Infectious DiseasesBethesda, Maryland

Len NovickExecutive DirectorNational Foundation for Infectious DiseasesBethesda, Maryland

Page 6: On-Site Progr

6

Eighth Annual ConferenceInvited Presenters*

Robert L. Atmar, M.D.Associate Professor Departments of Medicine and Molecular Virology and MicrobiologyBaylor College of MedicineHouston, TX

W. Ripley Ballou, M.D.Vice President Clinical Research and Development, Emerging DiseasesGlaxoSmithKline BiologicalsRixensart, Belgium

Alan D.T. Barrett, Ph.D.Professor and Vice Chairman for Research Department of Pathology Associate Director, SealyCenter for Vaccine DevelopmentUniversity of Texas Medical BranchGalveston TX

James E. Crowe, Jr. M.D.Professor of Pediatrics Vanderbilt University School of MedicineNashville, TN

Felicity Cutts, Ph.D.ProfessorInitiative for Vaccine ResearchWorld Health OrganizationPrevessin-Moens, Switzerland

Giuseppe Del Giudice, M.D.Vice President, Serology and Animal ModelChiron VaccinesSiena, Italy

Filip Dubovsky M.D., M.P.H.Scientific Director Malaria Vaccine Initiative, PATHBethesda, MD

David Goldblatt M.D., Ph.D.Professor of Vaccinology and ImmunologyInstitute of Child HealthUniversity College LondonLondon, United Kingdom

Lt. Col. Donald Gray Heppner, M.D.Director, Malaria Vaccine ProgramDepartment of ImmunologyWalter Reed Army Institute of ResearchSilver Spring, MD

David L. Heymann, M.D.Executive Director Communicable DiseasesWorld Health OrganizationGeneva, Switzerland

Richard E. Hill, Jr., D.V.M.Director Center for Veterinary BiologicsUSDA, Animal and Plant Health Inspection ServiceAmes, IA

Michael Houghton, Ph.D.Vice President and Fellow Hepatitis C ResearchChiron CorporationEmeryville, CA

Tracy Hussell, Ph.D.Reader, Kennedy Institute Technology and MedicineImperial College of ScienceLondon, England

Richard A. Insel, M.D.Executive Vice President of Research Juvenile Diabetes Research Foundation InternationalNew York, NY

Page 7: On-Site Progr

on Vaccine Research

7

Invited Presenters*

David R. Johnson, M.D., M.P.H.Director, Scientific & Medical Affairs sanofi pasteurSwiftwater, PA

Stefan H.I. Kappe, Ph.D.Independent Investigator Affiliate Professor of PathobiologyUniversity of WashingtonSeattle Biomedical Research InstituteSeattle, WA

W. Martin Kast, Ph.D.Walter A. Richter Cancer Research Chair Professor of Molecular Microbiology and ImmunologyNorris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos Angeles, CA

Jerome O. Klein, M.D.Professor and Vice Chairman for Academic Affairs Department of PediatricsBoston Medical CenterBoston, MA

Keith P. Klugman, MD, PhDProfessor, Infectious Diseases Department of International HealthEmory UniversityAtlanta, GA

Karen L. Kotloff, MDProfessor of Pediatrics and Medicine Center for Vaccine DevelopmentUniversity of Maryland School of MedicineBaltimore, MD

Antonio Lanzavecchia, M.D.DirectorInstitute for Research in BiomedicineBellinzona, Switzerland

Christopher P. Locher Ph.D.Project Leader Department of Infectious DiseasesMaxygen, Inc.Redwood City, CA

Suzanne M. Michalek, Ph.D.ProfessorDepartment of MicrobiologyUniversity of Alabama at BirminghamBirmingham, AL

Elizabeth Miller, M.D.Head, Immunization Deparatment Health Protection AgtencyCommunicable Disease Surveillance CenterLondon, United Kingdom

Thomas P. Monath, MDChief Scientific Officer Acambis, Inc.Cambridge, MA

Albert D.M.E. Osterhaus, D.V.M., Ph.D.Head, Department of Virology Erasmus University RotterdamRotterdam, Netherlands

John Robbins, M.D.ChiefLaboratory Development and Molecular Immunology NICHD/National Institutes of HealthBethesda, MD

Harriet L. Robinson, PhDAsa Griggs Candler Professor Chief, Division of Microbiology and ImmunologyYerkes Regional Primate Research CenterEmory UniversityAtlanta, GA

Page 8: On-Site Progr

8

Dale Schenk, Ph.D.Senior Vice PresidentChief Scientific Officer Elan PharmaceuticalsSouth San Francisco, CA

John W. Shiver, Ph.D.Vice President Vaccine and Biologics ResearchMerck and Company, Inc.West Point, PA

Claire-Anne Siegrist, M.D.Professor of Vaccinology Director of the Pediatric Department, Director of the WHOCollaborating Center for Neonatal VaccinologyUniversity of Geneva, CMUGeneva, Switzerland

Duncan Steele, M.D.Scientist and Responsible OfficerDepartment of Immunization, Vaccines and BiologicalsWorld Health OrganizationGeneva, Switzerland

David E. Swayne, D.V.M., Ph.D.Laboratory Director Southeast Poultry Research LaboratoryUSDA, Agricultural Research ServiceAthens, GA

*Speakers and presentations subject to change

Eighth Annual ConferenceInvited Presenters*

Page 9: On-Site Progr

on Vaccine Research

9

General Information

AMERICANS WITH DISABILITIES ACTThe Baltimore Marriott Inner Harbor Hotel is fully accessible to the public in accordance with the Americanswith Disabilities Act guidelines. If you have any special meeting needs or requirements, please contact eitherSharon Cooper-Kerr or a member of the hotel staff.

CONFERENCE INFORMATION DESKThe Conference Information Desk is located in the foyer area outside the Grand Ballroom. Conference staffwill be available at the desk throughout the conference.

CONFERENCE LANGUAGEThe official language for the conference is English.

CONFERENCE LOCATIONAll sessions of the conference will be held at:

Baltimore Marriott Inner Harbor Hotel110 South Eutaw StreetBaltimore, Maryland 21201(410) 962-0202

GENERAL CME INFORMATION

The National Foundation for Infectious Diseases (NFID) is accredited by the Accreditation Council forContinuing Medical Education (ACCME) to provide Continuing Medical Education (CME) for physicians.NFID takes responsibility for the content, quality, and scientific integrity of this CME activity.

NFID designates this CME activity for a maximum of 21 Category 1 credits toward the AMA Physician’sRecognition Award. Each physician should claim only those hours of credit that he/she actually spent in theeducational activity.

Designated CME ActivitiesSessions designated with a symbol have been approved for CME Credit. No other sessions are eligible for CME credit hours.

CME CertificatesIn order to ensure that you receive the CME credit hours to which you are entitled, please note the following:

1. Complete the CME application for credits located at the Conference Information Desk.2. Return your completed application and conference evaluation to conference staff at the Conference

Information Desk.

CME

Page 10: On-Site Progr

10

CME DisclosuresIn order for program sessions to be accredited, program presenters must disclose to the conference participantsany real or apparent conflict(s) of interest related to the content of their presentations. A summary of theseconflicts of interest is printed separately in this book under the heading Continuing Medical EducationDisclosures (see Table of Contents).

EXHIBIT HALLVisit the Exhibit Hall to meet with representatives from companies displaying the latest technologies in vaccine-related products and services. The exhibit hall hours are Tuesday, May 10, 10:30 a.m. – 3:30 p.m.; andWednesday, May 11, 10:00 a.m. – 3:30 p.m. A prize drawing will be held on Wednesday, May 11, at 1:15 p.m.Be sure to get your exhibitor passport stamped by each of the exhibitors to qualify for the drawing and return tothe conference registration desk by 1:00 p.m., Wednesday, May 11.

MESSAGESAll sleeping rooms in the Baltimore Marriott Inner Harbor Hotel are equipped with a voice mail system. Thissystem is accessible via the hotel operator using the house phone. In case of emergencies requiring immediateattention, your party should call the general hotel number listed below and instruct the switchboard to deliver amessage to Sharon Cooper-Kerr or Sheena Majette at the Vaccine Research Conference Information Deskoutside of the Grand Ballroom. The general hotel number is 1-410-962-0202.

NO SMOKING POLICYThe Baltimore Marriott Inner Harbor Hotel is a non-smoking facility except for specially designated guestrooms and smoking areas of the hotel bars and restaurants. No smoking is allowed in any of the session rooms,coffee break area or in the foyer adjoining the session rooms.

POSTER SESSIONSPosters will be on display on Monday, May 9 from 10:30 a.m. – 11:30 a.m. Presenters will be at their boardsduring this time to answer questions and discuss their research. There will also be a poster reception at 5:30 p.m. on Monday which will conclude the poster program. Posters and the Reception will be located in the Stadium Ballroom located on the second floor of the hotel.

PRESS ROOMNFID will have a Press Room located in the Westminster Room. Members of the press should sign in at theConference Information Desk during registration hours.

PROGRAM AND ABSTRACTSEach registered participant will receive one complimentary copy of the Final Program and Abstract Book as partof his/her registration fee. Additional copies, if available, can be purchased for $25. Orders for additionalcopies can be taken at the Conference Information Desk starting Tuesday, May 10, and after the conference bye-mail to [email protected], phone at (301) 656-0003 x19, or by fax at (301) 907-0878.

Eighth Annual Conference

Page 11: On-Site Progr

on Vaccine Research

11

REGISTRATION FEES AND HOURSThe onsite registration fee: US $450.00 (Non Member) and $405.00 (NFID Supporting Member)

Space is limited to the first 525 registrants. The registration fee includes a program/abstract book, continentalbreakfast on each day of the conference, all scheduled coffee breaks, lunch presentation on Monday, and thereceptions on Monday and Tuesday. Accommodations and additional meals (i.e. lunch) are not included.

Individuals interested in registering onsite may do so at the Conference Information Desk between thefollowing times:

Sunday, May 8 . . . . . . . . . . . . . . . . . . . . .7:00 p.m. - 9:00 p.m.Monday, May 9 . . . . . . . . . . . . . . . . . . . .7:00 a.m. - 5:00 p.m.Tuesday, May 10 . . . . . . . . . . . . . . . . . . . .7:00 a.m. - 5:00 p.m.Wednesday, May 11 . . . . . . . . . . . . . . . . .7:30 a.m. - noon

Speaker Ready Room and Audiovisual EquipmentA room has been set aside for speakers to preview their slides. All speakers should check in at the ConferenceInformation Desk to be directed to the speaker ready room. The room will be open during the registrationhours and will be equipped with a laptop for preview of your PowerPoint presentation.

Standard session room setup includes a PC, 250 zip drive, LCD projector, laser pointer, podium microphone,and aisle microphone.

Verification of AttendanceInternational attendees may obtain a letter of attendance verification from the staff at the ConferenceInformation Desk during registration hours.

Affiliated Events and Other MeetingsMONDAY, MAY 9, 2005Conference on Vaccine Research Organizing and Scientific Program Committees Meeting (Closed meeting)6:00 p.m. – 9:00 p.m.Patapsco/Severn Rooms

TUESDAY, MAY 10, 2005Albert B. Sabin Vaccine Institute Award Ceremony and Reception6:00 p.m. – 7:00 p.m., Grand Ballroom Foyer (Reception)7:00 p.m. - 8:00 p.m., Grand Ballroom (Ceremony)

Page 12: On-Site Progr

12

Eighth Annual ConferenceHotel Floor Plan

Page 13: On-Site Progr

on Vaccine Research

13

PROGRAM-AT-A-GLANCE

SUNDAY, MAY 8 MONDAY, MAY 9 TUESDAY, MAY 10 WEDNESDAY, MAY 11

7:00 Registration Registration Meet the ExpertsMeet the Experts Breakfast SessionBreakfast Session

7:30 Poster Set-up Continental Breakfast RegistrationContinental Breakfast/Exhibits

8:00 Continental Breakfast Symposium 3: Symposium 6: Influenza and Vaccines Therapeutic Vaccinesfor Emergency Pandemics

8:30 Welcome and Introductions

8:35 Keynote Address

9:20 Mary Lou Clements-Mann Memorial Lecture

10:00 Coffee Break Coffee Break/Exhibits

10:30 Poster Session Coffee Break/Exhibits Submitted Presentations 3&4

11:00 Submitted Presentations 1&2

11:45 Charles Mérieux Award Luncheon

12:00 Lunch (on your own)

12:30 Lunch (on your own)

1:00 Symposium 1: Symposium 7:Conjugate Vaccine Issues Vaccinology Impact of Recent

Advances in Immunology

1:30 Symposium 4:Concrete Proposals for Ensuring a Secure Supply of Vaccines in the U.S.

3:00 Coffee Break Coffee Break/Exhibits Coffee Break/Exhibits

3:30 Symposium 2: Symposium 5: Symposium 8:Update on Vaccines Against Vaccinology of Neglected Multi-Agent and Chimeric VaccinesEnteric and Oral Infections Diseases: Malaria

5:30 Adjournment Adjournment Adjournment/Participant EvaluationPoster Reception

6:00 Albert B. Sabin VaccineInstitute Reception

7:00 Registration Presentation of the Albert B. Sabin Gold Medal

Page 14: On-Site Progr

14

FINAL PROGRAM

SUNDAY, MAY 8, 2005

7:00 p.m. – 9:00 p.m. Early Registration Grand Ballroom Foyer

MONDAY, MAY 9, 2005

7:00 a.m. – 5:00 p.m. Registration Grand Ballroom Foyer

7:30 a.m. Poster Set-Up Stadium Ballroom

8:00 a.m. Continental Breakfast Grand Ballroom Foyer

8:30 a.m. Welcome and Introductions Grand Ballroom

Susan J. Rehm, M.D.National Foundation for Infectious DiseasesBethesda, MD

Keynote Address Grand Ballroom

Moderator: Bruce G. Weniger, M.D.Centers for Disease Control and Prevention

8:35 a.m. 1. Dilemmas in Public Health: From Smallpox to Polio, SARS, and Avian InfluenzaDavid L. Heymann, M.D.World Health OrganizationGeneva, Switzerland

9:10 a.m. Questions and Answers

Mary Lou Clements-Mann Memorial Lecture in Vaccine Sciences Grand Ballroom

Moderator: George R. Siber, M.D.Wyeth-Lederle Vaccines

9:20 a.m. 2. Use of Conjugate Vaccines as a Probe to Define the Role of the Pneumococcus in the Etiology of PneumoniaKeith P. Klugman, M.D., Ph.D.Emory University, Department of International HealthAtlanta, GA

9:50 a.m. Questions and Answers

CME

CME

Eighth Annual Conference

Page 15: On-Site Progr

on Vaccine Research

15

10:00 a.m. Coffee Break Stadium Ballroom

10:30 a.m. Poster Session Stadium Ballroom

11:45 a.m. Charles Mérieux Award Luncheon Grand BallroomHonoring Kristin L. Nichol, M.D., Ph.D.Professor of Medicine, University of Minnesota andChief of Medicine, VA Medical CenterMinneapolis, MN

Symposium 1: Conjugate Vaccine Issues Grand Ballroom Moderator: Myron M. Levine, M.D., D.T.P.H.

University of Maryland School of Medicine

1:00 p.m. 3. Avidity and Biological Activity of Antibodies Elicited by Pneumococcal, Hib, and Meningococcal Conjugate VaccinesDavid Goldblatt, M.D., Ph.D.University College of LondonLondon, United Kingdom

1:25 p.m. Questions and Answers

1:30 p.m. 4. Long-term Immunologic Memory: Insights from the UK Experience with Haemophilus Influenzae Type b and Meningococcal C Conjugates Elizabeth Miller, M.D.Communicable Disease Surveillance CenterLondon, United Kingdom

1:55 p.m. Questions and Answers

2:00 p.m. 5. Results of the Pneumococcal Conjugate Vaccine TrialFelicity Cutts, Ph.D.World Health OrganizationGeneva, Switzerland

2:25 p.m. Questions and Answers

2:30 p.m. 6. New Conjugate Vaccine to Prevent AnthraxJohn Robbins, M.D.National Institute of Child Health and Human Development, NIHBethesda, MD

FINAL PROGRAM

Page 16: On-Site Progr

16

MONDAY, MAY 9, 2005 (CONTINUED)

2:55 p.m. Questions and Answers

3:00 p.m. Coffee Break Stadium Ballroom

Symposium 2: Update on Vaccines Against Enteric Grand Ballroom

and Oral Infections Moderator: Richard J. Duma, M.D., Ph.D.

Halifax Medical Center

3:30 p.m. 7. New Rotavirus VaccinesDuncan Steele, M.D.World Health OrganizationGeneva, Switzerland

3:55 p.m. Questions and Answers

4:00 p.m. 8. Progress in Vaccines Against Norwalk VirusRobert L. Atmar, M.D.Baylor College of MedicineHouston, TX

4:25 p.m. Questions and Answers

4:30 p.m. 9. Immunology and Potential Vaccine Prevention of Clostridium difficile ColitisKaren L. Kotloff, M.D.University of Maryland School of MedicineBaltimore, MD

4:55 p.m. Questions and Answers

5:00 p.m. 10. Update on Microbiology, Immunology and Vaccine Prevention of Dental CariesSuzanne M. Michalek, Ph.D.University of Alabama at BirminghamBirmingham, AL

5:25 p.m. Questions and Answers

5:30 p.m. Adjournment and Poster Reception Stadium Ballroom

FINAL PROGRAM

CME

Eighth Annual Conference

Page 17: On-Site Progr

on Vaccine Research

17

TUESDAY, MAY 10, 2005

7:00 a.m.–5:00 p.m. Registration Grand Ballroom Foyer

7:00 a.m.–7:45 a.m. Meet the Experts Breakfast Session Patapsco Room

7:30 a.m. Continental Breakfast Grand Ballroom Foyer

Symposium 3: Influenza and Vaccines for Emergency Pandemics Grand BallroomModerator: Peter L. Nara, D.V.M., Ph.D.

Biological Mimetics, Inc

8:00 a.m. 11. Perspective of Vaccine ManufacturersGiuseppe Del Giudice, M.D.Chiron VaccinesSiena, Italy

8:25 a.m. Questions and Answers

8:30 a.m. 12. Vaccines Against SARS: Where Do We Stand?Albert D.M.E. Osterhaus, D.V.M., Ph.D.Erasmus Medical CollegeRotterdam, Netherlands

8:55 a.m. Questions and Answers

9:00 a.m. 13. Vaccines for Prevention, Management, and Eradication of Avian InfluenzaDavid Swayne, M.D.United States Department of AgricultureAthens, GA

9:25 a.m. Questions and Answers

9:30 a.m. 14. West Nile: An Overview of the Epidemic in North AmericaAlan D.T. Barrett, Ph.D.University of Texas Medical BranchGalveston, TX

9:55 a.m. Questions and Answers

10:00 a.m. 15. Vaccination for Emergency and Emerging Animal Health EventsRichard E. Hill, Jr., D.V.M.United States Department of AgricultureWashington, DC

FINAL PROGRAM

CME

Page 18: On-Site Progr

18

TUESDAY, MAY 10, 2005 (CONTINUED)

10:25 a.m. Questions and Answers

10:30 a.m. Coffee Break/Exhibits Stadium Ballroom

Submitted Presentations 1: New Vaccines and Novel Vaccine Use Grand Ballroom East/Salons A,B,C(Concurrent Session) Moderator: Giuseppe Del Giudice, M.D.

Chiron Vaccines

11:00 a.m. S1 Lipotechoic Acid Conjugate Vaccine for Staphylococcus A. Lees, J. KoKai-kun, A. LopezAcosta, J. Acevedo, J. MondBiosynexus Inc, Gaithersburg, MD

11:15 a.m. S2 Adult Formulation Tetanus and Diphtheria Toxoids with Acellular Pertussis Vaccine (Tdap) has Comparable Immunogenicity but Less Reactogenicity than DTaP-IPV for the Pre-school, Fifth-dose BoosterJ. M. Langley1, S. A. Halperin1, E. Mills2, A. Tomovici2, R. Guasparini3, G. Predy4, B. Law5, F. Diaz-Mitoma6, P. Whitsitt7, B. Tapiero8, M. Dionne9

1Dalhousie University, Halifax, NS, CANADA, 2Sanofi-Pasteur, Toronto, ON, CANADA,3TASC Research, Surrey, BC, CANADA, 4Capital-Health, Edmonton, AB, CANADA,5Univ-Manitoba, Winnipeg, MB, CANADA, 6CHEO, Ottawa, ON, CANADA,7Paradigm Clin Trials, Oshawa, ON, CANADA, 8Hop-Ste-Justine, Montreal, PQ,CANADA, 9CHUQ, Beauport, PQ, CANADA.

11:30 a.m. S3 An Antigen-antibody-complex- based Therapeutic Vaccine for Chronic Hepatitis B PatientsY. Wen1, D. Xu2, Z. Yuan1, K. Zhao3

1Department of Medical Molecular Virology, Shanghai Medical College, Fudan University,Shanghai, CHINA, 2Dept.Infectious Diseases, Di Tan Hospital, Beijing, CHINA, 3Beijing Institute of Biological Product, Beijingi, CHINA.

11:45 a.m. S4 Different Immune Response after Sequential Use of Pneumococcal Polysaccharide and Pneumococcal Conjugate Vaccine A. de Roux1, B. Schmoele-Thoma2, N. Ahlers2, W. Gruber3, G. Siber3, D. Sikkema3, T. Welte4, H. Lode1

1Heliosklinik Emil-von-Behring, Berlin, GERMANY, 2Wyeth, Muenster, GERMANY,3Wyeth, Pearl River, NY, 4University Hannover, Hannover, GERMANY.

12:00 p.m. S5 Induction of Therapeutic Antitumor Immunity via Chemokine ReceptorMediated Antigen Cross-presentationA. Biragyn, D. Baatar, R. SchiavoLaboratory of Immunology, GRC, National Institute on Aging, Baltimore, MD.

FINAL PROGRAM

CME

Eighth Annual Conference

Page 19: On-Site Progr

on Vaccine Research

19

12:15 p.m. S6 Directed Molecular Evolution Creates HIV-1 Novel gp120 Variants That Induce Broadly Neutralizing Antibodies in RabbitsL. Xu, X. Du, R. Whalen Infectious Diseases, Maxygen, Inc., Redwood City, CA.

12:30 p.m. Lunch (on your own)

Submitted Presentations 2: Vaccines for Enteric and Tropical Pathogens Grand Ballroom West/Salons D,E,F(Concurrent Session) Moderator: Carol J. Baker, M.D.

Baylor College of Medicine

11:00 a.m. S7 The Human Hookworm Vaccine Initiative (HHVI): Progress in the Product Development and Testing of the Na-ASP-2 Hookworm VaccineM. E. Bottazzi1, J. Bethony1, S. Brooker2, G. Goud1, A. Loukas3, S. Mendez1, B. Zhan1, K. Stoever4, P. Hotez1

1Microbiology and Tropical Medicine, The George Washington University, Washington, DC, 2London School of Hygiene and Tropical Medicine, London, UNITED KINGDOM, 3Queensland Institute of Medical Research, Brisbane, AUSTRALIA, 4Human Hookworm Vaccine Initiative, Sabin Vaccine Institute, Washington, DC.

11:15 a.m. S8 The Human Hookworm Vaccine Initiative (HHVI): Novel Design and Statistical Considerations to Estimate the Efficacy of a Helminth Vaccine in Field Trials in Endemic Region: Phase 2b Studies for a Human Hookworm VaccineJ Bethony1, S Brooker2, N Alexander2, S Geiger1, L Rodrigues3, ME Bottazzi1, K Stoever3, P Hotez1

1The George Washington University, USA, 2London School of Hygiene andTropical Medicine, UK, 3Sabin Vaccine Institute, USA

11:30 a.m. S9 Safety and Immunogenicity of Vaccines against Cholera and Enterotoxigenic Escherichia coliDiarrhea in Children In Bangladesh: Problems Encountered and Milestones AchievedF. Qadri, D. A. Sack Laboratory Sciences Division, ICDDR,B, Dhaka, BANGLADESH,

11:45 a.m. S10 Mass Vaccination Against Shigellosis: First Experience of Routine Immunization Against Shigella sonnei InfectionR. P. Chuprinina1, L. I. Pavlova1, T. I. Frolushkina1, A. V. Protodiakonov1, T. V. Gantcho2, M. E. Golovina2, V. I. Shmigol2, S. I. Elkina2, I. Y. Kurbatova2, V. L. Lvov2, P. G. Aparin2

1Carbohydrate Vaccines, Ministry of Health Russia, Moscow, RUSSIAN FEDERATION, 2Carbohydrate Vaccines, NRC-Institute of Immunology, Moscow, RUSSIAN FEDERATION.

FINAL PROGRAM

CME

Page 20: On-Site Progr

20

TUESDAY, MAY 10, 2005 (CONTINUED)

12:00 p.m. S11 Randomized, Double-blind Phase I Study to Assess the Safety, Tolerability, Immunogenicity, Dose Response, and Transmissibility of CVD 1208S, a guaBA, sen, and Set Deleted, Live, Oral Shigella flexneri 2a Soy Based VaccineK. L. Kotloff1, J. K. Simon1, M. Pasetti1, J. P. Nataro1, M. B. Sztein1, S. S. Wasserman2, W. C. Blackwelder1, E. M. Barry2, M. M. Levine1

1Pediatrics, University of Maryland, Baltimore, MD, 2Medicine, University of Maryland, Baltimore, MD.

12:15 p.m. S12 Phase 1 Study of the Safety and Immunogenicity of Amai-C1/Alhydrogel® Vaccine for Plasmodium Falciparum Malaria in Semi-Immune Malian AdultsA. Dicko1, D. Diemert2, I. Sagara1, M. Sogoba1, M. Niambele1, M. Assadou1, O. Guindo1, B. Kamate1, M. Baby1, M. Sissoko1, G. Mullen2, E. Malkin2, M. Sissoko1, M. Thera1, A. Dolo1, C. Long2, D. Diallo1, L. Miller2, A. Saul2, O. Doumbo1; 1MRTC/University of Bamako, Bamako, MALI, 2MVDB/NIAID, Rockville, MD.

12:30 p.m. Lunch (on your own)

Symposium 4: Concrete Proposals for Ensuring A Secure Grand Ballroom

Supply of Vaccines in the U.S. Moderator: Bruce G. Weniger, M.D.

Centers for Disease Control and Prevention

1:30 p.m. 16. Ideas from the National Vaccine Advisory Committee, the Institute of Medicine, and AcademiaJerome O. Klein, M.D.Boston Medical CenterBoston, MA

1:55 p.m. Questions and Answers

2:00 p.m. 17. Vaccine Supply: A Manufacturer’s Perspective on Current Challenges and OpportunitiesDavid R. Johnson, M.D.sanofi pasteurSwiftwater, PA

2:25 p.m. Questions and Answers

FINAL PROGRAM

CME

Eighth Annual Conference

Page 21: On-Site Progr

on Vaccine Research

21

2:30 p.m. 18. A Consensus Agenda to Strengthen U.S. Vaccine SupplyJohn M. ClymerPartnership for PreventionWashington, DC

2:55 p.m. Questions and Answers

3:00 p.m. Coffee Break/Exhibits Stadium Ballroom

Symposium 5: Vaccinology of Neglected Diseases: Malaria* Grand BallroomModerator: N. Regina Rabinovich

Bill & Melinda Gates Foundation

3:30 p.m. 19. Progress in the Field of Malaria VaccinologyFilip Dubovsky, M.D., M.P.H.Malaria Vaccine InitiativeBethesda, MD

3:55 p.m. Questions and Answers

4:00 p.m. 20. Novel and Classical Strategies for Attenuated Malaria VaccinesStephan Kappe, Ph.D.Seattle Biomedical Research InstituteSeattle, WA

4:25 p.m. Questions and Answers

4:30 p.m. 21. The Pathway Forward for Malaria Recombinant Vaccine (RTS,S): Implications of Study ResultsW. Ripley Ballou, M.D.GlaxoSmithKline BiologicalsRixensart, Belgium

4:55 p.m. Questions and Answers

5:00 p.m. 22. Developing Adjuvants for Malaria VaccinesLt. Col. Donald Gray Heppner, M.D.Walter Reed Army Institute of ResearchSilver Spring, MD

5:25 p.m Questions and Answers

5:30 p.m. Adjournment

*This symposium was made possible through an educational grant from the Bill & Melinda Gates Foundation

FINAL PROGRAM

CME

Page 22: On-Site Progr

22

TUESDAY, MAY 10, 2005 (CONTINUED)

6:00 p.m. Albert B. Sabin Vaccine Institute Reception Grand Ballroom Foyer

7:00 p.m. Presentation of the Albert B. Sabin Gold Medal Grand Ballroom

WEDNESDAY, MAY 11, 2005

7:00 a.m.–7:45 a.m. Meet the Experts Breakfast Session Patapsco Room

7:30 a.m.–12:00 p.m. Registration Grand Ballroom Foyer

7:30 a.m. Continental Breakfast/Exhibits Stadium Ballroom

Symposium 6: Therapeutic Vaccines Grand BallroomModerator: Diane E. Griffin, M.D., Ph.D.

Johns Hopkins Bloomberg School of Public Health

8:00 a.m. 23. Human Papilloma Virus Therapeutic VaccinesW. Martin Kast, Ph.D.University of Southern CaliforniaLos Angeles, CA

8:25 a.m. Questions and Answers

8:30 a.m. 24. Hepatitis C Therapeutic VaccinesMichael Houghton, Ph.D.Chiron CorporationEmeryville, CA

8:55 a.m. Questions and Answers

9:00 a.m. 25. Immunotherapy as a Treatment Possibility for Alzheimer’s DiseaseDale Schenk, Ph.D.Elan PharmaceuticalsSouth San Francisco, CA

9:25 a.m. Questions and Answers

FINAL PROGRAM

CME

Eighth Annual Conference

Page 23: On-Site Progr

on Vaccine Research

23

9:30 a.m. 26. Vaccines for Type 1 DiabetesRichard Insel, M.D.Juvenile Diabetes Research Foundation InternationalNew York, NY

9:55 a.m. Questions and Answers

10:00 a.m. Coffee Break/Exhibits Stadium Ballroom

Submitted Presentations 3: Vaccines for Epidemic Threats Grand Ballroom (Concurrent Session) Moderator: Susan J. Rehm, M.D. East/Salons A,B,C

National Foundation for Infectious Diseases

10:30 a.m. S13 Oculo-respiratory Syndrome (ORS) and Other Adverse Events Following Immunization (AEFIs) in Infants and Toddlers Given Influenza VaccineD. M. Skowronski1, S. A. Tweed1, V. Remple1, K. Pielak1, J. Daigneault2, P. Daly3, G. Arsenault4, E. Galanis1, T. Tam5; 1BC Centre for Disease Control, Vancouver, BC, CANADA, 2Direction de Sante Publique,Chicoutimi, PQ, CANADA, 3Vancouver Coastal Health Authority, Vancouver, BC, CANADA, 4Fraser Health Authority, Surrey, BC, CANADA, 5Public Health Agency of Canada, Ottawa, ON, CANADA.

10:45 a.m. S14 Improved Protective Antibody Responses Were Induced by Codon Optimized DNA Vaccines Expressing Hemagglutinin Antigens of Influenza H1 and H3 SerotypesS. Wang1, I. Mboujka1, J. Haran1, H. Cao1, X. Huang1, J. Taaffe2, A. Solórzano2, A. García-Sastre2, S. Lu1; 1Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 2Department of Microbiology, Mount Sinai School of Medicine, New York, NY.

11:00 a.m. S15 Protective Measures and Human Antibody Response to HPAI H7N3 in British Columbia (BC), CanadaD. M. Skowronski1, Y. Li2, S. A. Tweed1, T. Tam3, M. Petric1, S. Berger1, A. Larder4, N. Bastien2, A. King3, R. C. Brunham1; 1BC Centre for Disease Control, Vancouver, BC, CANADA, 2Public Health Agency of Canada, Winnipeg, MB, CANADA, 3Public Health Agency of Canada, Ottawa, ON, CANADA, 4Fraser Health, Abbotsford, BC, CANADA.

FINAL PROGRAM

CME

Page 24: On-Site Progr

24

WEDNESDAY, MAY 11, 2005 (CONTINUED)

11:15 a.m. S16 T Cell Multi-Epitope Vaccine for Pandemic InfluenzaJ. Alexander1, B. Stewart1, P. Bilsel1, J. Katz2, M. Newman1; 1Epimmune, San Diego, CA, 2Centers for Disease Control and Prevention, Atlanta, GA.

11:30 a.m. S17 Immunization with Salmonella enterica serovar Typhi Ty21a Expressing Anthrax Protective Antigen Protects Mice from an Anthrax Lethal Toxin ChallengeY. Wu, M. Osorio, S. Bhattacharyya, M. D. Bray, R. Walker, D. J. Kopecko;FDA-CBER, Bethesda, MD.

11:45 a.m. S18 Assessment of the Reactogenicity of Tdap in Children and Adolescents 7-19 Years of Age by Interval Since Prior Tetanus and Diphtheria Toxoids Containing VaccineS.A. Halperin1, L. Sweet2, D. Baxendale1, A. Neatby2, P. Rykers1, B. Smith1, M. Zelman2, D. Maus3, P. Lavigne3, M. Decker3

1Dalhousie University, Halifax, NS, CANADA, 2Department of Health and Social Services, Charlottetown, PE, CANADA, 3Sanofi Pasteur, Toronto ON, CANADA, and Swiftwater, PA, USA

12:00 p.m. Lunch (on your own)

Submitted Presentations 4: Innovations in Vaccine Design Grand Ballroom West/Salons D,E,F

(Concurrent Session) and Studies of Immune ResponseModerator: Connie Schmaljohn, Ph.D.

U.S. Army Medical Research Institute of Infectious Diseases

10:30 a.m. S19 Oral Vaccine Delivery by Salmonella Vaccine VectorsM. E. Gahan1, D. E. Webster1, S. L. Wesselingh1, B. B. Finlay2, R. A. Strugnell31Macfarlane Burnet Institute & Department of Medicine, Monash University, Melbourne, AUSTRALIA, 2Michael Smith Laboratories, University of British Columbia,Vancouver, BC, CANADA, 3Department of Microbiology and Immunology, University of Melbourne, Melbourne, AUSTRALIA.

10:45 a.m. S20 CpG Oligodeoxynucleotides Co-administered With the Microneme Protein MIC2 Protect Against Eimeria InfectionsR. A. Dalloul1, H. S. Lillehoj1, D. M. Klinman2, X. Ding1, W. Min1, R. A. Heckert1, E. P. Lillehoj31Animal Parasitic Diseases Laboratory, USDA, Beltsville, MD, 2Section of Retroviral Immunology, Center for Biologics Evaluation & Research, US FDA, Bethesda, MD, 3Department of Pediatrics, School of Medicine, University of Maryland, Baltimore, MD.

FINAL PROGRAM

CME

Eighth Annual Conference

Page 25: On-Site Progr

on Vaccine Research

25

11:00 a.m. S21 Immunogenicity of Combination DNA Vaccines for RVFV, TBEV, HTNV, and CCHFVK.W. Spik, A. Shurtleff, A.K. McElroy, M.C. Guttieri, J. W. Hooper, and C. SchmaljohnVirology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD

11:15 a.m. S22 Genetic Influence of HLA Haplotypes on Immune Responses Following Measles-Mumps-Rubella (MMR-II) Vaccination in Children I. G. Ovsyannikova1, S. V. Pankratz2, R. M. Jacobson1, R. A. Vierkant2, G. A. Poland1

1Vaccine Research Group, Mayo Clinic College of Medicine, Rochester, MN, 2Health Science Research, Mayo Clinic College of Medicine, Rochester, MN.

11:30 a.m. S23 Correlations among Measles Virus-Specific Antibody, Lymphoproliferation and Th1/Th2 Cytokine Responses Following MMR-II VaccinationN. Dhiman, I. G. Ovsyannikova, J. E. Ryan, R. M. Jacobson, R. A. Vierkant, S. V. Pankratz, S. J. Jacobsen, G. A. Poland Mayo Vaccine Research Group, Mayo Clinic College of Medicine, Rochester, MN

11:45 a.m. S24 Mumps Virus Vaccine Strain Urabe AM9: Identification of Nucleotide Changes Associated with Variability in the Neurovirulent PhenotypeC. J. Sauder, S. A. Rubin, K. M. Vandenburgh, K. M. Carbone; FDA/CBER, Bethesda, MD.

12:00 p.m. Lunch (on your own)

Symposium 7: Vaccinology Impact of Recent Advances Grand Ballroom

In Immunology Moderator: Paul-Henri Lambert, M.D.

Centre Medical Universitaire

1:00 p.m. 27. Limitations of B-cell Responses in Early LifeClaire-Anne Siegrist, M.D. University of GenevaGeneva, Switzerland

1:25 p.m. Questions and Answers

1:30 p.m. 28 Recent Advances in Studies of B and T cell Responses Early in LifeJames E. Crowe, Jr., M.D.Vanderbilt UniversityNashville, TN

1:55 p.m. Questions and Answers

FINAL PROGRAM

CME

Page 26: On-Site Progr

26

WEDNESDAY, MAY 11, 2005 (CONTINUED)

2:00 p.m. 29. Innate Imprinting and Inflammatory Lung DiseaseTracy Hussell, Ph.D.Imperial College LondonLondon, England

2:25 p.m. Questions and Answers

2:30 p.m. 30. Interrogating and Exploiting Memory B CellsAntonio Lanzavecchia, M.D.Institute for Research in Biomedicine.Bellinzona, Switzerland

2:55 p.m. Questions and Answers

3:00 p.m. Coffee Break/Exhibits Stadium Ballroom

Symposium 8: Multi-Agent and Chimeric Vaccines Grand BallroomModerator: Hana Golding, Ph.D.

Center for Biologics Evaluation and Research/FDA

3:30 p.m. 31. The Use of MVA as a Vaccine Delivery Vector to Elicit Protective Immune Responses Against PathogensHarriet L. Robinson, Ph.D. Emory UniversityAtlanta, GA

3:55 p.m. Questions and Answers

4:00 p.m. 32. Strategies for HIV Vaccine DevelopmentJohn W. Shiver, Ph.D.Merck and Company, Inc.West Point, PA

4:25 p.m. Questions and Answers

4:30 p.m. 33. Development of Novel Vaccine Candidates Using Directed Molecular EvolutionChristopher P. Locher, Ph.D.Maxygen, Inc.Redwood City, CA

4:55 p.m. Questions and Answers

FINAL PROGRAM

CME

Eighth Annual Conference

Page 27: On-Site Progr

on Vaccine Research

27

5:00 p.m. 34. ChimeriVaxTh: A Novel Platform for Genetically Engineered Live Viral VaccineThomas P. Monath, M.D.Acambis, Inc.Cambridge, MA

5:25 p.m. Questions and Answers

5:30 p.m. Adjournment/Participant Evaluation

Poster Session (Monday, May 9, 10:30 a.m. – 11:30 a.m., Stadium Ballroom; Posters will be on display throughout the day on Monday and will conclude with the Poster Reception at 5:30 p.m.)

Poster Group 1: Adjuvants and Immunomodulators

P1 Immune Responses Induced by Intranasal Immunization with Influenza H3N2-anti-H3N2 complex in MiceX. Yao, Y. Wen Fudan University, Shanghai, CHINA.

P2 Flagellin is an Effective Mucosal Adjuvant in the Development of a Protective Immune Response Against Yersinia pestisA. N. Honko, S. B. Mizel Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC.

P3 Induction of Active Immune Suppression by Co-immunization of DNA-protein VaccinesB. Wang, H. Jin, Y. KangState Key Lab for Agro-Biotechnology, China Agricultural University, Beijing, CHINA.

P4 Aluminum Phosphate is an Active Adjuvant for CRM197 Pneumococcal Conjugate Vaccine (PnC) in InfantsS. Lockhart1, W. Watson1, P. Fletcher2, A. Leeper3, S. Edwards4, M. McCaughey5, A. Dunning1, D. Sikkema1, G. Siber1

1Wyeth Research, Pearl River, NY, 2Woolwell Surgery, Plymouth, UNITED KINGDOM, 3Grove Surgery, Thetford, UNITED KINGDOM, 4North Cardiff Medical Centre, Cardiff, UNITED KINGDOM, 5Health Centre, Randalstown, UNITED KINGDOM.

P5 PyNTTTTGT Oligonucleotide IMT504 is a Potent Vaccine AdjuvantA. D. Montaner1, F. Elias2, J. M. Rodriguez2, J. Flo2, R. Lopez2, J. Zorzopulos1

1Instituto de Investigaciones Biomédicas, Fundación Pablo Cassará., Buenos Aires, ARGENTINA, 2Immunotech S.A., Buenos Aires, ARGENTINA.

FINAL PROGRAM

Page 28: On-Site Progr

28

POSTER SESSIONS (CONTINUED)

Poster Group 2: Antigen Presentation and Processing

P6 Novel Peptide Nanoparticles: A Platform for Vaccine Design D. Tropel1, A. C. Tissot2, C. Schellekens2, S. K. Raman1, A. Graff1, G. Machaidze1, M. F. Bachmann2, P. Burkhard3

1M.E Mueller Institute, Biozentrum, Basel, SWITZERLAND, 2Cytos Biotechnology, Schlieren, SWITZERLAND, 3The Institute of Materials Science, University of Connecticut, Storrs, CT

P7 Native Display of An HIV Tat Peptide on the Surface of Human Ferritin C. Li, E. Soistman, D. C. CarterNew Century Pharmaceuticals, Inc, Huntsville, AL.

Poster Group 3: Assessing Immunologic Response and Disease Protection

P8 Protection against hepatitis B carriage following infant vaccination may fall with ageM. E. Mendy, M. A. B. van der Sande, P. Waight, P. Rayco-Solon, P. Hutt, T. Fulford, C. Doherty, S. McConkey, D. Jefferies, A. Hall, H. WhittleViral Disease Programme, Medical Research Council, Banjul, GAMBIA.

P9 T cell responses to hepatitis B vaccine M. S. Hayney, N. A. WiegertUniversity of Wisconsin School of Pharmacy, Madison, WI.

P10 T cell responses following hepatitis A Immunizaton M. S. Hayney, N. A. Wiegert; University of Wisconsin School of Pharmacy, Madison, WI.

P11 Impact of a School Based Hepatitis B Immunization Program in a Low Endemic Area V. Gilca1, B. Duval2, N. Boulianne2, R. Dion2, G. D. Serres2

1Centre de recherche du Centre Hospitalier Universitaire de Québec, Quebec, PQ, CANADA, 2Institut National de Santé Publique du Québec, Quebec, PQ, CANADA, 2Laval University, Quebec, PQ, CANADA.

P12 Antibody Responses to Vaccinia Membrane Proteins Following Smallpox VaccinationS. Lawrence1, K. Lottenbach2, F. Newman2, M. Buller2, C. Bellone2, S. Koehm2, S. Stanley, Jr.1, R. Belshe2, S. Frey2

1Washington University, St. Louis, MO, 2Saint Louis University, St. Louis, MO

FINAL PROGRAM

Eighth Annual Conference

Page 29: On-Site Progr

on Vaccine Research

29

P13 Serum Neopterin for Early Assessment of Severity of Severe Acute Respiratory Syndrome J. W. Y. ChoiDepartment of Microbiology, The University of Hong Kong, Pokfulam, HONG KONG SPECIAL ADMINISTRATIVE REGION OF CHINA

P14 Determining Avidity for Anti-polysaccharide Antibodies S. Harris, J. Tam, P. Fernsten Vaccines Research, Wyeth, Pearl River, NY.

P15 Quantitation of Human Serum Immunoglobulin G Against O-Acetyl Positive and O-Acetyl Negative Serogroup W135 Meningococcal Capsular Polysaccharide P. C. Giardina1, E. Longworth2, R. Borrow2, P. Fernsten1

1Applied Immunology and Microbiology, Wyeth, Pearl River, NY, 2Meningococcal Reference Unit, Manchester Medical Microbiology Partnership, Health Protection Agency, Manchester, UNITED KINGDOM.

P16 Evaluating the Influences of Glycosylation on the Immunogenicity of the Ebola Virus Glycoprotein W. E. Dowling1, R. J. Hogan2, E. Thompson3, J. Paragas1, G. Bush1, J. Smith1, W. Capps1, L. Grey1, C. Badger1, C. S. Schmaljohn1

1Virology, USAMRIID, Fort Detrick, MD, 2Infectous Diseases, University of Georgia,Athens, GA, 3CBER, FDA, Rockville, MD.

Poster Group 4: Basic Science

P17 The Establishment of a Restriction Assorted Fragments Expression (RAFE) System for Vaccine Development of HIV-1B Subtype R. Shi1, W. Zheng2, J. Liu1, L. Li1, W. Ma1

1Institute of Molecular Biology, NanFang Medical University, Guangzhou, CHINA, 2South China Genomics Research Center, Guangzhou, CHINA.

P18 Toll-like Receptor 4 Plays Role in Activating Dendritic Cells by Necrotic Cells C. Kang, J. Choi, S. Lee, H. Moon, S. SeongMicrobiology and Immunoogy, Seoul National University College of Medicine, Seoul, REPUBLIC OF KOREA

P19 Construction of Recombinant BCG based HIV-1 Epitope Delivery System and Evaluation of its Immunogenicity in a Murine Model A. Vivekanandhan, N. Sujatha, P. NarayananImmunology, Tuberculosis Research Centre, Chennai, INDIA

FINAL PROGRAM

Page 30: On-Site Progr

30

POSTER SESSIONS (CONTINUED)

P20 Extending the Capsid Deletion Approach for Flavivirus Vaccine Development to the N-terminal Part of the ProteinR. M. Kofler, C. W. MandlClinical Institute of Virology, Medical University of Vienna, Vienna, AUSTRIA.

P21 Serotypes, Virulence Genes, and PFGE Patterns of Escherichia Coli Strains Isolated from Piglets with Diarrhea in SlovakiaH. Vu KhacDepartment of Bacteriology, Central Vietnam Veterinary Institute, Nha trang City, VIETNAM.

P22 Phase 2 & 3 Vaccine Research Agenda in the Kintampo District of Ghana.S. Owusu-Agyei Ministry of Health, Kintampo Health Research Centre, Kintampo, GHANA.

P23 A Multiplex Real-Time PCR Assay with An Internal Control for Quantitative Detection of Streptococcus pneumoniaeA. Hu, F. Li, P. Zhao, J. S. Tam, R. Rappaport, S. Cheng Wyeth, Pearl River, NY.

P24 Cloning and Characterization of the Polysaccharide Biosynthetic Genes for Shigelladysenteriae serotype 1 into Salmonella enterica serovar Typhi Ty21a D. Xu1, J. O. Cisar2, D. J. Kopecko1

1FDA-CBER, Bethesda, MD, 2NIH-NIDCR, Bethesda, MD.

Poster Group 5: Clinical Studies & Field Trials (e.g., Phase 1,2, or 3 studies)

P25 Pneumococcal Disease and Influenza Vaccination Acceptance Among Health Care WorkersJ. Wallenfels1, J. Rames2

1Ministry of Health of the Czech Republic, Praha, CZECH REPUBLIC, 2Institute of Hygiene and Epidemiology, First Faculty of Medicine, Charles University in Prague, Praha, CZECH REPUBLIC.

P26 Burden of Invasive Disease caused by Haemophilus influenzae Type b and Streptococcuspneumoniae Among Infants in Bamako, MaliS. O. Sow1, J. Campbell2, M. Tapia2, S. Diallo3, K. Kotloff2, M. Levine2

1Centre pour le Developpement des Vaccins, Bamako, MALI, 2University of Maryland School of Medicine, Center for Vaccine Development, Baltimore, MD, 3Clinical Bacteriology Laboratory and the Pediatric Service, Gabriel Touré Hospital, Bamako, MALI.

FINAL PROGRAM

Eighth Annual Conference

Page 31: On-Site Progr

on Vaccine Research

31

P27 Standardized Informed Consent for Low-literacy Audiences in African Research EnvironmentB. E. BekanProjet San Francisco, Kigali, RWANDA.

P28 Compatibility of Co-administered 7-valent Pneumococcal Conjugate, DTaP.IPV/PRP-T Hib and Hepatitis B Vaccines in InfantsD. W. Scheifele1, S. Halperin2, B. Smith2, K. Meloff3, D. Duarte-Monteiro3

1University of British Columbia, Vancouver, BC, CANADA, 2Dalhousie University, Halifax, NS, CANADA, 3Wyeth Pharmaceuticals, Toronto, ON, CANADA.

P29 Research Subject Satisfaction Assessment: A Missing Element of "Good Clinical Practices"C. LaJeunesse, A. Kallos, K. Marty, M. Mozel, D. W. ScheifeleVaccine Evaluation Center, University of British Columbia, Vancouver, BC, CANADA.

P30 Comparison of the Safety and Immunogenicity of Simultaneous or Sequential Administrationof an Adult Formulation Tdap Vaccine and Influenza VaccineS. McNeil1, F. Noya2, M. Dionne3, G. Predy4, W. Meekison5, C. Ojah6, S. Ferro7, E. Mills7, J. Langley1, S. Halperin1

1Dalhousie University, Halifax, NS, CANADA, 2Montreal Children's Hospital, Montreal, PQ, CANADA, 3INSPQ, Quebec City, PQ, CANADA, 4Capital-Health, Edmonton, AB, CANADA, 5Westcoast Clinical Research, Vancouver, BC, CANADA, 6Saint John Regional Hospital, Saint John, NB, CANADA, 7Sanofi-Pasteur, Toronto, ON, CANADA.

P31 Measurement of Tetanus Antitoxin in Oral Fluid: A Novel Method to Evaluate Vaccination ProgramsM. D. Tapia1, L. Cuberos1, S. O. Sow2, M. N. Doumbia2, M. Bagayogo2, M. Pasetti1, K. Kotloff1, M. Levine1

1University of Maryland School of Medicine, Baltimore, MD, 2Centre pour le Developpement des Vaccins - Mali, Bamako, MALI.

P32 Studies on a Live Oral Attenuated Cholera Vaccine, Peru-15 in BangladeshF. Qadri1, M. I. Chowdhury1, M. A. Salam1, S. M. Faruque1, T. Ahmed1, Y. A. Begum1, A. Saha1, L. V. Seidlein2, R. F. Breiman1, J. J. Mekalanos3, J. D. Clemens2, K. P. Killeen4, D. A. Sack1

1ICDDR,B: Centre for Health and Population Research, Dhaka, BANGLADESH, 2International Vaccine Institute, Seoul, DEMOCRATIC PEOPLE'S REPUBLIC OF KOREA, 3Harvard Medical School, Boston, MA, 4AVANT Immunotherapeutics, Needham, MA.

FINAL PROGRAM

Page 32: On-Site Progr

32

POSTER SESSIONS (CONTINUED)

Poster Group 6: Combination Vaccines

P33 A Novel Combination DNA and Inactivated Rabies Virus VaccineP. N. Rangarajan1, V. A. Srinivasan2

1Indian Institute of Science, Bangalore, INDIA, 2Indian Immunologicals Limited, Hyderabad, INDIA.

P34 Minicircle DNA Immobilized in Bacterial Ghost: A Novel Non-living Bacterial DNA-vaccine Carrier SystemC. Azimpour Tabrizi1, W. Jechlinger2, P. Becker3, T. Ebensen3, C. Guzman3, W. Lubitz1

1Microbiology and Genetic, Vienna, AUSTRIA, 2Inst. Bacteriology, Mycology and Hygiene, Vienna, AUSTRIA, 3German Research Centre for Biotechnology, Braunschweig, GERMANY.

P35 Multiple DNA Vaccine Plasmids Protect Mice from Acute Pulmonary Infection of Pseudomonas aeruginosaS. Saha1, F. Takeshita1, S. Sasaki1, T. Matsuda1, T. Matsumoto2, K. Yamaguchi2, K. Okuda1

1Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 2Toho University School of Medicine, Tokyo, JAPAN.

P36 Infanrix™-IPV-Hib (GSK) is Safe and Immunogenic Compared to Pentacel™ (Sanofi Pasteur) As a 4th Dose in 15-20 Month OldsS. A. Halperin1, B. Tapiero2, B. Law3, B. Duval4, F. Diaz-Mitoma5, D. Elrick6

1Pediatrics, Dalhousie University, Halifax, NS, CANADA, 2Pediatrics, Ste Justine Hospital, University of Montreal, Montreal, PQ, CANADA, 3Pediatrics, University of Manitoba, Winnipeg, MB, CANADA, 4Institut National de Santé Publique, Quebec City, PQ, CANADA, 5Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, CANADA, 6GlaxoSmithKline, Inc, Mississauga, ON, CANADA.

Poster Group 7: New Vaccines for Common Pathogens

P37 Immunization with Recombinant Adenovirus Synthesizing Secretory form of Japanese Encephalitis Virus Envelope Protein Protects Adenovirus-Exposed Mice Against Lethal EncephalitisS. VratiNational Institute of Immunology, New delhi, INDIA.

FINAL PROGRAM

Eighth Annual Conference

Page 33: On-Site Progr

on Vaccine Research

33

P38 A Novel Live Adenovirus Vaccine Vector Prototype: High-level Antigen Production from the Adenoviral Major Late Transcriptional UnitM. G. Berg1, B. Falgout2, G. W. Ketner1

1Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, 2CBER, Food & Drug Administration, Bethesda, MD.

P39 Construction of Live Attenuated Shigella Vaccine Candidates using RecombineeringR. Ranallo, S. Barnoy, S. Thakkar, M. Venkatesan Enteric Infections, WRAIR, Silver Spring, MD.

P40 Oral Immunization of Dogs with Baits Containing the Recombinant Rabies Virus Glycoprotein/Nucleoprotein-Canine Adenovirus Type 2R. Hu, S. Zhang, H. liVeterinary Institute, Academy of Military Medical Science, Changchun, CHINA.

P41 Immunological Properties of Conjugates Prepared from Pathogenic Candida Surface Antigens: Potential VaccinesS. BystrickyInstitute of Chemistry SAV, Bratislava, SLOVAKIA.

P42 A Novel Subunit Vaccine Protects Mice Against Systemic Disease and Intestinal Colonization by Salmonella entericaL. Wonderling1, D. Straub2, D. Emery2

1Syntiron, Saint Paul, MN, 2Epitopix, Willmar, MN.

P43 Partial Protection of Mice after DNA Vaccination Against Staphylococcus aureus Infection.M. C. Gaudreau1, P. Lacasse2, B. G. Talbot1

1Biologie, University of Sherbrooke, Sherbrooke, PQ, CANADA, 2Agriculture and Agri-Food Canada, Lennoxville, PQ, CANADA.

Poster Group 8: Novel Antigens

P44 Strong B- and T-cell Response after Protein, DNA, and DNA Prime/protein Boost Immunisation with HBV Cores Carrying HBV PreS1 SequencesD. Skrastina, I. SominskayaProtein Engineering, Biomedical Research and Study Centre, University of Latvia, Riga, LATVIA.

P45 Genetic Polymorphism and Positive Selection in a ‘Concealed’ Gut Potential Vaccine Antigen from Rhipicephalus appendiculatus.L. M. Kamau1, R. Skilton2, T. Musoke2, D. Wasawo2, J. Rowlands2, R. Bishop2

1Kenyatta University & International Livestock Research Institute (ILRI), Nairobi, KENYA, 2International Livestock Research Institute (ILRI), Nairobi, KENYA.

FINAL PROGRAM

Page 34: On-Site Progr

34

POSTER SESSIONS (CONTINUED)

P46 A Recombinant 63-kilodalton Form of Bacillus anthracis Protective Antigen Produced in the Yeast Saccharomyces cerevisiae Provides Protection in Two Inhalational Challenge Models of Anthrax Infection.R. W. Hepler1, R. Kelly1, T. B. McNeely1, H. Fan1, M. C. Losada1, H. A. George1, A. Woods1, L. D. Cope1, A. Bansal1, J. C. Cook1, G. Zang1, S. L. Cohen1, X. Wei1, P. M. Keller1, E. K. Leffel2, J. G. Joyce1, M. L. M. Pitt2, L. D. Schultz1, K. U. Jansen1, M. Kurtz1

1Merck Research Labs, West Point, PA, 2US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD.

P47 A Recombinant Leishmania Antigen Related to the Silent Information Regulatory 2 (SIR2) Protein Family Induces a B cell Activation and Antibody Specific Immune ResponseR. Silvestre1, A. Cordeiro-da-Silva1, A. Ouaissi21Faculdade Farmácia and I.B.M.C. of Universidade Porto, Porto, PORTUGAL, 2Institut de Recherche pour le Développement, UR008, Montpellier, FRANCE.

P48 DNA Vaccine Expressing D8L of Vaccinia Virus Enhanced the Efficacy of Multi-gene Smallpox DNA Vaccine FormulationsP. V. Sakhatskyy, S. Wang, T. Chou, S. Lu University of Massachusetts Medical School, Worcester, MA.

Poster Group 9: Preclinical Studies (e.g. laboratory animal)

P49 An Intranasal, Protollin™-RSV Subunit Vaccine Induces Mucosal IgA, Serum Neutralizing Antibodies and a Type-1 Cytokine-biased Response by Spleen and Lung Mononuclear Cells in Mice.S. L. Cyr1, T. Jones2, I. Stoica-Popescu2, S. Chabot1, D. S. Burt2, B. J. Ward1

1Microbiology and Immunology, McGill University, Montreal, PQ, CANADA, 2ID Biomedical, Laval, PQ, CANADA.

P50 Prevention of Serogroup A, C and W135 Meningococcal Disease in the Meningitis Belt of Africa by Targeting Outer Membrane ProteinsE. Rosenqvist, G. Norheim, E. Fritzsønn, T. Tangen, P. Kristiansen, D. A. Caugant, A. Aase, E. A. Høiby, I. S. AabergeDivision for Infectious Disease Control, Norwegian Institute of Public Health, Oslo, NORWAY.

FINAL PROGRAM

Eighth Annual Conference

Page 35: On-Site Progr

on Vaccine Research

35

P51 Development and Use of the Outer Membrane Vesicle Concept for Vaccines Against Meningococcal Group B Disease E. Rosenqvist1, G. Norheim1, L. Meyer Næss1, P. Kristiansen1, P. Costantino2, E. Wedege1, D. A. Caugant1, B. Feiring1, I. S. Aaberge1, R. Rappuoli3, J. Holst1

1Division for Infectious Disease Control, Norwegian Insitute of Public Health, Oslo, NORWAY, 2Chiron Vaccines S.r.l., Siena, ITALY, 3Chiron Vaccine S.r.l., Siena, ITALY.

P52 Reducing the Cost of Manufacturing Conjugate Vaccines: Effective Alternatives to Gel FiltrationA. Lees, D. E. ShaferBiosynexus Inc, Gaithersburg, MD.

P53 Aminooxy Reagents and Oxime Chemistry for the Preparation of Conjugate Vaccines A. Lees, A. LopezAcostaBiosynexus Inc, Gaithersburg, MD.

P54 Development of HIV-1 Subtype C Vaccine CandidatesP. SethMicrobiology, All India Institute of Medical Sciences, New Delhi, INDIA.

P55 Enhancement of Cell-mediated Immunity in Mice by the Model of a Whole HIV-1 Gag in Live Mycobacterium bovis BCG.D. PromkhatkaewDepartment Of Medical Sciences, Nonthaburi, THAILAND.

P56 Preclinical Development of Yeast-Based Immunotherapy for Chronic Hepatitis C Virus InfectionA. A. Haller, T. King, Y. Lu, C. Kemmler, D. Bellgrau, G. Gordon, D. Apelian, A. Franzusoff, T. C. Rodell, R. C. DukeGlobeImmune Inc., Aurora, CO.

Poster Group 10: Product Development

P57 Analytical Challenges for Novel Vaccine FormulationsE. Hartwell, S. D. Sen, B. J. Roser Cambridge Biostability Ltd, Cambridge, UNITED KINGDOM.

P58 Production and Control of a Brazilian Meningococcal C Conjugate VaccineI. A. SilveriaDepartamento de Desenvolvimento Tecnológico, Fundação Oswaldo Cruz / Biomanguinhos,Rio de Janeiro, BRAZIL.

FINAL PROGRAM

Page 36: On-Site Progr

36

POSTER SESSIONS (CONTINUED)

P59 Production and Control of a Brazilian Meningococcal B VaccineI. A. Silveria Departamento de Desenvolvimento Tecnológico, Fundação Oswaldo Cruz / Biomanguinhos,Rio de Janeiro, BRAZIL.

Poster Group 11: Routes of Antigen Administration and Vaccine Safety

P60 Needle-Free Delivery of Antigens to Ultrasound Pre-treated Skin Demonstrated in a Feasibility Clinical TrialD. H. Libraty1, S. P. Barman2

1Center for Infectious Diseases & Vaccine Research, University of Massachusetts Medical School, Worcester, MA, 2Transdermal Drug Delivery , Vaccines, Sontra Medical, Franklin, MA.

P61 Anaphylaxis Following Recombinant Hepatitis B Vaccines in Yeast-Sensitive Individuals: Reports to VAERSL. DiMiceli1, V. Pool1, S. V. Shadomy2, J. Iskander1

1CDC/National Immunization Program, Atlanta, GA, 2CDC/National Center for Infectious Diseases, Atlanta, GA.

P62 Histopathology Analysis of Local Reactions in Mice following Injection of Diphtheria-Tetanus-Acellular Pertussis (DTaP) vaccines A. Honjo, T. Katsuta, S. Tateyama, C. Nagaoka, T. Tokutake, Y. Arimoto, N. Nakajima, T. Goshima, T. Kato St.Mariannna University School of Medicine, Kawasaki-si, JAPAN.

Poster Group 12: Vaccines for New and Reemerging Pathogens and Vaccines for Tropical and/or Exotic Pathogens

P63 CAP adsorbed rPA Nasal Vaccine as an Approach to Mucosal Immune Protection Against Anthrax InfectionP. Nagappan1, J. Arroyo2, T. Morcol1, A. R. Mitchell1, L. Nerenbaum1, S. Billingsley1, S. J. D. Bell11BioSante Pharmaceuticals, Inc., Smyrna, GA, 2DVC LLC a CSC Company, Frederick, MD.

P64 Influenza Nucleoprotein Conjugated to Immunostimulatory DNA as a Potential Vaccine Against Pandemic InfluenzaT. dela Cruz1, D. Higgins1, G. Ott1, I. Mbawuike2, S. Tuck1, G. Van Nest1

1Dynavax Technologies, Berkeley, CA, 2Baylor College of Medicine, Houston, TX.

FINAL PROGRAM

Eighth Annual Conference

Page 37: On-Site Progr

on Vaccine Research

37

P65 Development of SARS Vaccine Using Recombinant Vaccinia Virus Derived from LC16m8M. Kitabatake1, F. Yasui2, S. Inoue3, K. Morita3, F. Murai4, M. Kidokoro5, K. Mizuno6, H. Shida7, K. Matsushima1, M. Kohara2

1Univ. Tokyo, Tokyo, JAPAN, 2The Tokyo Metro. Inst. Med. Sci., Tokyo, JAPAN, 3Inst. Tropical Med., Nagasaki Univ., Nagasaki, JAPAN, 4Post Genome Inst., Tokyo, JAPAN, 5Natl. Inst. Infect. Dis., Tokyo, JAPAN, 6The Chemo-Sero-Therapeutic Res. Inst., Kumamoto, JAPAN, 7Inst. Gen. Med., Hokkaido Univ., Sapporo, JAPAN.

P66 A Novel Subunit Vaccine Protects Mice Against Yersinia InfectionL. Wonderling1, D. Straub2, D. Emery2

1Syntiron, Saint Paul, MN, 2Epitopix, Willmar, MN.

P67 Cysteine Proteinases Based Vaccines for L. major and L. infantum InfectionsS. Rafati, T. Taheri, A. Zadeh Vakili, A. Nakhaee, F. Zahedifard, Y. Taslimi, F. Doustdari Immunology, Pasteur Institute of Iran, Tehran, IRAN (ISLAMIC REPUBLIC OF).

P68 Development of Attenuated Mutants as Potential Vaccine Candidates for Visceral leishmaniasisP. Salotra1, G. Sreenivas2, R. Singh1, A. Selvapandiyan3, R. Duncan3, H. L. Nakhasi31Institute of Pathology (ICMR), New Delhi, INDIA, 2Insitute of Pathology (ICMR), New Delhi, INDIA, 3CBER, FDA, Bethesda, MD.

P69 Protection Studies with Candidate Schistosoma mansoni DNAVaccinesA. M. Karim1, N. El-Ghazali1, A. Medhat1, S. F. Ibrahim2

1Ain Shams Univ. Fac. of Science, Cairo, EGYPT, 2Cairo Univ. Fac. of Science, Cairo, EGYPT.

P70 Protective Immunity of Single and Multiple Recombinant DNA or Protein Vaccines Against Lymphatic Filariasis.P. Kaliraj, Sr.1, S. Anand1, V. Murugan1, K. Kirithika2, M. Reddy2

1Anna University, Chennai, INDIA, 2MGIMS, Sevagram, INDIA.

Poster Group 13: Veterinary Vaccines

P71 Enhancing Brucellosis Vaccines, Vaccine Delivery Systems and Surveillance Diagnostics for Bison and Elk in the Greater Yellowstone AreaG. E. Plumb, Jr.1, B. Marsh2

1Yellowstone National Park, US National Park Service, Yellowstone National Park, WY, 2Board of Animal Health, State of Indiana, Indianapolis, IN.

FINAL PROGRAM

Page 38: On-Site Progr

38

POSTER SESSIONS (CONTINUED)

P72 Canine Herpesvirus Bacterial Artificial Chromosome Technology For Antifertility Vaccination of Foxes in AustraliaT. Strive, J. Wright, N. French, C. M. Hardy, G. H. ReubelSustainable Ecosystems, CSIRO, Canberra ACT, AUSTRALIA.

P73 Molecular Cloning and Sequence Analysis of Bm86 cDNA from a Thai Strain of the Cattle Tick, Boophilus microplusS. Jittapalapong1, S. Thanasilp1, T. Sirinarukmitr2, K. Kaewmongkol3, R. W. Stich4

1Veterinary Parasitology, Kasetsart University, Bangkok, THAILAND, 2Veterinary Pathology, Kasetsart University, Bangkok, THAILAND, 3Veterinary Companion Animal Medicine, Kasetsart University, Bangkok, THAILAND, 4Veterinary Preventive Medicine, The Ohio State University, Columbus, OH.

P74 Intranasal Vaccination of Mares to Protect Against Streptococcal Uterine InfectionsR. C. Causey Animal and Veterinary Sciences, University of Maine, Orono, ME.

P75 CpG Oligodeoxynucleotides Upregulate Antibacterial Systems and Induce Early, Non-specific Antiviral Protection in Fish.A. C. Carrington1, B. Collet2, C. J. Secombes1

1Department of Zoology, University of Aberdeen, Aberdeen, UNITED KINGDOM, 2Fisheries Research Services, Marine Laboratory, Aberdeen, UNITED KINGDOM.

P76 Summary of the Research of Recombinant HEV VaccineJ. LinLanzhou Institute of Biological Products, Gansu, CHINA

FINAL PROGRAM

Eighth Annual Conference

Page 39: On-Site Progr

on Vaccine Research

39

MEET THEEXPERT

PRESENTERS

Page 40: On-Site Progr

40

W. Ripley Ballou, M.D.Meet the Experts Breakfast SessionTuesday, May 10, 20057:00 am – 7:45 am

Dr. Ballou is Vice President, Early Development,GlaxoSmithKline Biologicals in Rixensart Belgium. Heis responsible for the clinical development of vaccinesagainst malaria, tuberculosis, HIV, as well as other earlydevelopment projects. Prior to his position at GSK, Dr.Ballou was Vice President, Vaccines and InfectiousDiseases at MedImmune, Inc. From 1984 to 1999, Dr.Ballou was an Infectious Disease Officer with the USArmy where his research focused on the development ofmalaria vaccines. From 1991 until his retirement fromthe military in 1999, he served as Chief, Department ofImmunology, Walter Reed Army Institute of Researchand was responsible for the Army’s malaria vaccinedevelopment program.

Dr. Ballou received his bachelor’s degree from theGeorgia Institute of Technology, Atlanta, Georgia, andhis medical degree from Emory University. He receivedhis internal medicine and infectious diseases training atthe Walter Reed Army Medical Center.

Dr. Ballou has authored and/or coauthored morethan 135 research papers and book chapters oninfectious disease topics. In addition, as an editorialboard member, he has reviewed several scientificjournals and numerous proceedings.

Keith P. Klugman, M.D., Ph.D.Meet the Experts Breakfast SessionTuesday, May 10, 20057:00 am – 7:45 am

Keith Klugman MB BCh, PhD, FRCPath (Lond) iscurrently Professor of Global Health at the RollinsSchool of Public Health at Emory University, in Atlanta, GA, USA. He is also Professor of Medicine inthe Division of Infectious Diseases of the School ofMedicine at that University and a Visiting Researcher in the Respiratory Diseases Branch of the Centers for

Disease Control and Prevention (CDC). He is also theDirector of the Respiratory and Meningeal PathogensResearch Unit at the University of the Witwatersrand inJohannesburg, South Africa.

He was previously the Director of the South AfricanInstitute for Medical Research in South Africa. He didhis undergraduate training and specialization in SouthAfrica, and his post doctoral research at RockefellerUniversity in New York.

Professor Klugman has served as a member ofnumerous international committees including those of the World Health Organization in Geneva and theInstitute of Medicine in Washington, DC. He currentlychairs the Wellcome Trust Tropical Interview Panel inLondon. He serves as an editor or member of theeditorial board of 9 international journals on medicine,infectious diseases and antimicrobial research.

Professor Klugman’s research interests are indeterminants of antimicrobial resistance, the clinicalrelevance of resistance and the development of vaccinesfor bacterial pathogens – particularly the pneumococcus.He has published more than 300 papers in the scientificliterature to date.

Karen L. Kotloff, M.D.Meet the Experts Breakfast SessionTuesday, May 10, 20057:00 am – 7:45 am

Dr. Kotloff is Professor of Pediatrics and Medicine atthe University of Maryland School of Medicine andChief of the Community Studies Section of the Centerfor Vaccine Development. She received her bachelor’sdegree from Washington University in St. Louis,Missouri and her medical degree from Temple Universityin Philadelphia, Pennsylvania. She completed herresidency in Pediatrics at the Children’s Hospital ofPittsburgh and her fellowship in Infectious Diseases atthe University of Maryland.

Dr. Kotloff has 18 years experience conductingepidemiologic research in infectious diseases and Phase I,II, and III clinical vaccine trials involving adults andchildren. She has particular interest in vaccines against

MEET THE EXPERT PRESENTERS

Eighth Annual Conference

Page 41: On-Site Progr

enteric infections, including Shigella, C. difficile, andHelicobacter pytori, among others. Recently, Dr. Kotloffhelped to establish a field site for evaluating vaccines andvaccine preventable diseases in Mali, West Africa.

Dr. Kotloff has served as a consultant for the NationalInstitutes of Health, the International Vaccine Institute,and the World Health Organization. She has authoredor co-authored more than 75 research papers and bookchapter on infectious disease topics.

Stanley A. Plotkin, M.D.Meet the Experts Breakfast SessionTuesday, May 10, 20057:00 am – 7:45 am

Dr. Stanley A. Plotkin is Emeritus Professor of theUniversity of Pennsylvania and Executive Advisor toSanofi Pasteur. Until 1991, he was Professor of Pediatricsand Microbiology at the University of Pennsylvania, andProfessor of Virology at the Wistar Institute and at thesame time, Director of Infectious Diseases and SeniorPhysician at the Children’s Hospital of Philadelphia. In1991, Dr. Plotkin left the University to join the vaccinemanufacturer, Pasteur-Mérieux-Connaught, where forseven years he was Medical and Scientific Director, based at Marnes-la-Coquette, outside Paris. The samecompany is now named Sanofi Pasteur.

Dr. Plotkin’s career included internship at ClevelandMetropolitan General Hospital, residency in pediatricsat the Children’s Hospital of Philadelphia and theHospital for Sick Children in London and three years in the Epidemic Intelligence Service of the Centers forDisease Control of the US Public Health Service.

He has been chairman of the Infectious DiseasesCommittee and the AIDS Task Force of the AmericanAcademy of Pediatrics, liaison member of the AdvisoryCommittee on Immunization Practices and Chairman of the Microbiology and Infectious Diseases ResearchCommittee of the National Institutes of Health. Dr. Plotkin received the Bruce Medal in PreventiveMedicine of the American College of Physicians, theDistinguished Physician Award of the PediatricInfectious Diseases Society and the Clinical Virology

on Vaccine Research

41

Award of the Pan American Society for Clinical Virology.In June 1998, he received the French Legion of HonorMedal, in June 2001, the Distinguished Alumnus Awardof the Children’s Hospital of Philadelphia, the SabinGold Medal in May 2002, and in September 2004 theBristol Award of the Infectious Diseases Society ofAmerica. A lecture in his honor has been established atthe Pediatric Academic Societies annual meeting. Hisbibliography includes over 600 articles and he has editedseveral books including the standard textbook onvaccines. He developed the rubella vaccine now instandard use, and has worked extensively on thedevelopment and application of other vaccines includingpolio, rabies, varicella, rotavirus and cytomegalovirus.

Gregory A. Poland, M.D.Meet the Experts Breakfast SessionTuesday, May 10, 20057:00 am – 7:45 am

Dr. Gregory Poland is the Director of the MayoClinic's Vaccine Research Group – a state-of-the-artresearch group and laboratory that investigates issuessurrounding vaccine response and novel vaccinesimportant to public health. Dr. Poland is a Professor of Medicine and Infectious Diseases and MolecularPharmacology and Experimental Therapeutics, theAssociate Chair for Research for the Department ofMedicine, the Director of the Immunization Clinic and the Director of the Program in TranslationalImmunovirology and Biodefense at the Mayo Clinic.He also serves as the President of the InternationalSociety for Vaccines and the American Editor for theJournal Vaccine.

In March 2005, Dr. Poland was elected as thePresident of the Armed Forces Epidemiological Board.He was appointed as the Mary Lowell Leary Professor inMedicine (the highest academic distinction for a facultymember) by Mayo Clinic’s Board of Trustees in February 2004, and in May 2003, he was awarded theSecretary of Defense Medal for Outstanding PublicService. In 1998, he received a joint award from theCenters for Disease Control and Prevention and the

MEET THE EXPERT PRESENTERS

Page 42: On-Site Progr

42

antigens of unrelated pathogens and deliver them to thehuman immune system. His clinical research hasinvolved studies of pathogenesis and the assessment of avariety of vaccines in adults and children in Baltimore,as well as in many developing countries. He has been a pioneer in carrying out clinical trials in developingcountries, including studies of vaccines developed at the CVD. He designed, arranged and supervised theperformance of several large-scale, randomized,controlled field trials investigating the efficacy of liveoral typhoid vaccines (which led to licensure of Ty21a by the FDA) and a vaccine to prevent invasive diseasecaused by Haemophilus influenzae type b.

Dr. Levine received his B.S. degree from City Collegeof New York, his Medical degree from the MedicalCollege of Virginia, and his D.T.P.H. diploma (withDistinction) from the London School of Hygiene andTropical Medicine.

Dr. Levine currently sits on the editorial board offour major research journals, is a consultant to manyorganizations including the World Health Organization,NIH, Institute of Medicine and the Department ofDefense. He holds memberships in numerous societiesincluding the Institute of Medicine of the NationalAcademy of Science, the Association of AmericanPhysicians, the American Society of ClinicalInvestigation and the Interurban Clinical Club. He ispast President of the American Epidemiological Societyand most recently was voted as President-elect of theAmerican Society of Tropical Medicine and Hygiene.He is the recipient of many honors, two of which are theAlbert B. Sabin Gold Medal Award for lifetimeachievement in the area of vaccine development andimplementation and he was voted by the editors ofBaltimore Magazine as “Baltimorean of the Year.” Thishonor was bestowed upon ten individuals in recognitionof their contribution to improving the way of life inBaltimore in 2001.

Dr. Levine has authored and co-authored 445scientific articles, 94 chapters, and is senior editor of“New Generation Vaccines,” the third edition premiertext on the discipline of modern vaccinology publishedin January 2004.

Health Care Financing Administration for hiscontribution to increasing adult immunization rates inthe US which was awarded by the Surgeon General ofthe United States. Also of major significance, in 1997,he was honored as the "Outstanding ClinicalInvestigator of the Year" by the Mayo Clinic.

Additionally, Dr. Poland participates on manynational and academic review committees and reviewsjournal articles for over 26 different publications such asThe Lancet, Annals of Internal Medicine and New EnglandJournal of Medicine. A prolific writer, Dr. Poland haspublished over 160 peer-reviewed scientific articles andbook chapters.

Dr. Poland received his medical degree from theSouthern Illinois University School of Medicine inSpringfield, Illinois, and completed his residency andadvanced post-graduate work at the University ofMinnesota/Abbott-Northwestern Hospital,Minneapolis, MN.

Myron M. Levine, M.D., D.T.P.H.Meet the Experts Breakfast SessionWednesday, May 11, 20057:00 am – 7:45 am

Myron M. Levine, M.D., D.T.P.H, Director of the internationally recognized Center for VaccineDevelopment at the University of Maryland School ofMedicine, holds faculty appointments as Professor infour departments at the University and is Chief of twodivisions. Dr. Levine has been Director of the Centerfor Vaccine Development since its inception, and hascreated therein an environment that is unusually rich in intellectual ferment and stimulation.

He is one of the most vocal advocates of mucosalimmunization, i.e., the administration of vaccines byoral and intranasal routes to avoid the unpleasantnessand occasional dangers of parenteral injections. Dr. Levine has made substantial contributions in basicvaccinology and clinical research. In recent years, hisbasic laboratory research has focused on the use ofattenuated Salmonella typhi as live oral typhoid vaccinesand as live vector vaccines that express the protective

MEET THE EXPERT PRESENTERS

Eighth Annual Conference

Page 43: On-Site Progr

Peter L. Nara, D.V.M., Ph.D.Meet the Experts Breakfast SessionWednesday, May 11, 20057:00 am – 7:45 am

Dr. Nara is currently Chief Executive Officer,President, Chairman & co-founder of BiologicalMimetics, Inc., a biotechnology company founded to translate the theory of “Deceptive Imprinting” andthe “Immune Refocusing” Technology, discovered by Dr. Nara and colleagues, into preventative vaccines,novel immuno-therapeutics and diagnostics toameliorate disease worldwide. Prior to this position, Dr. Nara was formerly the Section Chief of the VaccineResistant Diseases Section, Division of Basic Sciences, at the National Cancer Institute, National Institutes ofHealth.

Dr Nara’s scientific contributions include over 20book chapters and 160 scientifically peer-reviewedpublications. Some highlights include: his laboratory at the National Cancer Institute was designated both aNational and International AIDS Reference Laboratoryby the National Institute’s of Allergy and InfectiousDiseases and World Health Organization (WHO),Global Program on AIDS; he initiated a Internationaleffort which characterized over 120 HIV-1 isolates forfuture vaccine and clinical trial site development fromepidemic centers around the world, was made the HIV-1Immunotyping Group leader and chairman for theWHO Network for HIV Isolation and Characterization.He serves on numerous scientific advisory boards ofcorporate, private foundations and academic initiativesin the area of national biodefense, comparative medicineand immunology, vaccine discovery and immuno-therapeutics. He was a Howard Hughes, Ciba Corning,and Miles Invited Lecturer, nominated by Dr. JonasSalk; elected, and served as Founding President of theInternational Society for Vaccines 1994-2002; is one ofthe founding co-chairs for the Annual Conference onVaccine Research; served on the editorial boards of theleading virology, AIDS, vaccine, immunology journals;and is the inventor or co-inventor on 7 issued U.S.patents. This past year he was nominated asBiotechnology Executive of the Year in Frederick,County, Maryland.

on Vaccine Research

43

Dr. Nara’s formal education and training include his Bachelors of Science with Honors in 1977 fromColorado State University; his Masters of Science, cumlaude in Immuno-pharmacology in 1979 from The OhioState University; his combined Doctor of VeterinaryMedicine in 1984 and Doctorate of Philosophy inRetrovirology and Immunology in 1986 from The OhioState University. His post-doctoral training involved adual 4-year combined National Institutes of Health(NIH) medical post-doctoral fellowship/VeterinaryMedical Officer at the National Cancer Institute, and a4-year Comparative Pathology Residency at the ArmedForces Institute of Pathology, Washington D.C. (1986-1990). He was acting Head of the Virus Biology Sectionin the Office of the Institute’s Director from 1990-1992and was tenured and promoted to and remained SectionChief of the Virus Resistant Diseases Section from 1993-1998.

George R. Siber, M.D.Meet the Experts Breakfast SessionWednesday, May 11, 20057:00 am – 7:45 am

Dr. Siber joined Wyeth Lederle Vaccines as VicePresident and Chief Scientific Officer in August 1996.He was named Senior Vice President in August 1999and Executive Vice President in June 2002. In thiscapacity Dr. Siber is responsible for discovery research in bacterial vaccines, viral vaccines, immunology andgenetic vaccines, process and analytical development,clinical development, and scientific affairs for WyethVaccines Research. While at Wyeth Dr. Siber hasoverseen the development and approval of an acellularpertussis vaccine for infants (Acel-Imune), a vaccine toprevent Rotavirus diarrhea in infants (RotaShield), a glycoconjugate vaccine to prevent group Cmeningococcal meningitis (Meningitec), a 7 componentglycoconjugate vaccine to prevent pneumococcal diseasein infants and children (Prevnar), and a cold adaptednasally administered influenza vaccine in collaborationwith MedImmune (FluMist).

Prior to joining Wyeth Dr. Siber was Director of the Massachusetts Public Health Biologic Laboratories

MEET THE EXPERT PRESENTERS

Page 44: On-Site Progr

and Associate Professor of Medicine with the HarvardMedical School, Dana Farber Cancer Institute. During this time Dr. Siber oversaw research on acellularpertussis and Hemophilus influenza vaccines, thedevelopment and approval of CMV Immune Globulin(Cytogam®) and RSV Immune Globulin (Respigam®)and the production of DTP vaccines and immuneglobulins for the State of Massachusetts.

Dr. Siber’s research interests have included theevaluation of the human immune response topolysaccharide and protein antigens, the development of vaccines and immune globulins against Hib,pneumococci, meningococci, pertussis and RSV andmaternal immunization to prevent perinatal and earlyneonatal infections. Dr. Siber has authored more than150 scientific articles in peer-reviewed journals. Dr.Siber holds 3 issued patents which support a licenseddiagnostic test for meningitis (Bactigen®) and anantibody based preventative for respiratory syncytialvirus infections in high-risk children (Respigam®).

Dr. Siber has served on numerous advisorycommittees including the WHO/UNDP SteeringCommittee for Encapsulated Bacterial Vaccines, theSteering Committee for Development of StreptococcusPneumonia Vaccine for the Pan American HealthOrganization, the Institute of Medicine Committee on the Children’s Vaccine Initiative, the NIH BlueRibbon Panel for Bioterrorism and its Implications forBiomedical Research, Chairman of the review of the US Army’s HIV research program, and the Board ofScientific Counselors for the National Vaccine Center.

Dr. Siber was the recipient of the Canadian MedicalResearch Council Fellowship and is currently a fellow of the Infectious Diseases Society of America and thePediatric Infectious Disease Society. Dr. Siber received a B.Sc. degree at Bishop’s University and a MDCMdegree from McGill University both in Quebec, Canada. Dr. Siber trained as a medical intern and resident atRush Presbyterian Medical Center in Chicago and as asenior resident and infectious disease specialist at BethIsrael Hospital and Children’s Hospital in Boston.

44

Connie Schmaljohn, Ph.D.Meet the Experts Breakfast SessionWednesday, May 11, 20057:00 am – 7:45 am

Dr. Connie Schmaljohn is Chief of the MolecularVirology Department at the United States Army Medicalresearch Institute of Infectious Diseases (USAMRIID).She received her bachelors degree in Microbiology fromthe University of Nebraska and her Ph.D. inMicrobiology/Virology from Colorado State University.

Dr. Schmaljohn made key contributions to thediscovery and characterization of hantaviruses andremains a leading expert in hantavirus research. Mostrecently, her research has focused on molecular vaccinedevelopment with an emphasis on multiagent DNAvaccines for hantaviruses, bunyaviruses, flaviviruses,alphaviruses, and filoviruses.

Dr. Schmaljohn holds committee positions in theAmerican Society for Virology and the American Societyfor Tropical Medicine and Hygiene and is an EditorialBoard member for Virology, Emerging Infectious Diseases,Journal of Virology, Virology Journal, and Virus Research.

Dr. Schmaljohn has coauthored more than 110research papers, reviews and book chapters and hasedited several books.

MEET THE EXPERT PRESENTERS

Eighth Annual Conference

Page 45: On-Site Progr

on Vaccine Research

45

ABSTRACTS OFINVITED

PRESENTATIONS

Page 46: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

46

ABSTRACTS OF INVITED PRESENTATIONS

Dilemmas in Public Health: From Smallpox to Polio, SARS andAvian InfluenzaD. HeymannWorld Health Organization, Geneva, Switzerland

Smallpox vaccine, effectively used in a worldwide eradicationprogramme, led to certification of smallpox eradication in 1980. In1981, less than a year afterwards, AIDS was first identified. It is nowunderstood that had smallpox not been eradicated before AIDStransmission began to amplify worldwide, it would have been difficultif not impossible to eradicate using the same vaccine. The smallpoxeradication saga continues today as the threat of deliberate use of thesmallpox virus to cause harm, perceived by some nations, has led to anactive smallpox research programme for new vaccines and antivirals,using infective smallpox virus that was not destroyed after eradication.Efforts towards the eradication of a second viral disease, polio, haverecently been intensified and it is now feasible that polio transmissioncould be interrupted by the end of 2005. It is a paradox that thevaccine being used for polio eradication worldwide - trivalent oralpolio vaccine (OPV) - will itself become a risk after polio has beeneradicated because of recombination events that produce vaccine-derived polioviruses (VDPV). VDPV will change the risk benefit ofOPV once wild poliovirus has been eradicated, requiring that its use bediscontinued. SARS, for which there was no vaccine, is thought tohave emerged into human populations in China sometime in late2002. As many other emerging infections, SARS spreadinternationally, infected a disproportionate number of health workers,required a worldwide effort to contain, and caused severe economicloss. Vaccine development, begun with enthusiasm by severalpharmaceutical companies, some with government funding, has nowdecreased as further understanding about SARS epidemiology hasbecome clear. And finally, the challenge of avian influenza has shownthe importance of increasing vaccine production capacity in a worldwhere annual influenza vaccine production is less than 300 milliondoses, and the need to scale up production of a new influenza vaccineis a continuing, but poorly understood possibility.

References:1. Heymann DL, Aylward RB. Perspective article. Global Health:

Eradicating Polio. New England Journal of Medicine, 2004, 351 (13):1275-1277.

2. Heymann DL, de Gourville EM, Aylward RB. Protecting investmentsin polio eradication: the past, present and future of surveillance foracute flaccid paralysis. Epidemiology and Infection, October 2004, 132(5): 779-80.

3. Heymann DL. Smallpox containment updated: considerations for the21st century. International Journal of Infectious Diseases, October 2004,8 (S2): S15-S20 (ISSN 1201-9712).

4. Heymann DL, Rodier G. SARS: Lessons from a new disease. KnoblerS, Mahmoud A, Lemon S, Mack A, Sivitz L, Oberholtzer K, eds.Learning from SARS. Preparing for the Next Disease Outbreak. WorkshopSummary. Washington, DC, USA, The National Academies Press,2004: 234-246.

1 Use of Conjugate Vaccines as a Probe to Define the Role of the Pneumococcus in the Etiology of PneumoniaK. KlugmanEmory University, Department of International Health, Atlanta, GA

The bacterial etiology of pneumonia is underestimated asdiagnostic tests such as blood culture have very poor sensitivity.Conjugate vaccine trials have documented that the bacterial pathogensHaemophilus influenzae and Streptococcus pneumoniae each cause 20 -37% of bacterial pneumonia in children with X-ray documentation.These remain underestimates given the impact of misclassification onthis method of assessment. Non specific inflammatory markers such asprocalcitonin and C reactive protein may increase the specificity of theX-ray diagnosis of bacterial pneumonia. These bacteria may also play asignificant role in hospitalization of patients infected with respiratoryviruses. It is probable that the mortality associated with the 1918influenza pandemic was associated with a pneumococcal epidemicamong young adults in overcrowded conditions. Conjugate vaccineadministered to infants has been shown to reduce the burden ofinvasive pneumococcal disease in adults. The role that conjugatevaccine administration to children or adults may play in theprevention of pneumonia in the elderly remains to be established inclinical trials. Conjugate pneumococcal vaccine may play a role inreducing mortality from pneumonia in both children and adults.

References:1. Klugman KP et al. A trial of a 9-valent pneumococcal conjugate

vaccine in children with and those without HIV infection. N Engl JMed. 2003; 349:1341-8.

2. Black SB et al. Effectiveness of heptavalent pneumococcal conjugatevaccine in children younger than five years of age for prevention ofpneumonia. Pediatr Infect Dis J 2002; 21:810-5.

3. Madhi SA, Klugman KP, Vaccine Trialist Group. A role forStreptococcus pneumoniae in virus-associated pneumonia. Nat Med.2004;10:811-3.

4. Cutts FT et al. Efficacy of nine-valent pneumococcal conjugatevaccine against pneumonia and invasive pneumococcal disease inThe Gambia: randomised, double-blind, placebo-controlled trial.Lancet 2005;365:1139-46.

Avidity and Biological Activity of Antibodies Elicited by Pneumococcal, Hib, and Meningococcal Conjugate VaccinesD. GoldblattUniversity College of London, London, UK

Conjugate vaccines for Streptococcus pneumoniae, Haemophilusinfluenzae type b and Neisseria meningitidis Group C have changed theface of pediatric infectious diseases. These three pathogens have incommon a capsular polysaccharide (of different chemical structure),which gives them a unique advantage. Their capsules prevent thedeposition of complement and are thus important virulence factorsallowing them to evade host immunity yet conversely are targets forprotective antibody. Young children are unable to efficiently mount animmune response (of which the most important is antibody) topolysaccharide antigens, explaining their susceptibility to invasivedisease with these organisms. Conjugation of capsular polysaccharideto a protein carrier has resulted in glycoconjugate vaccines that induceprotective antibodies to polysaccharide when administered in the firstfew months of life. The protein carrier has the effect of changing the

2

3

Eighth Annual Conference

Page 47: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

47

(continued)

Results of the Pneumococcal Conjugate Vaccine TrialF. CuttsWorld Health Organization, Geneva, Switzerland

Pneumonia is estimated to cause 2 million deaths each year inchildren. Streptococcus pneumoniae is the most important cause of severepneumonia. We conducted a randomised, placebo-controlled double-blind trial in rural Gambia of a nine-valent pneumococcal conjugatevaccine, with the primary objective of evaluating efficacy against a firstepisode of radiological pneumonia. We randomised children aged 6-51weeks to receive 3 doses of pneumococcal conjugate vaccine or placebowith intervals of at least 4 weeks between doses. We conductedsurveillance for radiological pneumonia, invasive disease and adverseevents at a major health centre and hospital. We monitored mortality byrecording outcome of admissions and conducting three-monthly homevisits to each child. In per-protocol analyses, pneumococcal vaccineefficacy was 37% (95% CI: 27, 45) against radiological pneumonia;77% (95% CI: 51, 90) against vaccine-type invasive pneumococcaldisease; 50% (95% CI: 21, 69) against all invasive pneumococcaldisease; 15% (95% CI: 7, 21) against all-cause admissions, and 16%(95% CI: 3, 28) against mortality. In this rural African setting,pneumococcal conjugate vaccine has high efficacy against pneumoniaand invasive pneumococcal disease, and can significantly reduce hospitaladmissions and improve child survival.

Reference:1. Cutts FT, Zaman SM, Enwere G, Jaffar S, Levine OS, Okoko JB, et.

al.; Gambian Pneumococcal Vaccine Trial Group. Efficacy of nine-valent pneumococcal conjugate vaccine against pneumonia andinvasive pneumococcal disease in The Gambia: randomised, double-blind, placebo-controlled trial. Lancet 2005;365:1139-46.

New Conjugate Vaccine to Prevent AnthraxJ. RobbinsNational Institute of Child Health and Human Development, NIH, Bethesda, MD

Both the protective antigen (PA) and the poly(gamma-d-glutamicacid) capsule (gamma dPGA) are essential for the virulence of Bacillusanthracis. A critical level of vaccine-induced IgG anti-PA confersimmunity to anthrax, but there is little or no information about theprotective action of IgG anti-gamma dPGA. Because the number ofspores resulting from exposure due to a bioterrorist event might begreater than encountered in nature, we sought to induce capsularantibodies to expand the immunity conferred by available anthraxvaccines. Non-immunogenic gamma dPGA or corresponding syntheticpeptides were bound to bovine serum albumen, recombinant B.anthracis PA (rPA), or recombinant Pseudomonas aeruginosa exotoxin A(rEPA). To identify the optimal construct, conjugates of B. anthracisgamma dPGA, Bacillus pumilus gamma dLPGA, and peptides of varyinglengths (5-, 10-, or 20-mers), of the d or l configuration with activegroups at the N or C termini, were bound at 5-32 mol per protein. Theconjugates were characterized by physico-chemical and immunologicalassays, including GLC-MS and matrix-assisted laser desorptionionization time-of-flight spectrometry, and immunogenicity in 5- to 6-week-old mice. IgG anti-gamma dPGA and antiprotein were measuredby ELISA. The highest levels of IgG anti-gamma dPGA were elicited bydecamers of gamma dPGA at 10 -20 mol per protein bound to the N- or

6

immune response to the capsular polysaccharides by recruiting T cellsto help polysaccharide specific B cells make antibodies. In addition totheir enhanced immunogenicity, and in contrast to polysaccharidevaccines, conjugate vaccines induce immunological memory. Onemeasurable marker of that memory, is avidity maturation, an increasein the strength with which antibody binds to antigen, demonstrable inthe months following priming. Avidity is also an importantcomponent of the function of antibody, with animal modelssuggesting protection can be achieved with less high compared to lowavidity antibody. Such direct relationship in the protection of humanshas proven more difficult to illustrate.

Reference:1. Jodar L, Butler J, Carlone G, Dagan R, Goldblatt D, Kayhty H,

Klugman K, Plikaytis B, Siber G, Kohberger R, Chang I, CherianT. Serological criteria for evaluation and licensure of newpneumococcal conjugate vaccine formulations for use in infants.Vaccine. 2003 Jul 4;21(23):3265-72.

Long-term Immunologic Memory: Insights from the UK Experience with Haemophilus Influenzae Type b andMeningococcal C ConjugatesE. MillerCommunicable Disease Surveillance Center, London, UK

In the UK Hib was introduced as a primary immunization in1992 at 2/3/4 months of age (the schedule used for DTP vaccine)without a booster because it was thought that immunologic memoryinduced in infancy would provide long-term protection. The samelogic was applied to meningococcal serogroup C conjugate (MCC)vaccine introduced in 1999 in the UK, the first country to license thisnew vaccine based purely on immunologic correlates of protectionwithout supporting efficacy data. Clinical trials with MCC vaccinehad confirmed that it induced immune memory which persisted for atleast 3 years after primary immunization in infants. The subsequentresurgence of Hib disease in the UK in 1999 and the rapid decline inefficacy with MCC vaccines within one year after completion ofinfant immunization has prompted a major rethink of the role ofimmune memory in long term protection. Interestingly, efficacy ofboth Hib and MCC vaccines, and antibody levels, persist better inolder age groups (>1 year) given a single dose but no subsequentboosters. Despite the decline in efficacy of MCC vaccine in theyounger age groups, the control of the disease in the UK is stillexcellent. It is now clear, informed by mathematical models of carriageand disease, that protection against carriage induced by the conjugatevaccines has made a major contribution to the success of the Hib andMCC vaccine programs. The immunologic correlates of long-termprotection with conjugate vaccines remain unclear.

Reference:1. Snape MD, Pollard AJ. Meningococcal polysaccharide-protein

conjugate vaccines. Lancet Infect Dis. 2005 Jan;5(1):21-30

4

5

Page 48: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

48

ABSTRACTS OF INVITED PRESENTATIONS

C-terminal end. High IgG anti-gamma dPGA levels were elicited bytwo injections of 2.5 microg of gamma dPGA per mouse, whereasthree injections were needed to achieve high levels of proteinantibodies. rPA was the most effective carrier. Anti-gamma dPGAinduced opsonophagocytic killing of B. anthracis tox-, cap+. gammadPGA conjugates may enhance the protection conferred by PA alone.gamma dPGA-rPA conjugates induced both anti-PA and anti-gammadPGA.

References:1. Schneerson R, Kubler-Kielb J, Liu TY, Dai ZD, Leppla SH, Yergey

A, Backlund P, Shiloach J, Majadly F, Robbins JB. Poly(gamma-D-glutamic acid) protein conjugates induce IgG antibodies in mice tothe capsule of Bacillus anthracis: a potential addition to the anthraxvaccine. Proc Natl Acad Sci U S A. 2003; 100:8945-50.

2. Leppla SH, Robbins JB, Schneerson R, Shiloach J. Development ofan improved vaccine for anthrax. J Clin Invest. 2002; 110:141-4.

New Rotavirus VaccinesD. SteeleWorld Health Organization, Geneva, Switzerland

Rotavirus remains the most common cause of severe acutegastroenteritis in infants and young children worldwide and isassociated with approximately half a million childhood deaths eachyear.

Rotavirus vaccine development progresses rapidly with majorinvolvement from the international pharmaceutical industry and thecreation of the GAVI-funded Rotavirus Vaccine Program. Recently,alternative vaccine candidates have been taken up by vaccine producersin developing countries. Although certain rotavirus vaccines have beenlicensed in local national settings, and some candidates havecompleted enormous safety and efficacy trials, none of the variouscandidates have shown their clinical efficacy in infants in thedeveloping countries of Africa and Asia.

Challenges to rotavirus vaccine development include the lack ofefficacy data in infant populations most at risk of the disease;interaction with other routine EPI vaccines, especially OPV; safetyissues such as the risk of intussusception and administration to HIV-infected infants. Finally, issues such as supply and price of the vaccinesand equity of distribution of these new vaccines to the regions wherethey are most needed have not been creatively addressed yet by theinternational community nor the countries.

Reference:1. Glass R, Bresee J, Parashar U, Jiang B, Gentsch J. The future of

rotavirus vaccines: a major setback leads to new opportunities. TheLancet 363:1547-1550, May 2004

Progress in Vaccines Against Norwalk VirusR. AtmarBaylor College of Medicine, Houston, TX

Norwalk virus is a human calicivirus and is the prototype norovirusstrain. Noroviruses are the most common cause of epidemicnonbacterial gastroenteritis and are also common causes of sporadicgastroenteritis. These viruses cause significant morbidity in a variety

of populations, including persons in healthcare facilities, in the militaryand on cruise ships. The impact of these viruses as causes of foodborneand waterborne disease indicates that development of an effectivevaccine would be useful. However, there are many challenges that mustbe overcome. Human noroviruses cannot be grown in cell culture andno small animal model of infection exists. However, a human challengemodel is available. The antigenic and genetic diversity of noroviruseswill likely complicate the development of a broadly effective vaccine.Furthermore, correlates of immunity to noroviruses are poorly defined,although recent studies have shown that lack of expression of certainhisto-blood group antigens is associated with resistance to infection byNorwalk virus. Expression of the structural proteins of Norwalk virusleads to the formation of virus-like particles (VLPs) that areimmunogenic when administered to mice and humans. Norwalk virusVLPs are currently under investigation as a potential vaccine candidate.

References:1. Estes MK, Ball JM, Guerrero RA, Opekun AR, Gilger MA, Pacheco

SS, Graham DY. Norwalk virus vaccines: challenges and progress. J Infect Dis 2000;181:S367-73.

2. Tacket CO, Mason HS, Losonsky G, Estes MK, Levine MM, ArntzenCJ. Human immune responses to a novel Norwalk virus vaccinedelivered in transgenic potatoes. J Infect Dis 2000;182:302-5.

3. Hutson AM, Atmar RL, Estes MK. Norovirus disease: changingepidemiology and host susceptibility factors. Trends Microbiol2004;12:279-87.

Immunology and Potential Vaccine Prevention of Clostridium difficile ColitisK. KotloffUniversity of Maryland School of Medicine, Baltimore, MD

Clostridium difficile is a major cause of nosocomial diarrhea,particularly affecting the elderly and immunocompromised. Althoughusually responsive to medical therapy, infection can increase morbidity,prolong hospitalization, and produce life-threatening colitis. Toxins A and B are the principal virulence factors of this noninvasive organism.Considerable evidence supports the role of antitoxic immunity, mostnotably against toxin A, in prevention of and recovery from C. difficile-associated diarrhea (CDAD). Vaccines that induce antitoxic immunityare thus being explored as a means for protecting high-risk individuals.Approaches showing promise in preclinical models include recombinantfusion proteins containing the nontoxic binding domain of toxin A anddelivered to the mucosa either directly (with adjuvant) or via a liveattenuated bacterial vector. In the clinical arena, a Phase 1 trial has beencompleted to evaluate a parenteral vaccine containing C. difficile toxoidsA and B. Thirty healthy adults received four spaced inoculations ondays 1, 8, 30 and 60 with either 6.25 Fg, 25 Fg, or 100 Fg of vaccine.Vaccination was generally well-tolerated. Nearly all subjects ($90%)developed vigorous serum antibody responses to both toxins, asmeasured by IgG ELISA and neutralization of cytotoxicity, whereas fecalIgA increases occurred in approximately 50%. Serum antitoxin A IgGELISA titers in all vaccine recipients exceeded the concentrations thathave been associated with protection in clinical studies. More recently,the vaccine was evaluated in three patients with multiple episodes ofCDAD. Strong immune responses were seen in two patients, and nonehad additional recurrences of CDAD. Further development of thisvaccine as a prophylactic or therapeutic agent or for producing C.difficile hyperimmune globulin is warranted.

7

8

9

Eighth Annual Conference

Page 49: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

49

(continued)

Reference:1. Kotloff KL, Wasserman SS, Losonsky GA et al. Safety and

immunogenicity of increasing doses of a Clostridium difficile toxoidvaccine administered to healthy adults. Infect Immun 2001;69(2):988-995.

Update on Microbiology, Immunology and Vaccine Preventionof Dental CariesS. Michalek University of Alabama at Birmingham, Birmingham, AL

Dental caries (i.e., tooth decay) is an infectious disease that affectsthe population worldwide. In the U.S., almost 80% of youth by age 17have experienced carious lesions. This worldwide disease is mostdevastating to less developed countries (see: http://www.who.int/oral_health/publications/report03/en/). In the U.S. alone, dentalservice expenditures approximate $50 billion annually.

The mutans streptococci (MS) are the primary cause of dental cariesin humans. These gram-positive bacteria selectively and specificallycolonize tooth surfaces. An initial “window of infectivity” for MSoccurs in children around 18-24 months of age, a time correspondingwith the emergence of the primary molar teeth. Other “windows” mayopen at other stages in life. Scientific evidence from both animal andadult human clinical trial studies provides evidence for the effectivenessand safety of active and passive immunization strategies, as well asreplacement therapy for use in obtaining protection against dentalcaries. However, the effectiveness of these approaches in children,including infants, is still needed to establish the potential use of theseapproaches in this target population.

References:1. Caufield PW, Cutter GR, Dasanayake AP. Initial acquisition of

mutans streptococci by infants; evidence for a discrete window ofinfectivity. J. Dent. Res. 1993;72:37-45.

2. Childers NK, Zhang SS, Michalek SM. Oral immunization ofhumans with dehydrated liposomes containing Streptococcus mutansglucosyltransferase induces salivary immunoglobulin A2 antibodyresponses. Oral Microbiol. Immunol. 1994;9:146-153.

3. Childers NK, Zhang SS, Michalek SM. Oral immunization ofhumans with dehydrated liposomes containing Streptococcus mutansglucosyltransferase induces salivary immunoglobulin A2 antibodyresponses. Oral Microbiol Immunol. Jun 1994;9(3):146-153.

4. Childers NK, Tong G, Michalek SM. Nasal immunization ofhumans with dehydrated liposomes containing Streptococcus mutansantigen. Oral Microbiol. Immunol. 1997;12:329-335.

5. Childers NK, Tong G, Mitchell S, Kirk K, Russell MW, MichalekSM. A controlled clinical study of the effect of nasal immunizationwith a Streptococcus mutans antigen alone or incorporated intoliposomes on induction of immune responses. Infect. Immun.1999;67:618-623.

Perspective of Vaccine ManufacturersG. Del GiudiceChiron Vaccines, Siena, Italy

A generation of human-avian influenza virus reassortants has beenresponsible for the three pandemic waves that hit mankind during the XX

century (1918, 1957, 1968). In 1997 and again in 2003-2005, a highlypathogenic avian influenza virus (H5N1) has caused several cases in South-East Asia with a mortality rate of about 70%. The most powerful meansto prevent and control the risk of influenza pandemics depends on theproduction and availability of vaccines. Clinical data have shown that,since this vaccine will be used in immunologically naive individuals, morethan one dose may be required for optimal protection. Furthermore, dueto the current limited vaccine manufacturing capacity globally, a dosesaving approach may be required to allow the production of sufficient dosesof vaccine to cover the largest portion of the population worldwide. Thereis already evidence that vaccines against influenza pandemics may wellrequire adjuvants to be effective. Studies using the MF59 adjuvant havedemonstrated that this target is achievable and that even immunizationwith a vaccine not fully matched with the target virus can inducesubstantial cross-reacting protective immune responses. The preparationof vaccines effective against influenza pandemics is technically feasible.However, for regulatory, economical, ethical, legal and logistical reasons,amongst others, this target can be only achieved through a partnershipbetween public and private sectors.

Reference:1. Stephenson I, Bugarini R, Nicholson K, Podda A, Wood J, Zambon M,

Katz J. Cross-Reactivity to Highly Pathogenic Avian Influenza H5N1Viruses after Vaccination with Nonadjuvanted and MF59-AdjuvantedInfluenza A/Duck/Singapore/97 (H5N3) Vaccine: A Potential PrimingStrategy. JID 191:1210-1215, April 2005.

Vaccines Against SARS: Where Do We Stand?A. OsterhausErasmus Medical CollegeRotterdam, Netherlands

After the identification of SARS-CoV as the aetiologic agent of SARSand the rapid development of macaque-, ferret- and rodent models,studies concerning the pathogenesis and the development of interventionstrategies were vigorously pursued. Pegylated interferon-_ proved to bethe first clinically available drug that was effective in preventing and post-exposure treatment of SARS in the macaque model. Subsequentlypassive transfer of neutralizing polyclonal and monoclonal antibodies wasshown to significantly reduce the SARS-CoV load in the respectiveanimal models, when administered preventively. Several research groupsstarted the development of candidate SARS vaccines, using differentclassical and state-of-the-art technologies. It was realised that besidesefficacy, safety of the candidate vaccines should be given specialattention, since it has been shown that vaccination of cats with candidatefeline coronavirus vaccines predisposed them for antibody mediatedenhanced susceptibility upon challenge. In addition, adjuvantedinactivated whole virus vaccines should be given special attention in thisrespect, since the use of candidate measles- and RSV vaccines of this typehave predisposed children for more serious disease upon infection in thepast. Since macaque models have been developed to study thisphenomenon with these two candidate vaccines, it was decided tospecifically demonstrate the absence of this phenomenon with inactivatedcandidate SARS vaccines in macaques. Besides inactivated whole virusvaccines, recombinant-, (MVA-; adeno-), vectored and plasmid DNAvaccines have now been studied in animal models for SARS. In general,these are aiming at the induction of a specific immune response againstthe S protein of the virus. Most of the candidate vaccines studied so farproved to be protective in the animal models used, with S protein specific

10

11

12

Page 50: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

50

ABSTRACTS OF INVITED PRESENTATIONS

virus neutralizing antibodies as the most likely correlate of protection.No antibody-mediated enhancement has been observed, but there areindications that some of the vaccines tested may predispose for thedevelopment of immune pathology upon challenge. The first phase Iclinical study has now been carried out in the Peoples Republic ofChina.

Collectively, the data show that the collaborative activities by severalresearch groups have not only resulted in the rapid control of the SARSoutbreak, but also to the development of preventive and therapeuticstrategies and will soon lead to the availability of effective and safevaccines against SARS.

Vaccines for Prevention, Management, and Eradication of Avian InfluenzaD. SwayneUnited States Department of Agriculture, Athens, GA

Vaccines been used to prevent, manage and eradicate avianinfluenza (AI) in poultry. In the current epizootic of H5N1 AI in Asia,vaccines have emerged to be a valuable tool in control strategies. Fourinactivated AI vaccines, based on low and high pathogenicity H5outbreak strains, have been used in a variety of poultry includingchickens, ducks, geese and Japanese quail. Live fowl poxvirusrecombinants with H5 AI gene inserts will see some usage in chickensin the near future. In addition, AI vaccine strains generated throughreverse genetics will be used in future inactivated vaccines. Influenzavaccines in poultry have provided intense, longer-term and broaderprotection than have influenza vaccines in humans, thus requiring lessfrequent change in vaccine strains than for humans. However, vaccineefficacy should be re-evaluated every 2-3 years against prevailingcirculating AI strains. The AI vaccines provide protection by preventionof clinical signs and death, and reduce respiratory and intestinal virusreplication in poultry. Differentiation of vaccinated from infectedpoultry is critical to measure success of control programs.

Reference:1. Swayne, D.E. Application of New Vaccine Technologies for the Control

of Transboundary Diseases. Develop. Biol. 119:219-228, 2004.

West Nile: An Overview of the Epidemic in North AmericaA. BarrettUniversity of Texas Medical Branch, Galveston, TX

West Nile virus (WNV) has first isolated in Uganda in 1937 and,until recently, was found in parts of Asia, Africa and Europe. In 1999,WNV emerged in North America with an outbreak in New York andsurrounding areas. The virus has subsequently spread throughout theUnited States and into Canada, Mexico, Central America and CaribbeanIslands. The number of human and veterinary cases has increased since1999, including the largest epidemic of arboviral encephalitis everrecorded in the Americas in 2002. In addition to “traditional” mosquito-borne transmission, the virus has been transmitted by a variety of otherroutes including blood-borne. The virus has been found to infect at least48 species of mosquitoes, 285 species of birds and 29 animal species.West Nile virus is clearly a major public health and veterinary problemin the United States. Surprisingly, there is little evidence of human orveterinary disease in Mexico, which may in part be due to the

seroprevalence of antibodies to other flaviviruses. There is considerabledebate on what WNV will do in the future.

Reference:1. Granwehr BP, Lillibridge KM, Higgs S, Mason PW, Aronson JF,

Campbell GA, Barrett ADT. West Nile virus: where are we now?Lancet Infect Dis. 2004; 4:547-56.

Vaccination for Emergency and Emerging Animal Health EventsR. HillUnited States Department of Agriculture, Washington, DC

The Virus-Serum-Toxin Act of 1913 provides the legal basis for theregulation of veterinary biologicals in the United States, and the USDA’sCenter for Veterinary Biologics (CVB) has the authority for the issuanceof licenses and permits for such products. The law was intended toestablish standards and control the importation of products into theU.S. and the distribution of products assuring the purity, safety,potency, and efficacy of veterinary biological products. Prelicensingdata evaluation procedures are designed to assess the quality of eachproduct and support product label claims. Data from all phases ofproduct development are evaluated against these key elements. Underthe standard licensing process, this spectrum of evaluation includescomplete characterization of seed material and ingredients, laboratoryand host animal safety and efficacy studies. This comprehensiveevaluation may not be possible during the emergence of a new animaldisease; however, there are mechanisms that allow for the availability ofproducts in an emergency animal health situation. These mechanismsinclude a variety of licensing/permitting options, including theestablishment of vaccine banks. Historical examples of emerging animaldisease events in the United States will be used to illustrate theregulatory considerations for each type of product authorization.

Reference:1. R.E. Hill, Jr., P.L. Foley, M.Y. Carr, L.A. Elsken, D.M. Gatewood,

L.R. Ludemann, L.A. Wilbur. 2003. Regulatory Consideration forEmergency Use of Non-USDA Licensed Vaccines in the UnitedStates. Developments in Biologics. 114:31-52.

Ideas from the National Vaccine Advisory Committee, the Institute of Medicine, and AcademiaJ. KleinBoston Medical Center, Boston, MA

Recent shortages of vaccines have underlined the vulnerability ofvaccine supply in the United States. Five key issues needed to beaddressed to provide a reliable and predictable supply of routinelyadministered vaccines: 1. Financial incentives need to be identified thatwill maintain current manufacturers and encourage new manufacturersto enter the market; 2. Regulatory constraints on development andproduction of vaccines need to be reconsidered; 3. The Vaccine injuryCompensation Program needs to be strengthened to limit productliability for approved vaccines; 4. Impediments to utilization of vaccinestockpiles by Security and Exchange Commission regulations need to beremoved; and 5. Consumers and legislators, who now undervaluevaccines and are attentive only when vaccine is unavailable, need to bebetter informed about the assets and liabilities of vaccines. Although a

13

14

15

16

Eighth Annual Conference

Page 51: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

51

(continued)

variety of groups including the National Vaccine Advisory Committee,the General Accounting Office, the Institute of Medicine and the SabinVaccine Institute have made recommendations to strengthen vaccinesupply I recent years, progress in addressing the issues of vaccine supplyhas been modest. A robust domestic vaccine manufacturing capabilityshould be considered a national priority. Task forces, including allvaccine constituencies (consumers, purchasers, providers, manufacturers,regulatory authorities, national and local public health authorities,scientists, advocacy groups and legislators) should be established to meetat regular intervals until concrete proposals to strengthen vaccine supplyare developed that can be presented to authoritative agencies.

Reference:1. National Vaccine Advisory Committee. Strengthening the Supply of

routinely Recommended Vaccines in the United States;Recommendations form the National Vaccine Advisory Committee.JAMA 2003; 290: 3122-3128.

Vaccine Supply: A Manufacturer’s Perspective on CurrentChallenges and OpportunitiesD. Johnsonsanofi pasteur, Swiftwater, PA

The vaccine enterprise in this country is a remarkable success storythat has resulted in freedom from disease for millions of children andadults. The existing system fundamentally works, and should bestrengthened and stabilized to improve the delivery of today’s vaccines,as well as to bring forth a new generation of vaccines. Sanofi pasteurproposed a number of actions to ensure a stronger U.S. vaccine supplyto the National Vaccine Advisory Committee in February 2002. Sincethat time, these proposals have been conveyed to many stakeholdersinvolved in the vaccine enterprise. They include the need for the CDCto receive additional funding to create and implement expanded vaccinestockpiles; the inclusion of manufacturer expertise in formulatingimmunization policy; government and policy advisory boards actingwith greater predictability; and strengthening outreach to convey themessage that prevention is the most desirable intervention. There arealso practical steps that can strengthen vaccine predictability and supply,including pay-for-performance incentives to physicians and increasedvaccine reimbursement rates. We have made limited progress in the lastseveral years; government, policymakers, the health care community andindustry still have much work ahead to sustain vaccine discovery,development, production and stable supply.

References:1. CDC Program in Brief, Pediatric Vaccine Stockpiles, January 20042. Institute of Medicine: Financing Vaccines in the 21st Century:

Assuring Access and Availability, 20033. National Vaccine Advisory Committee: Strengthening the Supply of

Routinely Recommended Vaccines in the United States,Recommendations From the National Vaccine Advisory CommitteeJAMA. 2003;290:3122-3128.

4. Needs and Recommendations for the United States PoliovirusVaccine Stockpile, Report of the Joint Working Group of theNational Vaccine Advisory Committee (NVAC) and AdvisoryCommittee on Immunization Practices (ACIP), National VaccineAdvisory Committee Meeting, February 3-4, 2004

5. United States General Accounting Office, Report to CongressionalRequesters: Childhood Vaccines: Ensuring an Adequate Supply Poses

Continuing Challenges, September 20026. Sanofi pasteur: Strengthening Vaccine Supply: Principles to Ensure

Vaccine Supply, February 2002-present

A Consensus Agenda to Strengthen U.S. Vaccine SupplyJ. ClymerPartnership for Prevention, Washington, DC

Large gaps between U.S. adult immunization goals and actualvaccination rates have persisted for years, even as the gaps in pediatricimmunization have been narrowed or eliminated. Immunizationstakeholders – federal, state and local public health agencies;manufacturers; clinical practitioners; legislators, scholars; health plans,employers; public sector purchasers; and the public – have discussed theproblem and offered potential solutions but, lacking consensus, have notachieved significant policy action. Partnership for Prevention gatheredleaders of these stakeholder groups to develop one consensus policyrecommendation that, if passed, would move the needle on adultimmunization. The expert panel surpassed its goal, producing sixrecommendations.

This session will identify the specific recommendations, outline plansto translate them into policy, and discuss how the stakeholders overcametheir differences to achieve consensus.

Reference:1. Strengthening Adult Immunization: A Call to Action, Partnership for

Prevention, Washington, DC, 2005

Progress in the Field of Malaria VaccinologyF. DubovskyMalaria Vaccine Initiative, Bethesda, MD

There are multiple lines of evidence that assure the field that malariavaccines are technically feasible. In the recent past these experimentalscenarios have been partially replicated with vaccine constructs.Classically malaria vaccinology has focused different immunologiceffector mechanisms on different stages in the parasite life cycle. Pre-erythrocytic sterile immunity has been demonstrated with a prime-boostvaccine regime (Fowlpox/Modified Vaccinia Ankara bearing a string ofepitopes and the TRAP antigen) as well as recombinant proteins (amixed chimeric Hepatitis B virus-like-particle delivering CSPformulated in an oil-in-water emulsion with QS21 and MPL – termedRTS,S/AS02). Impact on parasite density has been demonstrated underfield conditions using a mixture of recombinant proteins delivered in awater-in-oil emulsion (RESA, MSP2, MSP1 in Montanide ISA 720).Globally there are many vaccine candidates that are being developed andfor the first time many of these candidates are using technologies thathave the potential to be manufactured at industrial scale. A pilot proof-of-concept efficacy trial has completed with RTS,S/AS02 in 1-4 year oldchildren in Mozambique. Over the six month observation period thevaccine was able to impact not only infection, but also clinical diseaseand severe disease.

Reference:1. Alonso PL, Jahit S, Aponte JJ et al. Efficacy of RTS,S/AS02A vaccine

against Plasmodium falciparum infection and disease in young Africanchildren: randomized controlled trial. Lancet 2004;364: 1411-1420

17

18

19

Page 52: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

52

ABSTRACTS OF INVITED PRESENTATIONS

Novel and Classical Strategies for Attenuated Malaria VaccinesS. KappeSeattle Biomedical Research Institute, Seattle, WA

Malaria is transmitted by means of inoculation of the Plasmodiumparasite sporozoite stage during a mosquito bite. Sporozoites infect theliver where they transform into liver stages, each developing intothousands of first generation red blood cell infectious merozoites.Using a mouse malaria model it was shown decades ago thatimmunization with irradiated sporozoites completely protected againstsubsequent nonirradiated sporozoite challenge. This findingdemonstrated that sterilizing immunity against malaria infection isachievable. Experimental immunization of humans with irradiatedsporozoites of human malaria parasites also conferred sterile protectionthus making the liver stage a prime malaria vaccine target. However,until recently the use of irradiated sporozoites as a malaria vaccine wasconsidered impractical for a variety of reasons. We have recently shownin a mouse malaria model that inactivation of a single parasite genearrests productive liver stage development and renders the parasiteunable to establish a blood stage infection. Immunization withsporozoites of such attenuated parasites confers complete protectionagainst infectious sporozoite challenge in mice. This protection is longlasting and stage-specific. We will discuss this new concept of agenetically attenuated malaria parasite vaccine and outline the possibleuse for human vaccinations.

Reference:1. Mueller, A. K., Labaied, M., S. H. I. Kappe and K. Matuschewski

(2005). Genetically modified Plasmodium parasites as a protectivelive attenuated experimental malaria vaccine. Nature. 433, 164-167

The Pathway Forward for Malaria Recombinant Vaccine (RTS,S): Implications of Study ResultsW. BallouGlaxoSmithKline Biologicals, Rixensart, Belgium

Plasmodium falciparum is responsible for at least 300 million casesof malaria each year and more than 1 million deaths, especially inchildren under 2 years of age in sub-Saharan Africa. RTS,S/AS02A is acandidate malaria vaccine that induces antibody and cellular immuneresponses that inhibit the ability of malaria sporozoites to successfullyinfect humans. Efficacy reflected in both sterile immunity and/or aprofound (>90%) reduction in infectious inoculum followinginfectious mosquito challenge has been confirmed in field trials inmalaria-endemic regions. The most impressive data come from arecently completed proof-of-concept trial in more than 2000 childrenaged 1-4 years in Mozambique, where significant vaccine efficacyagainst infection, clinical malaria and severe malaria were observed. Thenext steps include determination of an optimal vaccination schedule inthe target population (infants and children under 2), confirmation ofvaccine safety and immunogenicity in infants, and implementation ofan appropriate Phase III program that could lead to vaccine licensure.

Reference:1. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, Milman J,

Mandomando I, Spiessens B,Guinovart C, Espasa M, Bassat Q, AideP, Ofori-Anyinam O, Navia MN, Corachan S, Ceuppens M, DuboisMC, Demoitié MA, Dubovsky F, Menéndez C,Tornieporth N,

Ballou WR, Thompson R, Cohen J. Efficacy of the RTS,S/AS02Avaccine against Plasmodium falciparum infection and disease inyoung African children: randomised controlled trial. Lancet2004;364:1411 –20

Developing Adjuvants for Malaria VaccinesD. HeppnerWalter Reed Army Institute of Research, Silver Spring, MD

The Malaria Vaccine Program at the Walter Reed Army Institute ofResearch is developing a multi-antigen, multi-stage recombinant proteinsubunit-based vaccine intended to induce effective immune responsesagainst pre-erythrocytic and blood stages of Plasmodium falciparum.Although the immune correlates of vaccine-induced protection are notknown, it is assumed that an effective subunit malaria vaccine willrequire a safe, potent adjuvant able to induce both a sustained cellularand humoral immune responses against protective epitopes containedwithin soluble exogenous recombinant proteins. Additional selectioncriteria for a successful adjuvant include acceptable reactogenicity,antigen compatibility, and overall stability of the antigen-adjuvantformulation. Many adjuvants have been evaluated pre-clinically,relatively few have been systematically evaluated in humans, and feweryet in the context of clinical efficacy. Recently, the development ofRTS,S/AS02A, a purified subunit-based malaria vaccine formulatedwith a novel adjuvant, illustrates the potential for an improved adjuvantsystem to drive an effective immune response. Several adjuvants (CpG,oil-in-water emulsions, MPL, QS21 and others) will be reviewed in thecontext of recent malaria vaccine development with an emphasis onlessons learned from pre-clinical evaluations of candidate vaccines usingnon-human primate models

Reference:1. Heppner DG Jr, Kester KE, Ockenhouse CF, et. Al. Towards an

RTS, S-based, multi-stage, multi-antiagen vaccine against falciparummalaria: progress at the Walter Reed Army Institute of Research.Vaccine 23 (17-18):2243-50, March 2005.

Human Pailloma Virus Therapeutic Vaccines: Overview and Underlying ImmunologyW. KastUniversity of Southern California, Los Angeles, CA

Human papillomavirus (HPV) infection of cervical epithelium islinked to the generation of cervical cancer. Although most womeninfected with HPV clear their lesions, the long latency period frominfection to resolution indicates that human papillomavirus evolvedimmune escape mechanisms. Dendritic cells, targeted by vaccinationprocedures, incubated with HPV virus-like particles induce an HPV-specific immune response. Langerhans cells located at the sites ofprimary infection do not, implicating the targeting of Langerhans cellsas an immune escape mechanism utilized by HPV. Langerhans cellsincubated with HPV virus-like particles upregulate the PI3K pathwayand down regulate MAPK pathways. With the inhibition of PI3K andincubation with HPV virus-like particles, Langerhans cells initiate apotent HPV-specific response. PI3K activation in Langerhans cellsdefines a novel escape mechanism utilized by HPV. Apart from thesefundamental HPV immunological data an overview w ill be given of the

20

21

22

23

Eighth Annual Conference

Page 53: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

53

(continued)

current developments in the field of preventive and therapeutic vaccinesagainst HPV.

References:1. Brinkman JA, Caffrey AS, Muderspach LI, Roman LD and Kast

WM: The impact of anti HPV vaccination on cervical cancerincidence and HPV induced cervical lesions: Consequences forclinical management. Eur J Gyn Onc, In Press, 2005.

2. Fausch SC, Fahey LM, Da Silva DM and Kast WM: HPV can escapeimmune recognition through Langerhans cell PI3-kinase activation, JImmunol, In Press, 2005.

3. Da Silva DM and Kast WM: Vaccination against cervical cancer:hopes and realities. Am J Cancer, In Press 2005.

Hepatitis C Therapeutic VaccinesM. HoughtonChiron Corporation, Emeryville, CA

There is evidence that the liver disease associated with persistentinfection by the hepatitis C virus (HCV) is modulated by both humoraland cellular immune responses to the virus and that response toantiviral therapy is also predicated by such immune responses. It followstherefore that boosting of viral immunity by appropriate vaccinationcould be of therapeutic value, particularly when given as adjuncttherapy with antivirals. To this end, a clinical program is underwaydesigned to boost both HCV-specific humoral and cellular immuneresponses in chronically-infected HCV patients using vaccinemonotherapy and combination vaccine and antiviral therapies. It ishoped that use of recombinant envelope glycoprotein gpE1/gpE2vaccine formulations will boost cross-neutralising antibody titers andthus help to control disease progression and response to antiviraltherapy while an ISCOM-adjuvanted polyprotein formulation mayboost broadly cross-reactive CD4+ and CD8+ T cell responses to thevirus with similar benefits. Supporting pre-clinical data for theseexperimental approaches will be presented along with the status of theclinical development pathway.

Reference:1. Hsu HH, Abrignani S & Houghton M. Prospects for a Hepatitis C

Vaccine. Clin Liver Dis. 3(4) : 901-915, 1999

Immunotherapy as a Treatment Possibility for Alzheimer’s DiseaseD. SchenkElan Pharmaceuticals, South San Francisco, CA

The idea of immunizing patients suffering from an amyloidosis withthe offending monomer has emerged as a novel strategy to treat anumber of diseases including Alzheimer’s. The basis for such anapproach is that beta amyloid peptide is an unusual byproduct of amuch larger precursor protein, that once cleaved out, readily formsamyloid fibrils that accumulate in the brain tissue of Alzheimer’spatients and are thought to cause cognitive dysfunction as a result ofneuronal stress and eventually death. By

immunizing with beta amyloid peptide or its fragments, eitheractively or passively, antibodies generated to it evidently appear todisrupt the amyloidogenic process and result in reduced plaque lesionsin both animal models and in patients suffering from the disease. Both

the biologic and clinical implications of these findings will be presentedand discussed at the meeting.

Reference:1. Masliah E, Hansen L, Adame A, Crews L, Bard F, Lee C, Seubert P,

Games D, Kirby L, Schenk D (2005) Abeta vaccination effects onplaque pathology in the absence of encephalitis in Alzheimer disease.Neurology 64:129-131.

Vaccines for Type 1 DiabetesR. InselJuvenile Diabetes Research Foundation International, New York, NY

Type 1 (or insulin-dependent) diabetes is an autoimmune diseaseassociated with cell-mediated immune destruction of the insulin-producing beta cells of pancreatic islets. Approximately 16,000childhood cases of type 1 diabetes occur annually in the United States, arate similar to that of several infectious diseases for which vaccines havebeen developed and licensed. Both genetic and environmental riskfactors contribute to disease, whose incidence has been increasing for atleast the last four to five decades. The period between initial insulitisand sensitization to beta cell antigens and the onset of overt, clinicaldiabetes is quite variable, resulting in a large pool with prediabetes.

Primary prevention with vaccines targeted at environmental factorsis not currently possible. Secondary prevention with vaccines after thedisease process has commenced is being explored in both the at-risksetting, to prevent the onset of diabetes, and the recent-onset diabetessetting, to preserve functional beta cell mass. Current vaccineapproaches are targeted at altering function of pathogenic effector Tcells and inducing lasting immunologic tolerance or immunoregulationto beta cell antigens. Beta cell antigen-specific approaches underinvestigation include presentation of beta cell antigens by tolerogenicroutes (oral, intranasal), in tolerogenic forms (peptides, altered peptideligands), on tolerogenic cells (resting dendritic cells) or with co-stimulatory blockade. Islet cell antigens used in vaccines have included:insulin, proinsulin, insulin B chain or peptides (B:9-23), glutamic aciddecarboxylase (GAD65), and heat shock protein 60 (Hsp60)-derivedpeptides (p277). Antigen non-specific approaches include the use ofFcR nonbinding anti-CD3 monoclonal antibody (anti-CD3 Ab), whichcan induce lasting remission of diabetes in nonobese diabetic (NOD)mice that is associated with the generation of regulatory T cells. Short-term administration of humanized FcR nonbinding anti-CD3 Ab innewly diagnosed human type 1 diabetes preserves residual beta cellfunction for over a year, as evidenced by a decrease in the rate of loss ofinsulin production, improved glycemic control, and concomitantreduction of insulin dosing. To augment and prolong the effect, anti-CD3 Ab will be evaluated when administered for multiple courses,simultaneously with islet antigens, or with beta cell regenerationtherapeutics. In general, vaccine development for type 1 diabetes will beaccelerated by the identification of human beta cell T cell epitopes,improved approaches in quantifying pathogenic autoantigen-reactiveeffector T cells and protective regulatory T cells, and the generation ofsurrogate markers for therapeutic effects. A robust infrastructure fordeveloping and evaluating type 1 diabetes vaccines currently existsthrough support of multiple funding agencies, including JDRF.

References:1. Raz I, Eldor R, Naparstek Y. Immune modulation for prevention of

type 1 diabetes mellitus. Trends Biotechnol. 2005 Mar;23(3):128-34

24

25

26

Page 54: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

54

Eighth Annual Conference2. Chatenoud L. Anti-CD3 antibodies: towards clinical antigen-specific

immunomodulation. Curr Opin Pharmacol. 2004 Aug;4(4):403-7.Review

3. Bluestone JA, Tang Q. Therapeutic vaccination using CD4+CD25+antigen-specific regulatory T cells. Proc Natl Acad Sci U S A. 2004Oct 5;101 Suppl 2:14622-6. Review.

Limitations of B-cell Responses in Early LifeC. SiegristUniversity of Geneva, Geneva, Switzerland

Early immunization is required to induce immunity against diseasesthat may occur very early in life. This is however limited by the relativeimmaturity of the immune system. Vaccine antibody responses increasewith age in a step-wise manner, such that only the most immunogenicvaccines show significant protective efficacy after a single infant dose.Murine models assessing neonatal responses to human infant vaccinesindicated that this reflects a stepwise increase in Germinal Centerreactions, limited by the delayed postnatal development of folliculardendritic cells. Another hallmark of early life responses are the shortpersistence of vaccine antibodies. This reflects in mice a limited capacityof the neonatal bone marrow to support the establishment of long-livedplasma cells: plasmablasts efficiently migrate to the early life bonemarrow, but fail to receive appropriate differentiation / survival signals.Importantly, factors limiting the magnitude and duration of infantresponses do not prevent efficient priming, such that neonatally-triggered immune memory may be readily recalled later in life.Unfortunately, however, immune memory is not sufficient to confer full protection against pathogens requiring neutralizing antibodies either at the infection site (RSV, influenza) or rapidly after exposure(encapsulated bacteria), and these remain major threats to young infants.

Reference:1. Lambert P.H., Liu M. and C.A. Siegrist, Can successful vaccines teach

us how to induce efficient protective immune responses? NatureMedicine, April 5, 2005 (in press)

Recent Advances in Studies of B and T cell Responses Early in LifeJ. CroweVanderbilt University, Nashville, TN

This talk will review the evidence that the antibody response madeby infants in response to viral infection or vaccination is of lowmagnitude and poor quality. Using molecular studies of the humaninfant response to two major common viral pathogens of infancy,respiratory syncytial virus and rotavirus, we will explore the molecularbasis for poor quality infant antibodies. The talk will emphasize the roleof somatic hypermutation in enhancing the antiviral function ofantibodies, and the poor ability of infants to introduce mutations intoantibody gene sequences following primary infection or vaccination.

Reference:1. Weitkamp JH, Kallewaard N, Kusuhara K, Bures E, Williams JV,

LaFleur B, Greenberg HB, Crowe JE Jr. Infant and Adult Human BCell Responses to Rotavirus Share Common ImmunodominantVariable Gene Repertoires. Journal of Immunology 2003; 171: 4680-8.

Innate Imprinting and Inflammatory Lung DiseaseT. HussellImperial College London, London, England

Immune responses to infectious disease need to be regulated toprevent bystander tissue damage. Regulation is carefully orchestrated inorganized lymphoid tissue such as lymph nodes but less so in sites wherecompartmentalization of immune cells is not as apparent. The lung is aclassic example of a site where inflammation is not regulated (the site ofasthma, pneumonia and bronchiolitis). Lower respiratory tract viralinfections recruit a vast inflammatory infiltrate that, in clearing thevirus, occlude the airways. In addition, inflammatory cells releasemediators such as TNF that act on distant organs to cause the lifethreatening clinical symptoms of wasting, weight loss, fever and appetitesuppression. One therapeutic strategy would involve the induction ofregulation at this vulnerable site. We show that the lungmicroenvironment can be educated to respond to a variety of infectionsin a more controlled manner by instilling toll-like receptor ligands orimmunogenic proteins derived from bacteria such as LTK63. We havenamed this procedure “innate imprinting”, which is long lasting andprovides generic protection against a variety of serious lower respiratorytract infections.

Reference:1. Williams, A.E., Edwards, L., Humphreys,I.R., Snelgrove, R., Rae, A.,

Rappuoli, R. and Hussell, T. “Innate imprinting by the modifiedheat-labile toxin of Escherichia coli (LTK63) provides genericprotection against lung infectious disease”. J Immunol. 173 (2004),7435-43.

Interrogating and Exploiting Memory B CellsA. LanzavecchiaInstitute for Research in Biomedicine, Bellinzona, Switzerland

Our current interest is to understand the mechanisms that control Bcell priming and maintenance of memory B cells and serum antibodylevels. We developed a new strategy for the purification of human naïveB cells that was instrumental to determine their activation requirementsand in vivo turnover. We found that, in order to undergo clonalexpansion and differentiation, naïve B cells require, in addition to BCRstimulation and cognate T cell help, a third signal delivered by microbialproducts through Toll like receptors. In contrast, memory B cells canproliferate and differentiate in response to polyclonal stimuli such asbystander T cell help, TLR agonists and homeostatic cytokines.Consistent with these different activation requirements is theobservation that human memory B cells spontaneously turn over at highrate in vivo, while naïve B cells do not. To gain insights into themechanisms that sustain the serological response we monitored plasmacell generation, serum antibodies and frequency of memory B cells indonors boosted with tetanus toxoid. The results are consistent with atwo phase model in which a short antigen-driven phase of plasma cellgeneration sustains high antibody levels for 6 to 8 months. This isfollowed by a low level sustained plasma cell generation throughhomeostatic mechanisms that maintain for a lifetime constant antibodylevels proportional to the frequency of memory B cells. A mathematicalmodel of the serological response has been developed and will bediscussed in the context of vaccination strategies. Because of theircapacity of self renewal memory B cells represent a repository ofpotentially useful antibodies which have been selected in the course of

27

28

29

30

Page 55: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

55

(continued)

an immune response to human pathogens. We developed an improvedmethod of immortalization of human memory B cells and used it toisolate from a patient recovered from SARS coronavirus infection severalmonoclonal antibodies specific for different viral proteins, including 35antibodies with potent neutralizing activity. These results show that it ispossible to interrogate the memory repertoire of immune donors torapidly and efficiently isolate neutralizing antibodies which have beenselected in the course of natural infection.

The Use of MVA as a Vaccine Delivery Vector to Elicity ProtectiveImmune Responses Against PathogensH. RobinsonEmory University, Atlanta, GA

The development of an HIV/AIDS vaccine faces a number ofchallenges including the genetic diversity of the virus, the poor ability ofthe virus to be seen by neutralizing Ab, and the ability of the virus topersist in the presence of a strong immune response. Heterologousprime/boost immunization regimens have the potential for raising highlevels of both T cell and Ab responses. Working with collaborators atthe Emory Vaccine Center, the NIH and the CDC, we have developedsingle moiety Gag-Pol-Env expressing DNA and MVA vaccine vectors.These vaccines elicit a broad T cell response, good titers of anti-Env Aband have provided long term control of a virulent SHIV challenge in themacaque model. Macaques with controlled infections have stable low-levels of CD8 and CD4 T cells that co-produce IFN-_ and IL-2. Thevaccine is currently in early stage human trials.

Reference:1. Amara RR, Villinger F, Altman JD, Lydy SL, O’Neil SP, et al:

Control of a Mucosal Challenge and Prevention of AIDS in RhesusMacaques by Multiprotein DNA/MVA Vaccine. Science 292:69-74.Published online March 8, 2001; 0.1126/science.1058915.

Strategies for HIV Vaccine DevelopmentJ. ShiverMerck and Company, Inc, West Point, PA

Despite the critical need for a vaccine to prevent HIV-1 infection,development of an effective vaccine remains elusive. Research hasincreasingly focused on evaluating whether a vaccine based on elicitingT cell immunity against HIV-1 could control or prevent infection. This work is reflected in the large number of clinical trials currentlyevaluating vaccines that elicit cellular immunity against HIV-1. Wedeveloped a strategy for vaccine development comprised of twocomponents: (i) determine the best means for inducing cellularimmunity by comparing different vaccine approaches in animalimmunization studies; and (ii) identify which HIV-1 proteins are thebest antigens for T cell immune responses by studying cellular immuneresponses of HIV-infected subjects. Based on these studies DNA andreplication-defective adenovirus vector vaccines were selected for testingin clinical trials. The adenovirus vector will be tested in a proof-of-concept clinical trial to determine the effectiveness of this vaccine.Ongoing efforts also focus on developing an antigen that can elicitneutralizing antibodies against HIV-1. Such antibody responses shouldbe clinically relevant, i.e., able to potently neutralize diverse primaryviral isolates. Efforts have stalled on this objective throughout the HIV-

1 vaccine research field although numerous interesting researchstrategies are being devised and tested. Our current efforts towards thisobjective will be summarized.

Reference:1. Shiver JW, Emini EA. Recent advances in the development of HIV-1

vaccines using replication-incompetent adenovirus vectors. AnnuRev Med. 2004;55:355-372.

Development of Novel Vaccine Candidates Using DirectedMolecularEvolutionC. LocherMaxygen, Inc., Redwood City, CA

Directed molecular evolution strategies include DNA shufflingmethods, such as multi-gene and synthetic shuffling, to create largelibraries of genetically recombined molecules. Chimeric molecules fromthese libraries are then screened for improved characteristics. Moreeffective vaccines can be developed because chimeras are selected forimproved immunogenicity and increased reactivity to multiple antigenicvariants. Genetically recombined chimeric molecules can also haveincreased recombinant polypeptide expression through substitution ofhydrophilic/acidic amino acid residues. Moreover, increased viral growthkinetics (for improved inactivated or live attenuated viral vaccinemanufacturing) is another application. These technologies have beenused to create chimeric dengue virus vaccine antigens that generateneutralizing antibodies to all four serotypes; chimeric hepatitis B virussurface antigens with improved immunogenicity; chimeric HIV-1envelope antigens that induce neutralizing antibodies against multiplestrains of viruses; improved Venezuelan equine encephalitis virusenvelope vaccine antigens; and improved chimeric Plasmodiumfalciparum Erythrocyte Membrane Protein-1 (PfEMP-1) antigens thatinduce antibodies to multiple antigenic variants of the malaria parasite.Directed molecular evolution strategies have also been used to developmore potent adjuvants that are co-administered with vaccine candidates.Statistical analyses of multiple amino acid substitutions impartingimproved characteristics and repeated DNA shuffling may lead tofurther improvements of vaccine efficacy.

Reference:1. Locher, C.P., M. Paidhungat, R. Whalen, and J. Punnonen. 2005.

DNA Shuffling and Screening Strategies for Improving VaccineEfficacy. DNA and Cell Biology 24(4):256-263.

ChimeriVaxTh: A Novel Platform for Genetically Engineered Live Viral VaccineT. MonathAcambis, Inc., Cambridge, MA

A number of platform technologies for new vaccine developmenthave been explored, and a few have shown promise in clinical trials. One of the most promising approaches utilizes live, recombinant viruseswhich express one or more foreign genes encoding the antigen(s) ofinterest. We have used yellow fever (YF) 17D as a vector platform forcreating new recombinant vaccines against West Nile (WN), Japaneseencephalitis (JE), and dengue (DEN) viruses. Construction of the

31

32

33

34

Page 56: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

56

Eighth Annual Conferencevaccines involved infectious clone technology, in which the prM-E genesof YF 17D are replaced with the corresponding donor genes of thevaccine target. In the case of the WN and JE vaccines, mutationsinserted in the E gene were inserted to establish an improved attenuatedphenotype. The chimeric vaccines have reduced virulence compared toparental YF 17D vaccine, yet elicit strong neutralizing antibody levelsspecific to the donor gene. The vaccine candidates protect monkeysagainst challenge with wild-type JE, WN or dengue viruses.ChimeriVax™ vaccines against WN, JE and dengue have enteredclinical trials. In Phase 1 and 2 trials they have proven to be welltolerated and highly immunogenic, without restriction by preexistingimmunity to the vector (yellow fever). In addition a chimeric WNvaccine is in the late stages of development for protection of horses. Thepresentation will provide an update on the status of preclinical andclinical development of these new vaccines.

References:1. Lai CJ and Monath TP. Chimeric flaviviruses: novel vaccines against

dengue fever, tick-borne encephalitis, and Japanese encephalitis. InChambers TJ and Monath TP (eds). The Flaviviruses: Detection,Diagnosis, and vaccine Development. Adv Virus

2. Pugachev K, Monath TP, Guirakhoo F. Chimeric vaccines againstJapanese encephalitis, dengue and West Nile. In Levine M et al (Eds)New Generation Vaccines, 3rd Edit, Marcel Dekker, New York, 2004,pp. 559-71.

Page 57: On-Site Progr

on Vaccine Research

57

ABSTRACTS OFORAL SUBMITTED PRESENTATIONS

Page 58: On-Site Progr

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

58

Lipotechoic Acid Conjugate Vaccine for Staphylococcus A. Lees, J. KoKai-kun, A. LopezAcosta, J. Acevedo, J. Mond Biosynexus Inc, Gaithersburg, MD.

Staphylococcus is an opportunistic, Gram-positive bacterial genusthat is increasingly becoming antibiotic resistant. Lipotechoic acid(LTA) is a common and obligatory element of Gram-positivebacteria; no LTA deficient mutants are known. LTA from manyspecies have poly(glycerolphosphate) (PGP) as a common structuralelement, although it is often decorated with D-alanine andcarbohydrate. A protective antibody to S. epidermidis LTA, A110, isbeing evaluated in clinical trials for its ability to protect againstStaphylococcus bacteria. A110 crossreacts with LTA from bacteriacontaining PGP LTA but not Gram-positive bacteria that containnonPGP LTA. A110 also reacted with deacylated S. aureus LTA(DeAcLTA), which has been stripped of its pendant D-alanines andfatty acids. Both LTA and DeAcLTA are poorly immunogenic inmice. DeAcLTA was oxidized, functionalized with a mercapto-amino-oxy reagent and subsequently linked to maleimide derivatized-tetanus toxoid (TT). Due to the extreme negative charge on theDeAcLTA, the highest levels of coupling were obtained underrelatively acidic pH. Unlike the unconjugated antigen, the DeAcLTA-TT conjugate vaccine induced high levels of anti-LTA IgGantibodies. The response was boostable, indicating conversion of theDeAc LTA from a T-cell independent to a T-cell dependent antigen.The antibodies cross reacted with intact LTA. Furthermore, the serawere highly protective in an opsophagocytic assay against S.epidermidis bacteria. Because PGP is a common element of mostGram-positive bacteria, this conjugate vaccine has promise forproviding broad protection.

Adult Formulation Tetanus and Diphtheria Toxoids with AcellularPertussis Vaccine (Tdap) has Comparable Immunogenicity butLess Reactogenicity than DTaP-IPV for the Pre-school, Fifth-dose BoosterJ. M. Langley1, S. A. Halperin1, E. Mills2, A. Tomovici2, R. Guasparini3, G. Predy4, B. Law5, F. Diaz-Mitoma6, P. Whitsitt7, B. Tapiero8, M. Dionne9

1Dalhousie University, Halifax, NS, CANADA, 2Sanofi-Pasteur, Toronto, ON,CANADA, 3TASC Research, Surrey, BC, CANADA, 4Capital-Health, Edmonton,AB, CANADA, 5Univ-Manitoba, Winnipeg, MB, CANADA, 6CHEO, Ottawa, ON,CANADA, 7Paradigm Clin Trials, Oshawa, ON, CANADA, 8Hop-Ste-Justine,Montreal, PQ, CANADA, 9CHUQ, Beauport, PQ, CANADA.

Background: The pre-school (4-6 year), fifth-dose booster withDTaP-IPV is associated with increased injection site reactions,perhaps related to pertussis antigen content. Methods: Healthychildren age ≥ 4 to <7 years with completed primary series and 4thdose of DTaP-IPV-Hib (Pentacel™) were randomized to Tdapfollowed by IPV 4-6 weeks later, or DTaP-IPV (Quadracel™).Results: 593 children were enrolled at 8 sites. Safety endpoints days0-14 were less frequent in the Tdap group than the DTaP-IPV group:erythema (34.6 v. 51.7%), swelling (24.2 v. 33.8%), pain (39.6 v.67.2%) and fever (8.7 v. 16.9%). 100% (95% CI 98.6, 100) ofparticipants had seroprotective antibody levels to diphtheria andtetanus at 4-6 weeks (≥ 0.10 IU/ml). The Tdap v. DTaP-IPV 4-foldantibody rise was: pertussis toxoid 91.9% (95% CI 87.9, 94.9) v.96.8% (93.8, 98.6), filamentous hemagglutinin 88.1% (83.6, 91.8)v. 92.8 (88.9, 95.7), pertactin 94.3% (90.7, 96.7) v. 92% (88.0,95.1) and fimbriae 94.6% (91.2, 97.0) v. 87.6% (82.9, 91.5).

Pertussis and tetanus antibody GMTs were comparable; anti-diphtherialevels were lower after the lower diphtheria content Tdap. Conclusions:Tdap is less reactogenic than DTaP-IPV without inferiorimmunogenicity.

References:1. National Advisory Committee on Immunization. Canadian Guide to

Immunization. Ottawa, ON: Health Canada. 2002.2. Rennels MB. Pediatrics 2000; 105;e12.

An Antigen-Antibody Complex-based Therapeutic Vaccine forChronic Hepatitis B PatientsY. Wen1, D. Xu2, Z. Yuan1, K. Zhao3

1Department of Medical Molecular Virology, Shanghai Medical College, FudanUniversity, Shanghai, CHINA, 2Dept.Infectious Diseases, Di Tan Hospital,Beijing, CHINA, 3Beijing Institute of Biological Product, Beijingi, CHINA.

Yeast derived hepatitis B surface antigen complexed to anti-HBsproved effective in treating HBsAg positive transgenic mice. Thiscomplex increased uptake of HBsAg by antigen presenting cells andmodulated processing and presentation of HBsAg. In stage I phase Iclinical trial 22 healthy volunteers were vaccinated with three doses of30_g, 60_g and 90_g of HBsAg in the complex at 4-week intervals. Instage II phase I clinical trial, 9 volunteers received 90_g of HBsAg for 6injections. The renal, liver function and blood chemistry tests were allwithin the normal range and all recipients developed high titer of anti-HBs.. In phase IIA clinical trial, 36 chronic hepatitis B patients werevaccinated with 60_g , 90_g of HBsAg or adjuvant for six injections.Around 30% of the treated patients showed a 2-4 log decrease in serumHBV DNA and some were sero-converted from HBeAg strong positiveand anti-HBe negative to coexistence of anti-HBe positive and HBeAgweak positive. In two treated patients anti-HBs was detected, but withno significant changes in their HBeAg or serum HBV DNA status.Patients that had a decrease in serum HBV DNA showed “flares” intheir liver function assays, but returned to normal limits later. Whilethis therapeutic vaccine is effective for certain chronic hepatitis Bpatients, the contradictions and indications of this vaccine will bestudied.

References:1. Zheng BJ, Ng MH, He LF, et al. Therapeutic efficacy of hepatitis B

surface antigen-antibodies-recombinant DNA composite in HBsAgtransgenic mice. Vaccine 2001; 19:4219-4225.

2. Zheng BJ, Zhou J, Qu D, et al. Selective functional deficit indendritic cell-T cell interaction is a crucial mechanism in chronichepatitis B virus infection. J Viral Hepat 2004; 11:217-214.

Different Immune Response after Sequential Use of PneumococcalPolysaccharide and Pneumococcal Conjugate Vaccine A. de Roux1, B. Schmoele-Thoma2, N. Ahlers2, W. Gruber3, G. Siber3, D. Sikkema3, T. Welte4, H. Lode1

1Heliosklinik Emil-von-Behring, Berlin, GERMANY, 2Wyeth, Muenster, GERMANY,3Wyeth, Pearl River, NY, 4University Hannover, Hannover, GERMANY.

Background: Pneumococcal conjugate vaccine (7vPnC) in adultsmay complement the polysaccharide vaccine (23vPS). This studyexamined the immunogenicity of conjugate administered before or after

S1

S3

S2

S4

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

Eighth Annual Conference

Page 59: On-Site Progr

on Vaccine ResearchABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

59

(continued)

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

polysaccharide. Methods: 217 individuals aged ≥ 70 years withoutprior pneumococcal immunization were equally randomized in an openlabel study to receive either 7vPnC (Prevnar® 2µg saccharide/dose,except 4µg for 6B, Wyeth), or 23vPS (Pneumovax®23, 25µgsaccharide/dose, Aventis Pasteur MSD). One year later, 7vPnCrecipients received 23vPS and 23vPS recipients received 7vPnC. Bloodsamples were obtained prior to and one month post vaccination.Antibody responses (GMCs) were measured by ELISA. Results: Afterinitial 7vPnC, GMCs of all serotypes, except 19F (non-inferior), weresuperior to initial 23vPS. 7vPnC/ 23vPS recipients had higher GMCs(point estimates) compared to 23vPS alone. 23vPS/7vPnC recipientshad lower GMCs compared to 7vPnC alone.

Pneumococcal ELISA GMCs (µg/ml) with 95% CI, All AvailableImmunogenicity Population

Serotypes\Treatments N 4 6B 9V 14 18C 19F 23F

7vPnC 110 3.1 8.0 9.8 17.1 13.0 5.5 12.42.2, 4.3 6.0, 10.8 7.5, 12.8 12.3, 24.0 10.1, 16.7 4.1, 7.4 9.0, 17.0

7vPnC/23vPS 36 2.0 5.4 5.7 14.5 7.6 8.4 7.41.2, 3.5 3.3, 9.0 3.6, 8.9 8.8, 23.9 5.2, 11.1 5.3, 13.1 4.0, 13.6

23vPS 107 1.4 4.4 3.6 8.5 6.8 4.4 3.81.1, 2.0 3.4, 5.8 2.8, 4.6 6.0, 12.1 5.2, 8.9 3.4, 5.8 2.9, 5.0

23vPS/7vPnC 78 0.9 2.2 3.0 6.7 5.1 2.1 3.00.6, 1.3 1.5, 3.2 2.2, 4.0 4.5, 9.9 3.7, 6.8 1.5, 3.0 1.9, 4.8

Conclusion: In pneumococcal vaccine-naïve individuals aged ≥ 70years, 7vPnC induces a superior immune response to 23vPS for six ofseven 7vPnC serotypes and appears to increase antibody response tosubsequent 23vPS. However, initial 23vPS may inducehyporesponsiveness as measured by decreased antibody responses tosubsequent 7vPnC.

Reference:1. Kuhnke A, Lode H, De Roux A, Ahlers N, Thoma B, Madore D,

Welte T. Comparison of immunogenicity of pneumococcalconjugate (PnC) and 23-valent polysaccharide vaccine (23vPS) inelderly patients. ICAAC 2004; G-1675-2004.

Induction of Therapeutic Antitumor Immunity via ChemokineReceptor Mediated Antigen Cross-Presentation.A. Biragyn, D. Baatar, R. SchiavoLaboratory of Immunology, GRC, National Institute on Aging, Baltimore, MD.

We have recently established a novel strategy for rendering weaklyor non-immunogenic self tumor antigens immunogenic. The strategy isbased on use of proinflammatory chemokines to deliver antigens toimmature DCs through targeting chemokine receptors differentiallyexpressed on APCs. Herein, we report that chemokine receptormediated targeting is an efficient strategy to cross-present antigens toelicit potent CD4+ and CD8+ responses. The mechanism by which thefusions elicit responses is efficient uptake, processing and presentationof antigens via the MHC pathways. Experiments with inhibitors ofintracellular trafficking suggest that chemo-attractant fusion proteins,but not antigen alone, were processed and presented through early/lateendosomal and Golgi compartments, and stimulated antigen-specific Tcells both in vitro and in vivo. Moreover, our data indicate for the first

time that the chemokine receptor- targeted antigens are degraded byproteosomes and efficiently cross-presented to MHC class I. Thestrategy we developed is very simple and potent. Protein or DNAimmunizations elicit therapeutic antitumor immunity against widevariety of tumors, which express non-immunogenic or weaklyimmunogenic tumor antigens, such as a recently discovered embryonicantigen OFA. In addition, use of chemokines also allows induction ofcontrolled and polarized immune responses individually tailored for thespecific disease at will.

References1 Biragyn A, Tani K, Grimm MC, Weeks S, Kwak LW. Genetic fusion

of chemokines to a self tumor antigen induces protective, T-celldependent antitumor immunity. Nat Biotechnol 1999; 17(3):253-258.

2. Biragyn A, Ruffini PA, Coscia M, Harvey LK, Neelapu SS, Baskar S,Wang JM, Kwak LW. Chemokine receptor-mediated delivery directsself-tumor antigen efficiently into the class II processing pathway invitro and induces protective immunity in vivo. Blood 2004;104(7):1961-1969.

Directed Molecular Evolution Creates HIV-1 Novel gp120 VariantsThat Induce Broadly Neutralizing Antibodies in RabbitsL. Xu, X. Du, R. Whalen Infectious Diseases, Maxygen, Inc., Redwood City, CA.

Background: Attempts to develop a preventative vaccine to HIV-1capable of inducing neutralizing antibodies have been hindered by thelack of appropriate immunogens. We hypothesize that directedmolecular evolution can create novel Envelope variants that renderexisting neutralizing epitopes more immunogenic. Methods: In vitrohomologous DNA recombination was used to create novel chimericvariants of the Env protein from wild-type env sequences encoding cladeB gp120 and gp120 Core. Variants were characterized by the binding ofhuman monoclonal antibodies. Rabbits were immunized with plasmidDNA using electroporation followed by boosting with a heterologousadjuvanted gp120 protein. Sera and purified IgG were used to measureneutralization activity using a pseudovirus entry assay. Results:Chimeric variants created by in vitro recombination exhibited novelantigenicity, based on antibody binding, with respect to the parentalgenes from which they were derived. Analysis of the neutralizationactivity of antibodies induced by the variants using a panel ofpseudoviruses derived from primary clade B and non-clade B virusesshowed that gp120 variants presented improved neutralization activity.Neutralization activity induced by chimeric gp120Core sequences wasalso greater than that given by a parental gp120Core. Conclusion: Theseresults indicate that the use of directed molecular evolution can improvethe immunogenicity of the HIV-1 Env protein.

References:1. Mascola JR. Defining the protective antibody response for HIV-1.

Curr Mol Med 2003; 3:209-216.2. Locher CP, Soong NW, Whalen RG, Punnonen J. Development of

novel vaccines using DNA shuffling and screening strategies. CurrOpin Mol Ther 2004; 6:34-39.

S6

S5

Page 60: On-Site Progr

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

60

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

The Human Hookworm Vaccine Initiative (HHVI): Progress in the Product Development and Testing of the Na-ASP-2Hookworm VaccineM. E. Bottazzi1, J. Bethony1, S. Brooker2, G. Goud1, A. Loukas3, S. Mendez1, B. Zhan1, K. Stoever4, P. Hotez1

1Microbiology and Tropical Medicine, The George Washington University,Washington, DC, 2London School of Hygiene and Tropical Medicine, London,UNITED KINGDOM, 3Queensland Institute of Medical Research, Brisbane,AUSTRALIA, 4Human Hookworm Vaccine Initiative, Sabin Vaccine Institute,Washington, DC.

Background: Human hookworm infection, a leading cause of iron-deficiency anaemia and malnutrition, affects 740 million people indeveloping countries of the tropics. Currently, the major approach tohookworm control relies on frequent deworming with benzimidazoleanthelminthics. As complementary approach for hookworm control,the HHVI is developing a first-generation recombinant vaccine withthe goal to reduce hookworm disease burden. Methods: TheAncylostoma Secreted Protein-2 (ASP-2), an abundant protein secretedby infective hookworm larvae was selected on the basis of humanimmunoepidemiological studies and results from laboratory animalvaccinations. Furthermore, it was shown that anti-ASP-2 inhibitshookworm larval invasion in vitro. A product development strategy tomanufacture and test ASP-2 was generated in preparation for initialsafety studies in humans. Results: The antigen from Necatoramericanus, Na-ASP-2, was cloned and expressed in the yeast Pichiapastoris. Process development, assay development and qualification,cGMP pilot lot manufacture, toxicity and stability studies of the alum-formulated Na-ASP-2 Hookworm Vaccine have been completed.Conclusions: Vaccination against hookworm would alleviate the publichealth deficiencies of drug treatment alone. After an initial Phase 1study, the the clinical development of Na-ASP-2 Hookworm Vaccinewill include pilot efficacy studies in Brazil and Africa to evaluateclinical endpoints unique to helminth infections.

References:1. Hotez PJ, Brooker S, Bethony JM, Bottazzi ME, Loukas A, Xiao S.

Hookworm infection. N Eng J Med 2004; 351(8):799-807.2. Hotez PJ, Zhan B, Bethony J, et al. Progress in the development of a

recombinant vaccine for human hookworm disease: the humanhookworm vaccine initiative. Int J Parasitol 2003; 33(11):1245-1258.

The Human Hookworm Vaccine Initiative (HHVI): Novel Designand Statistical Considerations to Estimate the Efficacy of aHelminth Vaccine in Field Trials in Endemic Region: Phase 2bStudies for a Human Hookworm Vaccine.J Bethony1, S Brooker2, N Alexander2, S Geiger1, L Rodrigues3, ME Bottazzi1, K Stoever3, P Hotez1

1The George Washington University, USA, 2 London School of Hygiene andTropical Medicine, UK, 3 Sabin Vaccine Institute, USA

Background: An understanding of the epidemiology andtransmission dynamics of helminth infection is important fordetermining the design, evaluation, and ultimate use of a hookwormvaccine. Hookworms do not replicate within their hosts and, as such,the traditional estimates of vaccine efficacy, which are based on thecomparison of incidence rates of infection and/or disease in vaccinatedand unvaccinated populations, are not appropriate. Furthermore, the

clinical hallmark of hookworm disease is iron deficiency anemia, whichis influenced by numerous other factors (e.g., background nutritionalstatus, co-infections, etc). Finally, the marked heterogeneity ofhookworm infections in endemic areas, including over-distribution ofinfection in community (such that a minority of people harbor amajority of the infection), marked geographic variation, age-related andgender-related differences in prevalence and intensity of infection arefurther obstacles in designing and evaluating the efficacy of ahookworm vaccine.

Methods: Worm burden is a key determinant of hookwormmorbidity and transmission dynamics. Hence, a reduction in wormburden is an appropriate measure of vaccine efficacy. In this context,vaccine efficacy has been defined as 1-(AWV/AWU), where AWU andAWV are the average (arithmetic mean) worm burdens in theunvaccinated and vaccinated, respectively. An efficacy of 50% impliesthat vaccinated individuals have 50% fewer worms than unvaccinatedindividuals. In addition to these parasitologic outcomes, the impact ofthe vaccine on clinical outcomes also requires evaluation. Themorbidity from hookworm disease is directly related to intestinal bloodloss caused by the adult hookworms. Diminished worm burden andfecal egg counts should be expected to result in reductions in hostintestinal blood loss, with the latter determined by quantitative fecalheme measurements. The primary sample size calculation had theobjective of detecting a vaccine efficacy of 30%; 1211 and 1730 eggsper gram in the unvaccinated and vaccinated arms, respectively. Totake account of the highly skewed distribution of the egg counts, thiscalculation used the negative binomial distribution, and yielded 505people per arm, or 632 after allowing for losses to follow-up. Sincefecal heme loss has been found to be approximately proportional to eggcount, the above efficacy implies that mean fecal heme will also bereduced by 30%. Fitting a gamma distribution to existing data fromthe prospective field site, we estimated our power to detect such aneffect to be 98%. Results: We have designed a double-blind,randomized placebo controlled Phase 2b proof of concept trial toestimate of the efficacy of the Na-ASP-2 Hookworm Vaccine inreducing intensity of re-infection with hookworm in an adulthookworm infected populations from endemic areas of Brazil.Additional objectives are to investigate the effect of the vaccine on theclinical outcomes of hookworm disease by measuring using intestinalblood loss (fecal heme). Conclusions: This vaccine brings to bear anumber of innovative clinical trial design features. Including (a)measuring intensity of infection rather than incidence of disease; (b)using a treatment and reinfection design to determine the efficacy ofthe vaccine; (c) sampling across numerous geographic regions to avoidinterruption of transmission; (d) using fecal heme as a biomarker forthe influence of hookworm infection on IDA; and (e) novel statisticalapproaches to deal with the over-dispersion of many of the parametersof hookworm infection.

References:1. Brooker S, Bethony J, Rodrigues L, Alexander N, Geiger S, Hotez P.

Epidemiologic, immunologic and practical considerations indeveloping and evaluating a human hookworm vaccine. Expert Rev Vaccines 2005; 4(1):35-50.

2. Hotez PJ, Brooker S, Bethony JM, Bottazzi ME, Loukas A, Xiao S. Hookworm infection. N Engl J Med 2004; 351(8):799-807.

S7

S8

Eighth Annual Conference

Page 61: On-Site Progr

on Vaccine ResearchABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

61

(continued)

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

Safety and Immunogenicity of Vaccines against Cholera andEnterotoxigenic Escherichia coli Diarrhea in Children In Bangladesh-Problems Encountered and Milestones AchievedF. Qadri1, D. A. Sack2

1Laboratory Sciences Division, ICDDR,B, Dhaka, BANGLADESH, 2ICDDR,B,Dhaka, BANGLADESH.

Background. In Bangladesh, the two major bacterial pathogenswhich contribute to the large burden of secretory diarrheal disease areVibrio cholerae O1 and enterotoxigenic Escherichia coli (ETEC). Cholerais more common in those more than two years of age and ETECinfections in children less than three years of age. The vaccines tested atthe ICDDRB include killed and live oral vaccines for cholera and a killedoral vaccine for ETEC. Methods. Oral vaccines were the whole cellkilled ETEC and cholera (SBL) as well as the live attenuated choleravaccine, Peru-15 (AVANT) which were tested in adults and children inBangladesh. Results. The ETEC vaccine was found to be safe andimmunogenic in adults and children up to 18 months of age (n=300).In infants, the full dose resulted in vomiting, requiring a reduction to aquarter dose. The reduced dose appeared safe and immunogenic (n=200)suggesting that this dose may be effective in primed populations. Inprevious field studies the oral killed whole cell cholera vaccine provedefficacious, but protection waned more quickly in children, howeversupplementation with zinc increased its immunogenicity (n=240). Analternative strategy is a single dose, live, vaccine candidate, Peru-15. Thisvaccine proved to be both safe and immunogenic in adults (n=70) andchildren (n=120). Conclusions. Oral vaccines for cholera and ETEC aresafe and immunogenic in adults in children in Bangladesh and furtherstudies are needed to establishtheir efficacy, effectiveness and usefulnessin this region.

References:1. Firdausi Q, Tanvir A, Firoz A, et al. Safety and immunogenicity of an

oral, inactivated enterotoxigenic Escherichia coli plus cholera toxin Bsubunit vaccine in Bangladeshi children 18-36 months of age.Vaccine 2003; 21:2394-2403.

2. Albert MJ, Qadri F, Wahed MA. et al. Supplementation with zinc,but not vitamin A, improves seroconversion to vibriocidal antibodyin children given an oral cholera vaccine. J Infect Dis 2003; 187:909-913.

Mass Vaccination Against Shigellosis: First Experience of RoutineImmunization Against Shigella sonnei InfectionR. P. Chuprinina1, L. I. Pavlova1, T. I. Frolushkina1, A. V. Protodiakonov1, T. V. Gantcho2, M. E. Golovina2, V. I. Shmigol2, S. I. Elkina2, I. Y. Kurbatova2, V. L. Lvov2, P. G. Aparin2 1Carbohydrate Vaccines, Ministry of Health Russia, Moscow, RUSSIANFEDERATION, 2Carbohydrate Vaccines, NRC-Institute of Immunology, Moscow,RUSSIAN FEDERATION.

Newly-developed low-endotoxic lipopolysaccharide Shigella sonneivaccine SHIGELLVAC protected civic population from natural infectionin endemic region with efficacy rate higher than 90% (phase III clinicaltrials). The vaccine was officially approved for routine immunization ofchildren older than 3 years old and adults by MOH Russia in September2003. The strategy of routine immunization against food-borneshigellosis was formulated based on the vaccine characteristics andepidemiological background of disease distribution. Approximately100,000 persons from 3 to 70 years were single immunized with Shigella

sonnei in the framework of the vaccination program initiated in theCentral, Volga, Northern Caucasus, Black Sea, Ural, and Yakutiya regionsof the Russian Federation. Chromatography-grade quality vaccineSHIGELLVAC was well tolerated by vaccinees in different age groups.Personal of food factories, food-handlers, and the staff of canteens andcafés represented the immunized contingent. Among them specialattention was paid to immunization of the staff of milk factories and milkfarms. Another aim of routine immunization was immunization of thestaff of kindergartens and children’s summer camps, especially workersconnected with food processing. None of 60 food handlers immunized bySHIGELLVAC was infected during a large shigellosis outbreak inKrasnoturiinsk (mid-Ural area). Three months after vaccination,immunized individuals demonstrated GM IHA titers of 1:1800, whichwas dramatically higher than titers observed after natural infection.

Reference:1. Aparin PG, Golovina ME, Ershov VI, Gancho TV, Shmigol VI,

Pavlova LI, Chuprynina RP, Yolkina SI, Rachmanov RS, Lvov VL.Systemic and mucosal (local) immune response after immunizationwith new type low-endotoxic LPS Vaccine Shi

Randomized, Double-blind Phase I Study to Assess the Safety,Tolerability, Imunogenicity, Dose Response, and Transmissibility ofCVD 1208S, a guaBA, sen, and Set Deleted, Live, Oral Shigellaflexneri 2a Soy Based Vaccine.K. L. Kotloff1, J. K. Simon1, M. Pasetti1, J. P. Nataro1, M. B. Sztein1, S. S. Wasserman2, W. C. Blackwelder1, E. M. Barry2, M. M. Levine1

1Pediatrics, University of Maryland, Baltimore, MD, 2Medicine, University ofMaryland, Baltimore, MD.

Background: CVD1208S is a live, oral, S. flexneri 2a vaccine whichhas been attenuated by deleting locus guaBA encoding two enzymesintegral to guanine nucleotide biosynthesis as well as set and sen, whichencode Shigella enterotoxin (ShET)1 and ShET2. CVD 1208S wasconstructed on animal-free media to address regulatory concerns aboutpossible contamination with agents that cause Bovine SpongiformEncephalopathy (BSE). Methods: Healthy inpatient adult volunteerswere randomized in a double-blind fashion to receive one oral dose ofCVD 1208S (108 or109 CFU, n=7 per dosage) or placebo (n=2).Clinical, immunologic, and microbiologic responses were evaluated.Results: One vaccine recipient (6%) experienced a brief (4 hour) episodeof low grade fever and another (6%) experienced mild diarrhea. Anti-LPSresponses as measured by antibody secreting cells, serum, or fecalantibody, occurred in 57% of 108 CFU and 100% of 109 CFUrecipients. Over 86% of high dose vaccinees shed the vaccine strain for 2or more days. Transmission to placebo recipients was not detected.Conclusions: CVD 1208S is well tolerated, immunogenic, easilyeradicated, and not transmitted to placebo recipients during a 10 dayinpatient stay. CVD1208S is a promising Shigella vaccine candidatesuitable for outpatient phase II testing.

References:1. Kotloff, KL, Pasetti MF, Barry EM, et al. Deletion in the Shigella

enterotoxin genes further attenuates Shigella flexneri 2a bearingguanine auxotrophy in a phase 2 trial of CVD 1204 and CVD 1208.J Infect Dis 2004; 190(10):1745-1754.

2. Kotloff KL, Noriega FR, Taraz Samandari T, et al. Shigella flexneri 2astrain CVD 1207, with specific deletions in virG, sen, set, and guaBA,is highly attenuated in humans. Infect Immun 2000; 68(3):1034-1039.

S9

S10

S11

Page 62: On-Site Progr

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

62

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

Phase 1 Study of the Safety and Immunogenicity of Amai-C1/Alhydrogel® Vaccine for Plasmodium falciparum Malaria inSemi-Immune Malian AdultsA. Dicko1, D. Diemert2, I. Sagara1, M. Sogoba1, M. Niambele1, M. Assadou1, O. Guindo1, B. Kamate1, M. Baby1, M. Sissoko1, G. Mullen2, E. Malkin2, M. Sissoko1, M. Thera1, A. Dolo1, C. Long2, D. Diallo1, L. Miller2, A. Saul2, O. Doumbo1

1MRTC/University of Bamako, Bamako, MALI, 2MVDB/NIAID, Rockville, MD.

Apical membrane antigen-1 (AMA1) - a leading malaria vaccinecandidate - is a surface protein expressed during the asexual blood stageof P. falciparum that has been implicated in parasite invasion oferythrocytes. AMA1-C1/Alhydrogel® consists of an equal mixture ofAMA1 from the FVO and 3D7 clones of P. falciparum, producedseparately as recombinant proteins expressed by Pichia pastoris, thenmixed and adsorbed onto Alhydrogel®. In a double-blinded Phase 1study, 54 healthy Malian adults were enrolled into one of three dosecohorts (n=18 per cohort) and randomized 2:1 to receive either AMA1-C1/Alhydrogel® or Recombivax® hepatitis B vaccine. The first,second and third cohorts were vaccinated successively at three-weekintervals, and received 5, 20 and 80 mcg of AMA1-C1, respectively. 53subjects received both of 2 scheduled immunizations on Days 0 and 28,and all cohorts have completed 180 days of follow-up. No vaccine-related serious or grade 3 adverse events have been observed. Allinjection site reactions were mild in severity, whereas systemic reactionswere mild to moderate in severity. Anti-AMA1 antibody responses arebeing measured by ELISA on sera collected at pre- and post-vaccinationtime-points. The data so far demonstrate that AMA1-C1/Alhydrogel®has an excellent safety profile. Full safety and immunogenicity resultswill be reported.

Reference:1. Stowers AW, Kennedy MC, Keegan BP, Saul A, Long CA, Miller LH.

Vaccination of monkeys with recombinant Plasmodium falciparumapical membrane antigen 1 confers protection against blood-stagemalaria. Infect Immun 2002; 70(12):6961-6967.

Oculo-respiratory Syndrome (ORS) and Other Adverse EventsFollowing Immunization (AEFIs) in Infants and Toddlers GivenInfluenza VaccineD. M. Skowronski1, S. A. Tweed1, V. Remple1, K. Pielak1, J. Daigneault2, P. Daly3, G. Arsenault4, E. Galanis1, T. Tam5

1BC Centre for Disease Control, Vancouver, BC, CANADA, 2Direction de SantePublique, Chicoutimi, PQ, CANADA, 3Vancouver Coastal Health Authority,Vancouver, BC, CANADA, 4Fraser Health Authority, Surrey, BC, CANADA, 5PublicHealth Agency of Canada, Ottawa, ON, CANADA.

Background: ORS following influenza vaccine (FV) was firstreported in Canada in 2000-2001. The rate of ORS in children in 2000(10-15%) was highest among first-time vaccinees (24%)[1]. In 2004-2005, expert groups recommended influenza vaccination for childrenaged 6-23 mos for the first time. We assessed ORS and other AEFIsamong infants/toddlers receiving FV in Quebec (QC) and BritishColumbia (BC), Canada in fall, 2004. Methods: Study consent wasobtained from parents bringing their infant/toddler to public clinics forinfluenza immunization. Telephone interviews were conducted 6-10days later using a standard questionnaire. Results: 320 children in QCand 370 in BC participated; first-time vaccinees accounted for 307(96%) and 291 (79%), respectively. The same lot number was

administered to 87%, overall. The rate of ORS was 18/320 (5.6%;95%CI 3.6%-8.7%) and 3/370 (0.8%; 95%CI 0.3%-2.4%) in QC andBC, respectively. 95% of parents would immunize their child again ifthe vaccine was free. Other AEFIs occurring within 72 hrs ofvaccination included fussiness (19%; 95%CI 16-22%), fever (11%;95%CI 9-14%), drowsiness (7%; 95%CI 5-9%) and vomiting (2%;95%CI 1-4%). Conclusions: Influenza vaccine was well-tolerated. Thefrequency of ORS was low and consistent with the background rate ofacute respiratory illness in this agegroup[2]. Differences in rates ofORS between QC/BC may relate to interview approach or distributionof background confounders (infection/allergy/asthma).

References:1. Skowronski DM, Bjornson G, Husain E, Metzger DL, Scheifele

DW. Oculorespiratory syndrome after influenza immunization inchildren. Pediatr Infect Dis J. 2005; 24:63-69.

2. Monto AS, Ullman BM. Acute respiratory illness in an Americancommunity : the Tecumseh study. JAMA 1974;227:165-169.

Improved Protective Antibody Responses Were Induced by CodonOptimized DNA Vaccines Expressing Hemagglutinin Antigens ofInfluenza H1 and H3 SerotypesS. Wang1, I. Mboujka1, J. Haran1, H. Cao1, X. Huang1, J. Taaffe2, A. Solórzano2, A. García-Sastre2, S. Lu1

1Department of Medicine, University of Massachusetts Medical School,Worcester, MA, 2Department of Microbiology, Mount Sinai School of Medicine,New York, NY.

Background: Influenza viral infection (flu) remains an importantpublic health issue. Alternative vaccination strategies are needed toimprove the quality of immune responses and to overcome thelimitation of the manufacturing process for the current inactivated fluvaccines. Early generations of flu DNA vaccine, while protective insmall animal models, could not elicit detectable antibody responsesprior to viral challenge. We have developed codon optimized Flu HAgenes to improve the immunogenicity of Flu DNA vaccines. Methods:Codon optimized HA inserts for A/H1N1/NewCal/20/99 (H1) andA/H3N2/Panama/2007/99 (H3) were produced and individuallysubcloned into DNA vaccine vector pSW3891. The HA gene insertswere designed to express either cell associated or secreted forms of HAantigens. New Zealand White rabbits received 4 bi-weekly gene gunimmunizations. Rabbit sera were collected for antibody analyses.Results: Codon optimized HA DNA vaccines elicited significant levelsof anti-HA IgG responses with strong serotype specificity asdetermined by ELISA and Western blot. Co-delivery of HA DNAvaccines from two serotypes expanded the breadth of anti-HA IgGresponses to include both serotypes. Significant levels of HI activitieswere also observed with codon optimized HA DNA vaccines.Conclusions: Codon optimization was able to improve theimmunogenicity of influenza DNA vaccines and it offers an alternativevaccination strategy to the current flu vaccines.

Reference:1. Palese P, Garcia-Sastre A. Influenza vaccines: present and future. J

Clin Invest 2002; 110:9-13.

S12

S13

S14

Eighth Annual Conference

Page 63: On-Site Progr

on Vaccine ResearchABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

63

(continued)

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

Protective Measures and Human Antibody Response to HPAIH7N3 in British Columbia (BC), CanadaD. M. Skowronski1, Y. Li2, S. A. Tweed1, T. Tam3, M. Petric1, S. Berger1, A. Larder4, N. Bastien2, A. King3, R.C. Brunham1

1BC Centre for Disease Control, Vancouver, BC, CANADA, 2Public Health Agencyof Canada, Winnipeg, MB, CANADA, 3Public Health Agency of Canada, Ottawa,ON, CANADA, 4Fraser Health, Abbotsford, BC, CANADA.

Background: In spring,2003 a poultry outbreak of highly-pathogenic-avian-influenza (HPAI)-H7N7 in the Netherlands caused89 human infections. Modified hemagglutination inhibition (HI) assaydetected antibody in >50% of 500 potentially-exposed persons[1]. Inspring,2004 an HPAI-H7N3 poultry outbreak in BC, Canada causedtwo human infections. We report compliance with protective measuresand sero-survey for additional cases in BC. Methods: All sera fromvolunteers among those potentially exposed to AI-infectedpoultry/products in BC were tested by microneutralization (MN) assay.Sera from two confirmed human infections, a random sample of tenparticipants and four persons with unprotected exposure to AI-infectedbirds were also tested by Western blot, routine and modified HI (usinghorse erythrocytes)[2]. Results: 167 persons participated between May7 and July 26, 2004. Of 91 persons who handled or shared confinedairspace with AI-infected poultry, 21 (23%) experiencedconjunctivitis/influenza-like illness. Among 65 persons present in barnswith AI-infected birds and sawdust, 85% received influenza vaccine,74% took oseltamavir and 85%, 83% and 55% always wore gloves,mask or goggles, respectively. All assays were negative for antibody toH7. Conclusions: Improved compliance with protective measures,especially goggles, is needed during poultry outbreaks. Possible reasonsfor fewer infections compared to the Netherlands include: lowerpathogenicity of H7N3, better compliance with protective measuresand/or sampling/testing considerations.

References:1. Bosman A, Mulder YM, de Leeuw JRJ, et al. Executive summary

avian flu epidemic 2003: public health consequences. RIVM rapport630940003. Bilthoven:RIVM; 2004.http://www.rivm.nl/bibliotheek/rapporte

2. Stephenson I, Wood JM, Nicholson KG, Zambon MC. Sialic acidreceptor specificity on erythrocytes affects detection of antibody toavian influenza hemagglutinin. J Med Virol 2003; 70:391-398.

T Cell Multi-Epitope Vaccine for Pandemic InfluenzaJ. Alexander1, B. Stewart1, P. Bilsel1, J. Katz2, M. Newman1

1Epimmune, San Diego, CA, 2Centers for Disease Control and Prevention,Atlanta, GA.

The goal of our project is to develop vaccines for use againstpandemic strains of influenza virus. Vaccine design was based on use ofmultiple sequence conserved T cell (CTL and HTL) epitopes. Vaccinesthat induce T-cell responses can be designed and producedprospectively and used early in a pandemic, prior to availability ofstrain-matched vaccines which induce antibody responses. Epitopeswere identified using sequences of influenza virus strains which arepotential components of a pandemic influenza virus including agents ofpast pandemics and zoonotic influenza infections of man. An effectiveprocess based on motif search algorithms and HLA-peptide bindingassays was employed to identify epitopes that are restricted by multipleHLA types and with predictable immunogenicity levels. Confirmation

that epitopes were processed and presented to the immune systemduring natural infection was tested using in vitro recall responses asmeasured using PBMC from normal donors, peptide pools andELISPOT IFN-_ assays. In our initial studies, 19 of 39 HLA-A2-restricted peptides induced measurable responses in the range of 10-6,000 spot forming cells/106 PBMC. These preliminary datademonstrate that virally conserved CTL epitopes can be identified andsupport our belief that a vaccine designed for use in a “first response”mode during the early stage of a pandemic is feasible.

References:1. Seo SH, Peiris M, Webster RG. Protective cross-reactive cellular

immunity to lethal A/Goose/Guangdong/1/96-like H5N1 influenzavirus is correlated with the proportion of pulmonary CD8+ T cellsexpressing gamma interferon. J Virol 2002; 76:4886-4890.

2. Webby RJ, Andreansky S, Stambas J, Rehg JE, Webster RG, DohertyPC. Protection and compensation in the influenza virus-specificCD8+ T cell response. Proc Natl Acad Sci U S A

2003;100:7235-7240.

Immunization with Salmonella enterica serovar Typhi Ty21aExpressing Anthrax Protective Antigen Protects Mice from anAnthrax Lethal Toxin ChallengeY. Wu, M. Osorio, S. Bhattacharyya, M. D. Bray, R. Walker, D. J. KopeckoFDA-CBER, Bethesda, MD.

The current FDA-approved anthrax vaccine is an aluminumhydroxide-adsorbed, formalin-treated culture supernatant of toxigenic,nonecapsulated strain of B. anthracis. The principal protectivecomponent of this vaccine is the protective antigen (PA) protein.Although the vaccine is safe and efficacious, it does have somelimitations such as a high index of local and systemic reactions, it isadministered subcutaneously, and requires multiple doses to stimulateimmunity. In an attempt to design an improved anthrax vaccine for easyemergency immunization, we have chosen to test the feasibility of usingan attenuated Salmonella serovar typhi strain (Ty21a) expressing PA toinduce an immune response to PA and thus protect against anthraxinfection. In these studies, the anthrax PA was cloned behind anenvironmentally regulated promoter in a genetically stable low copyplasmid vector, pGB-2. Preliminary mouse studies showed that the livevector expressing PA elicited a robust immune response measured byPA-specific IgG ELISA titers. Antibody titers were significantlyincreased when the live vector expressed an extracellularly secreted formof PA. A challenge of immunized mice with approximately 5 LD50doses of anthrax lethal toxin (given i.v.) showed that vaccine constructsinduced protective immunity in 75 % of vaccinated animals. Ty21a hasbeen administered to greater than 25 million people and proven to besafe. Thus, a live attenuated oral vaccine such as Ty21a expressing PAcould form the basis of a safe and effective vaccine against anthrax

References:1. Pitt ML, Little S, Ivins BE, Fellows P, Boles J, Barth J, Hewetson J,

Friedlander AM. In vitro correlate of immunity in an animal model ofinhalational anthrax. J Appl Microbiol 1999; 87(2): 304.

2. Xu DQ, Cisar JO, Ambulos Jr N Jr, Burr DH, Kopecko DJ.Molecular cloning and characterization of genes for Shigella sonneiform I O polysaccharide: proposed biosynthetic pathway and stableexpression in a live salmonella vaccine vector. Infect Immun 2002;70(8):4414-4423.

S15

S17

S16

Page 64: On-Site Progr

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

64

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

Assessment of the Reactogenicity of Tdap in Children andAdolescents 7-19 Years of Age by Interval Since Prior Tetanus andDiphtheria Toxoids Containing VaccineS. A. Halperin1, L. Sweet2, D. Baxendale1, A. Neatby2, P. Rykers1, B. Smith1, M. Zelman2, D. Maus3, P. Lavigne3, M. Decker3

1Dalhousie University, Halifax, NS, CANADA, 2Department of Health and SocialServices, Charlottetown, PE, CANADA, 3Sanofi Pasteur, Toronto ON, CANADA,and Swiftwater, PA, USA

Background: The incidence of pertussis is increasing amongstadolescents and adults, despite widespread immunization prior toschool entry. Adult-formulation diphtheria and tetanus toxoids andacellular pertussis vaccines (Tdap) have been developed to preventpertussis in adolescents and adults. Methods: To evaluate whetherrecent receipt of diphtheria-tetanus-toxoid-containing-vaccine(TD/Td) might contraindicate receipt of Tdap, we performed an open-label, province-wide, clinical trial comparing the reactogenicity of Tdap(Adacel‘, Sanofi-Pasteur) given 2-9 years versus ≥10 years after previousTD/Td. Results: 7001 children/adolescents were enrolled in the study(464-963/interval cohort); adverse event diaries were completed by5926 (85%). No Arthus reactions or serious adverse events related tovaccination were reported. No differences in reports of fever werefound by interval since last immunization. Injection-site erythema andswelling were modestly increased among those with most recent priorTD/Td. Compared to the 10-year-interval group, the maximumincrease for any other group was £8.4% for any erythema, £6% formoderate/severe erythema, £10.2% for any swelling, £6.9% formoderate/severe swelling, £5.1% for any pain, and £3.5% formoderate/severe pain. Conclusion: Although there is a modest increasein injection-site events with decreasing interval since a previousimmunization, Adacel™ (Tdap) can be safely administered at intervalsof ≥2 years since a previous TD/Td vaccine.

References: 1. National Advisory Committee on Immunization. Prevention of

pertussis in adolescents and adults. Can Commun Dis Rep 2003;28:ACS-5,6.

2. Rennels MB, Deloira MA, Pichichero ME, et al. Extensive swellingafter booster doses of acellular pertussis-tetanus-diphtheria vaccines.Pediatrics 2000; 105(1):e12.

Oral Vaccine Delivery by Salmonella Vaccine VectorsM. E. Gahan1, D. E. Webster2, S. L. Wesselingh2, B. B. Finlay3, R. A. Strugnell4

1Macfarlane Burnet Institute & Department of Medicine, Monash University,Melbourne, AUSTRALIA, 2Macfarlane Burnet Institute, Melbourne, AUSTRALIA,3Michael Smith Laboratories, University of British Columbia, Vancouver, BC,CANADA, 4Department of Microbiology and Immunology, University ofMelbourne, Melbourne, AUSTRALIA.

Background: Salmonella enterica is an ideal candidate for oraldelivery of vaccine antigens. We report on the effects of plasmid vectorand Salmonella strain on the immune response to the C-fragment oftetanus toxin (Cfrag). Methods: Cfrag (+pCMV) was ligated into theplasmids pcDNA3, pAT153, pBBR122, pBR322, pRSF1010, pUC18and pACYC184 and transformed into _aroAD Salmonella. In vitro andin vivo plasmid stability was examined and correlated with Cfragantibody response. Additional Salmonella Pathogenicity Island (SPI)mutations (ssrA, invA and sifA) were transduced into _aroAD. Organcounts and Cfrag antibody titres were determined. Results:

pAT153/Cfrag was the most stable plasmid in vitro and in vivo.Although all plasmids elicited similar antibody responses to SalmonellaLPS, only pAT153/Cfrag induced a Cfrag antibody response. At day 28all SPI mutants were able to colonise, however, _aroAD(ssrA) wassignificantly reduced in the Peyer’s patches compared with _aroAD(p=0.03). Only 2/5 (day 28) and 1/5 (day 56) mice immunised with_aroAD(ssrA) produced a Cfrag antibody response. At day 56 _aroADhad significantly higher Ig and IgG1 Cfrag titres than all SPI mutants(p<0.03).Conclusion: Our research shows attenuated Salmonella enterica can beused to orally deliver vaccine antigens. However, consideration must begiven to the plasmid vector and strain of Salmonella used as these impacton the response to the vaccine.

Reference:1. Dunstan, SJ, Simmons CP, Strugnell RA. In vitro and in vivo stability

of recombinant plasmids in a vaccine strain of Salmonella enterica var.Typhimurium. FEMS Immunol Med Microbiol 2003; 37(2-3):111-119.

CpG Oligodeoxynucleotides Co-administered With the MicronemeProtein MIC2 Protect Against Eimeria InfectionsR. A. Dalloul1, H. S. Lillehoj1, D. M. Klinman2, X. Ding1, W. Min1, R. A. Heckert1, E. P. Lillehoj3

1Animal Parasitic Diseases Laboratory, USDA, Beltsville, MD, 2Section ofRetroviral Immunology, Center for Biologics Evaluation & Research, US FDA,Bethesda, MD, 3Department of Pediatrics, School of Medicine, University ofMaryland, Baltimore, MD.

Short synthetic oligodeoxynucleotides containing unmethylatedCpG motifs (CpG ODNs) were previously shown to exert a positiveeffect on weight loss and oocyst shedding associated with Eimeriainfection when injected in vivo. The present work investigated theeffects of in ovo vaccination with CpG ODNs and an Eimeriarecombinant microneme protein (MIC2), alone or in combination, onchicken susceptibility to coccidiosis. In ovo injection of CpG ODNsalone enhanced resistance to experimental E. acervulina infection as bestexemplified by reduced oocyst shedding. Two CpG ODNs reduced theoocyst load, but did not affect weight gain. When co-administered withthe recombinant microneme protein, both ODNs reduced oocystshedding; however, only ODN D19 plus MIC2 consistently improvedweight gain. Vaccinating with ODN 2006 or MIC2 protein curtailedoocyst shedding but did not enhance weight gain in E. tenella-infectedbirds. Co-administration of CpG ODN and MIC2 did not have anadditive effect in reducing the oocyst output; however, it resulted in thehighest and lowest Ab response before and after E. tenella infection,respectively. Collectively, CpG ODNs administered in ovodemonstrated immunoenhancing and adjuvant effects following Eimeriainfections.

References:1. Dalloul RA, Lillehoj HS, Klinman DM, et al. In ovo administration

of CpG oligodeoxynucleotides and the recombinant micronemeprotein MIC2 protects against Eimeria infections. Vaccine 2005; inpress.

2. Klinman DM. Immunotherapeutic uses of CpGoligodeoxynucleotides. Nat Rev Immunol 2004; 4:249-259.

S18

S19

S20

Eighth Annual Conference

Page 65: On-Site Progr

on Vaccine ResearchABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

65

(continued)

Immunogenicity of Combination DNA Vaccines for RVFV, TBEV,HTNV, and CCHFVK.W. Spik, A. Shurtleff, A.K. McElroy, M.C. Guttieri, J. W. Hooper, and C.SchmaljohnVirology Division, United States Army Medical Research Institute of InfectiousDiseases, Fort Detrick, Frederick, MD 21702

Background: Rift Valley fever virus (RVFV), Crimean Congohemorrhagic fever virus (CCHFV), Hantaan virus (HTNV), and tick-borne encephalitis virus (TBEV) cause serious infections in humans.Licensed vaccines against these pathogens are not currently available.Methods: We constructed DNA vaccines for RVFV and CCHFV bycloning cDNA representing M segment coding regions into a plasmidwith a CMV promoter. We vaccinated mice by gene gun with thesevaccines alone or combined with previously developed DNA vaccinesfor TBEV (1) and HTNV (2). We measured antibody responses byimmune precipitation of proteins and by neutralization assays. Results:The TBEV vaccine and a RVFV vaccine encoding the envelopeglycoproteins elicited neutralizing antibodies and afforded protectiveimmunity when delivered alone or combined with other DNA vaccines.Although low levels of antibodies were elicited with the HTNV orCCHFV vaccines, no major differences were observed when they weredelivered alone or combined with other DNA vaccines. Conclusions:These data suggest that combination DNA vaccines for these agents arepossible, but further studies in other animal models and with otherdelivery methods are desirable.

References:1. Schmaljohn C, Custer D, VanderZanden L, Spik K, Rossi C, Bray

M. Evaluation of tick-borne encephalitis DNA vaccines in monkeys.Virology 1999; 263:166-174.

2. Hooper, JW, Custer DM, Thompson E, Schmaljohn CS. DNAvaccination with the Hantaan virus M gene protects hamsters againstthree of four HFRS hantaviruses and elicits a high-titer neutralizingantibody response in Rhesus monkeys. J Virol 2001; 75:8469-8477.

Genetic Influence of HLA Haplotypes on Immune ResponsesFollowing Measles-Mumps-Rubella (MMR-II) Vaccinationin Children I. G. Ovsyannikova1, S. V. Pankratz2, R. M. Jacobson1, R. A. Vierkant2, G. A. Poland1

1Vaccine Research Group, Mayo Clinic College of Medicine, Rochester, MN, 2Health Science Research, Mayo Clinic College of Medicine, Rochester, MN.

Background: The variability of humoral (antibody, Ab) and cellular(lymphocyte proliferation) immune responses modulated by HLAgenes is a significant factor in the protective effect of the MMRvaccine. Methods: We studied the association between measures ofimmune responses and HLA alleles among 346 children who previouslyreceived two doses of the MMR vaccine. Haplotype effects wereestimated using an approach that accounts for linkage phase ambiguityvia an Expectation Maximization algorithm. Results: Mean values formeasles, mumps and rubella Abs and stimulation indices (SI) were2040, 1034, 50 IU/ml and 5.1, 7.3, 3.3, respectively. Specifically,DPB*04-DQB*03-DRB*07 (p=.001) and DPB*04-DQB*06-DRB*02(p=.02) haplotypes were associated with lower levels of measles-inducedAbs. In contrast, DPB*03-DQB*03-DRB*04 (p=.02) was positivelyassociated with lymphoproliferation to measles antigens, whereasDPB*04-DQB*02-DRB*03 (p=.01) was negatively associated with

measles-specific SI. Of all haplotypes analyzed, A*02-Cw*07-B*07(p=.03), A*01-Cw*07-B*08 (p=.02), DPB*04-DQB*02-DRB*03(p=.006) were associated with lower cellular responses to mumps.Interestingly, A*26-Cw*12-B*38 (p=.01) and DPB*04-DQB*05-DRB*01 (p=.008) were associated with high SI. Specific haplotypesassociated with both rubella high and low SI were DPB*03-DQB*06-DRB*06 (p<.001) and DPB*11-DQB*02-DRB*07 (p=.003),respectively. Conclusion: These data further confirm that humoral andcellular responses to MMR vaccine are genetically restricted by HLAgenes.

Reference: 1. Ovsyannikova IG, Jacobson RM, Vierkant RA, Jacobsen SJ, Pankratz

VS, Poland GA. Human leukocyte antigen class II alleles and rubella-specific humoral and cell-mediated immunity following measles-mumps-rubella-II vaccination. J Infect Dis 2005; 191:515-519.

Correlations among Measles Virus-Specific Antibody,Lymphoproliferation and Th1/Th2 Cytokine Responses FollowingMMR-II VaccinationN. Dhiman, I. G. Ovsyannikova, J. E. Ryan, R. M. Jacobson, R. A. Vierkant, S. V. Pankratz, S. J. Jacobsen, G. A. PolandMayo Vaccine Research Group, Mayo Clinic College of Medicine, Rochester, MN.

Introduction: Measles immunity is conferred by humoral andcellular immune responses, later being critical in long-term protectionagainst measles infection. Methods: We examined measles-specificantibodies, lymphoproliferation and Th1/Th2 signature cytokines, IFN-_ and IL-4, in population-based cohort of healthy children fromOlmsted County, Minnesota after measles-mumps-rubella-II (MMR-II)two-dose vaccination. Results: We detected positive measles-specificcellular and humoral immunity in majority of our study population.However, a small proportion of subjects demonstrated an immuneresponse skewed towards the Th2 type, characterized by the presence ofeither IL-4 and/or measles-specific antibodies and a lack of positivelymphoproliferation and IFN-_ production. Further, we observedsignificant positive correlation between lymphoproliferation andsecretion of IFN-_ (r= 0.20, p=0.0002) and IL-4 (r=0.15, p=0.005).Measles antibody levels were associated with lymphoproliferation(r=0.12, p=0.03), but lacked correlation to either cytokine type.Conclusions: We demonstrated both cellular and humoral responsesafter the recommended MMR-II vaccination in a significant proportionof study subjects. Further, a positive correlation betweenlymphoproliferation and IL-4 and IFN-_ suggests that immunity tomeasles may be maintained by both the Th1 and Th2 cells. On the otherhand, biased humoral response observed in a subset of subjects may beinsufficient to provide long-term immunity. Lack of correlation betweenantibody levels and Th1/Th2 cytokines may explain skewed immuneresponses in these individuals. Further work on the determinants ofTh1/Th2 skewing of the immune response is needed.

References:1. Dhiman N, Ovsyannikova IG, Jacobson RM, et al. Correlates of

lymphoproliferative responses to measles, mumps, and rubella(MMR) virus vaccines following MMR-II vaccination in healthychildren. Clin Immunol 2005; in press.

2. Ovsyannikova IG, Jacobson RM, Ryan JE, et al. HLA class II allelesand measles virus-specific cytokine immune response following twodoses of measles vaccine. Immunogenetics 2005; 56:798-807.

S21

S22

S23

Page 66: On-Site Progr

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

66

ABSTRACTS OF ORAL SUBMITTED PRESENTATIONS

Mumps Virus Vaccine Strain Urabe AM9: Identification ofNucleotide Changes Asociated with Variability in theNeurovirulent PhenotypeC. J. Sauder, S. A. Rubin, K. M. Vandenburgh, K. M. CarboneFDA/CBER, Bethesda, MD.

Mumps virus is a highly neurotropic virus. We previously describeda rat model for prediction of mumps virus neurovirulence in humans.The degree of hydrocephalus in brains of rats following neonatalinfection with different mumps virus strains correlated with the strain’sapparent neurovirulent potential in humans. The Urabe AM9 vaccinestrain, which causes meningitis in about 1 in 10,000 vaccinees,exhibited considerable neurovirulence in this rat model. In order toidentify the genetic basis of Urabe AM9 neurovirulence, we sought toalter the virus phenotype by repeated passaging in various cell lines.Employing our neonatal rat model, we found that the in vitro passagedvirus exhibited various degrees of attenuation. Attenuation was foundto be most pronounced in Urabe AM9 passaged six times in chickenembryo fibroblasts (CEF) and human 293 cells. The nucleotidesequence of parental Urabe AM9 as well as of the virus passaged in CEFcells was determined. Attenuation was associated both with adisappearance and evolution of nucleotide heterogeneity on 20positions, 13 of which involving amino acid changes. Using thisknowledge, the relevance for neurovirulence of individual mutationscan be analysed in future studies employing mumps virus reversegenetics technology.

References:1. Rubin SA, Pletnikov M, Taffs R, et al. Evaluation of a neonatal rat

model for prediction of mumps virus neurovirulence in humans. J.Virol. 2000; 74:5382-5384.

2. Amexis G, Fineschi N, Chumakov K. Correlation of geneticvariability with safety of mumps vaccine Urabe AM9 strain. Virology2001; 287:234-241.

S24

Eighth Annual Conference

Page 67: On-Site Progr

on Vaccine Research

67

ABSTRACTS OFSUBMITTED POSTER

PRESENTATIONS

Page 68: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

68

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

Immune Responses Induced by Intranasal Immunization withInfluenza H3N2-anti-H3N2 Complex in MiceX. Yao, Y. WenFudan University, Shanghai, CHINA.

Intranasal delivery of inactivated influenza virus in mice can provideprotection against the homologous virus as well as heterologous strainsdue to the high cross-reactivity of IgA antibody in mucosal secretions.Our previous study showed that Hepatitis B Surface Antigen/Antibodiescomplexes can induce strong humoral immune responses when deliveredby a mucosal route. In this study, 15_g H3N2 antigen complexed withmouse antibodies against H3N2 (anti-H3N2) was administered toBALB/c mice by intranasal inhalation. Mice were divided into 5 groups(8 mice per group), namely, intranasal immunization with only H3N2,H3N2+CpG, H3N2+Polyethylene glycol, H3N2+anti-H3N2 and thecontrols. Each group of mice was immunized with H3N2 15_g withdifferent adjuvants for three times of immunization at three-weekintervals. Compared to mice immunized with H3N2 alone, miceimmunized with antigen-antibody complex showed significant increasein mucosal IgA (Geometric mean: H3N2 16.82, H3N2+anti-H3N2113.14, H3N2+CpG 226.27, H3N2 Vs H3N2+anti-H3N2) (P< 0.01)H3N2+anti-H3N2 Vs H3N2+CpG (P>0.05). The statisticalsignificance of the difference between groups was calculated by Student’stwo-tailed t test using the Excel program(Microsoft). Serumimmunoglobulin G (IgG) antibodies specific to Influenza virus was alsodetected in all study groups. By ELISA, anti-H3N2 titer ranged from51,200 to 819,200; the highest being H3N2+CPG group, and thesecond high was the H3N2 antigen-antibody group, the lowest wasH3N2 group. These results indicate that intranasal immunization usinghemagglutinin antigen- antibody complex, or antigen with CpG caninduce antibodies in serum and in tracheal lung wash. Currently theapproach of intranasal immunization using avian influenza virusantigen-antibody complex is studied in chickens to evaluate whethersimilar immune response can be induced in birds.

References:1. Tumpey T.M., M.Renshaw, J.D. Clements,and J.M.Katz. Mucosal

delivery of inactivated influenza vaccine induces B-cell-dependentheterosubtypic cross-protection against lethal influenza A H5N1virus infection. J.Virol 2001;75:5141-5150.

2. Qu Di, BJ Zheng, Xin Yao, et al. Intranasal immunization withinactivated SARS-CoV(SARS-associated coronavirus) induced localand serum antibodies in mice. Vaccine 23(2005):924-31.

Flagellin is an Effective Mucosal Adjuvant in the Development of a Protective Immune Response Against Yersinia pestisA. N. Honko, S. B. Mizel Microbiology and Immunology, Wake Forest University School of Medicine,Winston-Salem, NC.

Background: Pneumonic plague is a highly transmissible form ofYersinia pestis infection that is fatal without immediate medicaltreatment. Although the shift in vaccine development from completepathogens to individual antigens has led to safer vaccines, efficacy has,in many cases, been markedly reduced. Vaccine adjuvants are necessaryto promote strong adaptive responses to recombinant protein antigens.We evaluated the efficacy of a recombinant mucosal vaccine consistingof the F1 antigen of Y. pestis and Salmonella flagellin as an adjuvant.

Methods: Female mice were immunized intratracheally (i.t.) orintranasally (i.n.) with recombinant, endotoxin-depleted F1 antigen andflagellin or a mutant flagellin and boosted at 4 weeks. Plasma antibodytiters to F1 antigen were determined by ELISA. Immunized and controlmice were challenged i.n. with 1.2x105 cfu Y. pestis CO92, a doseequivalent to 100 times the LD50. Statistical significance and LD50values were determined using SigmaStat3.1. Results: Flagellin-treatedmice had a dramatic increase in anti-F1 plasma IgG titers that remainedstable over time. Using C3H/HeN and C3H/HeJ mice, we found thatsignaling via TLR5/4 heteromeric complexes was not required for theadjuvant effect of flagellin. Type I and II interferons, TNF-_, and IL-6are also not essential for adjuvant activity. Finally, immunization withflagellin and the F1 antigen was protective for i.n. challenge withvirulent Y. pestis CO92. Conclusion: Mucosal immunization withrecombinant F1 antigen and flagellin promotes a strong humoralresponse that is protective against intranasal challenge with Y. pestis.

Reference:1. Honko AN, Mizel SB. Mucosal administration of flagellin induces

innate immunity in the mouse lung. Infect Immun 2004; 72:6676-6679.

Induction of Active Immune Suppression by Co-immunization of DNA-protein VvaccinesB. Wang, H. Jin, Y. Kang State Key Lab for Agro-Biotechnology, China Agricultural University, Beijing, CHINA.

Background: Induction of immune tolerance via protein-basedvaccines has been exploited for treating autoimmune disease such asmultiple sclerosis; whereas the more recently introduced DNA-basedvaccines have been developed for alleviating allergy such as asthma.However, in general, such vaccines work mainly by redirecting immunepolarization (Th1 vs. Th2) or inducing peripheral deletion and arelimited to passive immune regulatory mechanisms. Methods: Co-administration of the DNA-protein regimens into animal has beenemployed to induce an antigen specific T cell suppression has beenevaluated by MLR, DTH, cytokine profiles, adoptive transfer and FACSassays. Results: The co-administration did not produce enhancedimmunity, but rather resulted in impaired delayed type hypersensitivityand antigen-specific T cell proliferation, while the levels of total IgGremain the same. Adoptive transfer of T cells from those co-immunizedanimals conferred the suppressive activity to naïve and active immunizedrecipient animals, suggesting the involvement of suppressor T cells. Theelicited suppressor T cells are antigen non-specific and able to inhibitthe activation of allogenic T cells and are also correlated with a shift ofcytokine balance, as reflected by an elevated level of IL-10 and reducedlevel of IFN-_ or IL-2. Conclusion: Our results illustrate thecomplexity of the interplay between DNA and protein vaccines duringimmunization and are the first study to demonstrate that co-immunization with DNA- and protein-based vaccines may represent anovel means for inducing active suppression against untowardimmunity.

References:1. Amara, R. R., F. Villinger, et al., Control of a Mucosal Challenge and

Prevention of AIDS by a Multiprotein DNA/MVA Vaccine.SCIENCE 2001, 292:69

2. Horner, A., J. Van Uden, J. Zubeldia, D. Broide, and E. Raz. DNA-based immunotheraputics for the treatment of allergic disease.Immunol Reviews 2001, 179:102.

P1

P2

P3

Eighth Annual Conference

Page 69: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

69

(continued)

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

Aluminum Phosphate is an Active Adjuvant for CRM197Pneumococcal Conjugate Vaccine (PnC) in InfantsS. Lockhart1, W. Watson1, P. Fletcher2, A. Leeper3, S. Edwards4, M. McCaughey5, A. Dunning1, D. Sikkema1, G. Siber1

1Wyeth Research, Pearl River, NY, 2Woolwell Surgery, Plymouth, UNITEDKINGDOM, 3Grove Surgery, Thetford, UNITED KINGDOM, 4North CardiffMedical Centre, Cardiff, UNITED KINGDOM, 5Health Centre, Randalstown,UNITED KINGDOM.

Background. This study was designed to compare theimmunogenicity and reactogenicity of combined 9-valentpneumococcal (9vPnC) and meningococcal C (MnCC) CRM197conjugates with (AlPO4 [+]) or without AlPO4 (AlPO4 [-]) in humaninfants. Methods. 224 UK infants were randomized to 9vPnC/MnCCat 2, 3, 4 and 12 months formulated as AlPO4 [+] or AlPO4 [-].Subjects also received Infanrix-Hib and oral polio vaccine. Ab wasassayed by ELISA. Results. IgG Ab responses were higher with AlPO4group for MnC and most PnC serotypes after two or more doses. GM[Ab] after 3 doses in 94 evaluable AlPO4 (+) and 91AlPO4 (-)recipients were:

PnPs9 4 5 6B 9V 14 18C 19F 23F MnCPs

AlOP4 + 2.46* 2.08* 1.15* 1.05 1.43* 4.69 1.40 2.94 1.19 9.63*

AlPO4 - 1.79 1.41 0.84 0.88 1.06 3.87 1.17 2.99 0.95 7.13

*significant at p <.05Reactions: AlPO4 (+) 9vPnC/MnCC induced significantly moreerythema, swelling and tenderness than AlPO4 (-) but similar rates toInfanrix-Hib. No significant differences were seen in fever, and othersystemic reactions after any dose. Conclusions. AlPO4 increased Abresponses to most PnC serotypes and MnCC in human infants. Localreactions were higher with AlPO4 but similar to those with licensedvaccines.

References:1. Wernette CM, Frasch CE, Madore D, et al. Enzyme-linked

immunosorbent assay for quantitation f human antibodies topneumococcal polysaccharides. Clin Diagn Lab Immunol2003;10(4):514-519

2. Sikkema DJ, Friedman KE, Corsaro B, et al. Relationship betweenserum bactericidal activity and serogroup-specific immunoglobulinG concentration for adults, toddlers, and infants immunized withNeisseria meningitidis serogroup C vaccines. Clin D

PyNTTTTGT Oligonucleotide IMT504 is a Potent Vaccine AdjuvantA. D. Montaner1, F. Elias2, J. M. Rodriguez2, J. Flo2, R. Lopez2, J.Zorzopulos1

1Instituto de Investigaciones Biomédicas, Fundación Pablo Cassará., BuenosAires, ARGENTINA, 2Immunotech S.A., Buenos Aires, ARGENTINA.

PyNTTTTGT oligodeoxinucleotides (ODNs) cause activation,proliferation and immunoglobulin secretion on B cells, and theexpression of co-stimulatory molecules on plasmacytoid dendritic cellsof primates and rats (1)

The goal of this study was to investigate the adjuvant properties ofPyNTTTTGT prototype IMT504 ODN on different human vaccinessuch as Flu, Hepatitis B, Hepatitis A and Rabies.Addition of IMT504 to commercial vaccines, compared to the antigen

alone, resulted in:a) higher antibody responses (Flu, Hepatitis B and Hepatitis A)

b) earlier antibody responses (Flu and Hepatitis A) andc) the need of much less antigen to obtain a good antibody response

(Flu and Rabies)

Since rat and monkey cells are significantly less immunostimulatedin vitro by PyNTTTTGT ODNs than human cells, it can be predictedan excellent performance for human vaccination. Moreover, preclinicaltrials have demonstrated that IMT504 is an extremely safe drug.

Novel Peptide Nanoparticles: A Platform for Vaccine DesignD. Tropel1, A. C. Tissot2, C. Schellekens2, S. K. Raman1, A. Graff1, G.Machaidze1, M. F. Bachmann2, P. Burkhard3

1M.E Mueller Institute, Biozentrum, Basel, SWITZERLAND, 2Cytos Biotechnology,Schlieren, SWITZERLAND, 3The Institute of Materials Science, University ofConnecticut, Storrs, CT.

Background: We have recently described structure-based design of anovel type of nanoparticles, which self-assemble from single polypeptidechains. Such nanoparticles have a regular polyhedral symmetry and adiameter similar to small viruses, so we have investigated the possibilityto use them as a repetitive antigen display system. Methods: We haveestablished a recombinant Escherichia coli protein expression system toeasily produce nanoparticles displaying an antigenic Salmonella epitope.These nanoparticles were injected to four mice at day 0 and 14 andantibody production was studied by Elisa assay. Results: We haveconstructed an expression vector for Escherichia coli, which allowed thefusion between the DNA sequences coding for an antigenic Salmonellaepitope and the peptide forming the nanoparticle. The resulting His-tagged polypeptide was purified via metal affinity chromatographyunder denaturing conditions and correct self assembling intonanoparticles was obtained by successive dialyses. The serum of miceimmunized with nanoparticles displaying Salmonella epitope wasanalyzed at day 21. High titers of epitope-specific IgG were detected,suggesting a strong humoral immune response. Conclusions: Sinceother peptide epitopes can easily be displayed on the surface of thesenanoparticles this novel technology represents a versatile platform forefficient vaccine design. Furthermore, since the nanoparticles are builtfrom protein domains this allows for a highly specific design in terms ofshape, size and stability.

Reference:1. Lechner F, Jegerlehner A, Tissot AC, et al. Virus-like particles as a

modular system for novel vaccines. Intervirology 2002; 45:212-217

Native Display of An HIV Tat Peptide on the Surface of HumanFerritinC. Li, E. Soistman, D. C. CarterNew Century Pharmaceuticals, Inc, Huntsville, AL.

In order to improve immunogenicity and efficacy, vaccinationstrategies are becoming increasingly sophisticated. Recently, we havedevelopped a protein nanoparticle based technology platform for thepresentation of protein and peptide immunogens. As a demonstration,an HIV-1 Tat peptide has been engineered and expressed in E.coli. as aferritin fusion protein.SDS-PAGE and Transmission ElectronMicroscopy studies confirmed proper capsid assembly and orderly

P4

P6

P5P7

Page 70: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

70

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

display of the HIV Tat prptide on the surface of this proteinnanoparticle. Animal studies demonstrated that this chimeric particleelicits a potent humoral response orders of magnitude greater thanconjugation of the peptide with KLH. After final immunization, theserum immune response lasts more than three months without notebleloss of the activity. Concentrations of fusion protein which are 100times (Mole ratio) less than that utilized to immunize with soluble Tatpeptide alone, are capable to induce considerable immune response.Preliminary studies using an HIV virus Tat-based neutralization assayand the antibody generated with this chimeric particle indicatedneutralization of the virus. Orally immunized animals with this fusionprotein also produce high titer anti-Tat antibodies. These resultsindicated that the protein nanoparticle (ferritin)-mediated delivery ofHIV Tat peptide can dramatically increase the antigen accessibility tohost immune system and induce specific neutralizing antibodies. Oralactivity suggested that peptide expressed on this platform can resistenzymatic degradation and be absorbed into the bloodstreamefficiently. Further examples which include applications for influenza,malaria and foot and mouth disease are in development.

References:1. Kramer RM, Li C, Carter DC, Stone MO and Naik RR. Engineered

protein cages for nanomaterial synthesis. Journal of AmericanChemistry Society. 2004; 126(41): 13282-13286.

2. Morris CB, Thanawastien A, Sullivan DE and ClementsJD.Identification of a peptide capable of inducing an HIV-1Tat-specificCTL response. Vaccine.2002; 20: 12-15

Protection Against Hepatitis B Carriage Following InfantVaccination May Fall With AgeM. E. Mendy, M. A. B. van der Sande, P. Waight, P. Rayco-Solon, P. Hutt, T. Fulford, C. Doherty, S. McConkey, D. Jefferies, A. Hall, H. WhittleViral Disease Programme, Medical Research Council, Banjul, GAMBIA.

Background. Chronic carriage of HBV is a major risk factor forliver cirrhosis and hepatocellular carcinoma. Infant vaccination hasbeen effective in preventing horizontal transmission in earlychildhood. It is not known whether this protection will be maintainedagainst sexual transmission in early adulthood. Methods. Infantvaccination was introduced in The Gambia in 1984. Serologicalassessment of 1099 fully vaccinated participants between the ages of 1and 24 was conducted in 2003, in order to determine vaccine efficacy(VE) against infection and chronic carriage two decades postintrocudction of vaccination. The core and surface antibody tests wereperformed by radio immunoassay or ELISA and HBsAg was detectedby RPHA or immunochromatography. Data were entered andvalidated in access 2000 and data analysis was done using Stata 8.Results. Overall VE against infection and chronic carriage was 83.4%(95%CI 79.8-86.6) and 96.5% (93.9-98.9) respectively; againstcumulative infection and carriage it was 78.0% (74.1-81.7) and 95.0%(92.0-97.7). However, both VE and surface antibody levels declinedwith age. Less than half of the vaccinees still had detectable antibody15 years after vaccination. Conclusions. HBV vaccination in early lifecan provide long-lasting protection into early adulthood, in spite ofloss of antibody levels and an increasing rate of core antibodyconversion, but decreases progressively with age. The role of sub-clinical boosting, and the necessity of a booster vaccine, need to beevaluated.

References:1. Bah E, Parkin DM, Hall AJ, et al. Cancer in the Gambia: 1988-97.

Br J Cancer 2001;84:1207-142. Whittle HC, Jaffar S, Wansborough M, et al Observational study of

vaccine efficacy 14 years after trial of hepatitis B vaccination inGambian children. BMJ 2002;325:569-72

T Cell Responses to Hepatitis B VaccineM. S. Hayney, N. A. Wiegert University of Wisconsin School of Pharmacy, Madison, WI.

Background: T cell function is critical to the biology ofimmunization. The hypotheses are that the T cell responses to hepatitisB vaccine will be CD4+ cell dominant and that the vigor of theresponses will correlate with cytokine genotype. Methods: Ten healthyhepatitis B vaccine responders were enrolled. Peripheral bloodmononuclear cells (PBMC) and CD4+ and CD8+ populations wereisolated for the trans-vivo delayed-type hypersensitivity (DTH) assayfrom blood samples obtained. PBMC alone, CD4+ or CD8+

lymphocytes and antigen presenting cells alone, with hepatitis B surfaceantigen (HBsAg) and with HBsAg and antibodies to interferon _(IFN_), interleukin-4 (IL), or IL-10 were injected into the footpad ofimmunodeficient mice. The resulting swelling is an index of human Tcell sensitization. Cytokine genotyping of IFN_, IL-4 and IL-10 wasdone using the polymerase chain reaction. Results: The mean responseby CD4+ cells (35.0+6.6 x10-4 inches) and CD8+ cells (23.5+6.4 x10-4

inches) to HBsAg were similar (p>0.2; paired t test). The CD4+ cellresponses depended on IL-4 and IL-10 such that the vigor of theresponses decreased when these cytokines were inhibited (CD4+

35.0+6.6 x10-4 inches vs. CD4+ antiIL-4/10 18.0+3.3x10-4 inches;p<0.03) The CD8+ cell response was not consistently regulated bycytokines. Those with high IL-10 producing genotypes were particularlydependent on Th2 cytokines for DTH response (low producers32.5+7.5x10-4 inches vs. high producers 14.4+2.7x10-4 inches p<0.03).Conclusion: Hepatitis B immunization elicits both CD4+ and CD8+

immune responses. The vigor of the CD4+ response is regulated by Th2cytokines—IL-4 and IL-10.

References:1. VanBuskirk AM, Burlingham WJ, Jankowska-Gan E, et al. Human

allograft acceptance is associated with immune regulation. J. Clin.Invest. 2000;106:145-55

2. Hahn AB, Kasten-Jolly JC, Constantino DM, et al. TNFa, IL-6,IFNg, and IL-10 gene expression polymorphisms and the IL-4receptor a-chain variant Q576R: Effects on renal allograft outcome.Transplantation 2001;72:660-5

T Cell Responses Following Hepatitis A ImmunizatonM. S. Hayney, N. A. WiegertUniversity of Wisconsin School of Pharmacy, Madison, WI.

Background: Interferon _ (IFN_) and interleukin (IL)-10 areproduced with hepatitis A infection and following immunization. Thehypotheses are that the T cell responses to hepatitis A vaccine will beCD4+ cell dominant and that the vigor of the responses will correlatewith cytokine genotype. Methods: Eight healthy hepatitis Aseronegative individuals were immunized with hepatitis A vaccine.

P10

P8

P9

Eighth Annual Conference

Page 71: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

71

(continued)

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

Peripheral blood mononuclear cells (PBMC) and CD4+ and CD8+populations were isolated for the trans-vivo delayed-typehypersensitivity (DTH) assay from blood samples obtained 10-12 daysafter immunization. PBMC alone, CD4+ or CD8+ lymphocytes andantigen presenting cells alone, with hepatitis A antigen (hepA), andwith hepA and antibodies IFN_, IL-4, or IL-10 were injected into thefootpad of immunodeficient mice. The resulting swelling is an indexof T cell sensitization. Cytokine genotyping of IFN_, IL-4 and IL-10was done using the polymerase chain reaction. Results: The meanresponse by CD4+ cells (26.1+12.4 x10-4 inches) and CD8+ cells(16.7+9.6 x10-4 inches) to hepA vaccine virus were similar (p=0.4;paired t test). The CD8+ cell response was regulated by IL-4 and IL-10 (CD8+ 8+14 vs. CD8+ anti-IL-4/10 24+15 x10-4 inches; p<0.05;paired t test). Additionally, all four individuals with intermediate tohigh IFN_, IL-4 and IL-10-producing genotypes maintained orincreased CD8+ cell responses to hepA with anti-IL-10 indicating therole of IL-10 in regulating the immune response (p<0.05; Fisher’sexact). Conclusion: Hepatitis A immunization elicits CD4+ andCD8+ immune responses. The vigor of the CD8+ response isregulated by IFN_, IL-4 and IL-10.

References:1. VanBuskirk AM, Burlingham WJ, Jankowska-Gan E, et al. Human

allograft acceptance is associated with immune regulation. J. Clin.Invest. 2000;106:145-55

2. Hayney MS, Buck JM and Muller D. Production of interferon gand interleukin-10 after inactivated hepatitis A immunization.Pharmacotherapy 2003;23:431-5

Impact of a School Based Hepatitis B Immunization Program in aLow Endemic AreaV. Gilca, PhD1, B. Duval, MD2, N. Boulianne, MSc2, R. Dion, MD2, G. D. Serres, PhD3

1Centre de recherche du Centre Hospitalier Universitaire de Québec, Quebec,PQ, CANADA, 2Institut National de Santé Publique du Québec, Quebec, PQ,CANADA, 3Laval University, Quebec, PQ, CANADA.

Background: In 1994, a school-based routine HB immunizationprogram was implemented in Quebec in grade 4 children in parallelwith increased vaccination of high risk groups. Ten years later weconducted a study to assess the impact of the program, vaccinefailures, and number of cases that would have been prevented by aninfant program. Methods: Acute HB cases reported for 1994-2003were retrieved from the provincial Register of Notifiable Diseases.Detailed information on risk factors and vaccination status wereextracted from regional Public Health Department files for all 0-20year-old cases. Results: Between 1994 and 2003, a 4-fold HBincidence decline was observed in the general population, 10-fold inthe 10-20 year-olds. In the same period, from 37 cases observed in 10-20 year-olds, 30 (81%) cases were in older cohorts not covered bythe school immunization program. In this age group, 32 cases werenon vaccinated, one case fully vaccinated, and 4 others incompletelyvaccinated. From 45 cases notified in under 10 year-old children, 24 (53%) were born outside Canada and 14 (31%) were born fromHBsAg positive mothers. Up to 26 (58%) cases in children under 10 years old could have been prevented by an infant immunizationprogram in Quebec. Conclusions: The preteen vaccination program ishighly effective. An infant program would bring some additionalbenefits.

References:1..Patrick DM, Bigham M, Ng H, White R,et all. Elimination of acute

hepatitis B among adolescents after one decade of an immunizationprogram targeting Grade 6 students. Pediatr Infect Dis J.2003;22(10):874-877

2. Mele A, Stroffolini T, Zanetti A, et al. Hepatitis B in Italy: where arewe ten years after the introduction of mass vaccination? J MedVirology 2002;67:440-443

Antibody Responses to Vaccinia Membrane Proteins FollowingSmallpox VaccinationS. Lawrence1, K. Lottenbach2, F. Newman2, M. Buller2, C. Bellone2, S. Koehm2, S. Stanley, Jr.1, R. Belshe 2, S. Frey 2

1Washington University, St. Louis, MO, 2Saint Louis University, St. Louis, MO.

Background: Vaccinia virus extracellular enveloped virion (EEV)and intracellular mature virion (IMV) membrane proteins are potentialcandidates for use in smallpox subunit vaccines. Human antibodyresponses to these proteins after vaccinia vaccination are not known.Methods: Paired (pre- and 28-days post-vaccination) sera from 29vaccinia-naïve and 29 non-naïve adults vaccinated with live vacciniavirus (Dryvax®) were examined for presence and titers of anti-EEVprotein (B5R, A33R) and anti-IMV protein (L1R, A27L) antibodies byELISA. Chi-square, non-parametric, and t-tests were used to compareantibody responses. Results: In naïve vaccinees, post-vaccinationantibodies were detected in 69.0% [B5R], 100% [A33R], 93.1%[A27L], and 62.1% [L1R] of naïve vaccinees. In non-naïve vaccineespre-existing antibodies were present in 72.4% [B5R], 58.6% [A33R],40.7% [A27L], and 6.9% [L1R], and were boosted ≥4-fold in 62.1%[B5R], 34.5% [A33R], 59.3% [A27L], and 3.5% [L1R]. Interestingly,anti-A33R responses were more vigorous in naïve vaccinees (10.0- vs2.8-fold geometric mean titer boost for naïve vs. non-naïve, p<0.001),resembling neutralizing antibody responses (88.2- vs. 14.3-fold boost,p<0.001), while anti-B5R responses were more robust in non-naïvevaccinees (3.1- vs. 6.5-fold boost, p=0.010), similar to whole virusbinding (ELISA) antibodies (6.3- vs. 15.6-fold boost, p<0.001).Conclusions: Our findings suggest that Dryvax® vaccination elicitsanti-EEV and anti-IMV membrane protein antibody responses in mostnaïve vaccinees. Except for anti-L1R, these specific antibody responsesappear durable and are substantially boosted by re-vaccination. Furthercorrelation with other immunologic and clinical parameters is needed.

References:1. Frey SE, Newman FK, Yan L, Belshe RB. Response to smallpox

vaccine in persons immunized in the distant past. JAMA2003;289:3295-9.

2. Belshe RB, Newman FK, Frey SE, et al. Dose-dependentneutralizing-antibody responses to vaccinia. J Infect Dis2004;189:493-497.

P12

P11

Page 72: On-Site Progr

ABSTRACTS OF INVITED PRESENTATIONS

72

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

inhibition ELISA protocol are linear, reproducible and cover a largedynamic range.

References:1. Harris, SL, King, WJ, Ferris, W, Granoff, DM. Age-related disparity

in functional activities of human group C serum anticapsularantibodies elicited by meningococcal polysaccharide vaccine Infect.Immun. 2003;71(1):275-286.

2. Goldblatt, D, Borrow, R, Miller, D. Natural and vaccine-inducedimmunity and immunologic memory to Neisseria meningitidisserogroup C in young adults. J. Infect. Dis., 2002; 185(3):397-400.

Quantitation of Human Serum Immunoglobulin G Against O-AcetylPositive and O-Acetyl Negative Serogroup W135 MeningococcalCapsular PolysaccharideP. C. Giardina1, E. Longworth2, R. Borrow2, P. Fernsten1

1Applied Immunology and Microbiology, Wyeth, Pearl River, NY,2Meningococcal Reference Unit, Manchester Medical Microbiology Partnership,Health Protection Agency, Manchester, UNITED KINGDOM.

Background: The capsule of Neisseria meningitidis serogroup W135is expressed as both O-acetyl positive (OA+) and O-acetyl negative (OA-) forms. The purpose of this study is to understand the impact of OAstatus on serological measurements of anti-W135 IgG antibodies.Methods: Sera from 28 adults immunized with a licensedmeningococcal polysaccharide vaccine (Menomune; A, C, Y and W135vaccine, Aventis) were analyzed for W135-specific serum antibodyconcentrations against the standard reference serum, CDC1992, byenzyme-linked immunosorbent assay (ELISA) and for functional killingactivity by serum bactericidal assay (SBA). Results: A concentration of10.13_g/ml IgG against OA- antigen was established for CDC1992 bycross-standardization against OA+ antigen (16.2_g/ml). Overall, serumIgG against OA+ antigen (geometric mean concentration (GMC) =7.16_g/ml) was higher compared to OA- antigen (GMC = 2.82_g/ml).However, seven specimens contributed disproportionately to thisdifference. These sera were also distinguished by the inability of fluidphase OA- antigen to compete for antibody binding to OA+ solid phaseantigen. Although there were no overall differences in SBA titers againstOA+ and OA- target bacteria (GMT = 9642 and 9045, respectively),three specimens showed relatively large differences, which may reflectdifferent epitope specificities. Conclusion: The relationship betweencapsule-specific IgG concentrations measured by ELISA against OA+and OA- W135 antigens is serum specific and does not reflect thefunctional (killing) activity for some specimens, in vitro.

References:1. Elie, C. M., P. K. Holder, S. Romero-Steiner, and G. M. Carlone.

2002. Assignment of Additional Anticapsular AntibodyConcentrations to the Neisseria meningitidis Group A, C, Y, and W-135 Meningococcal Standard Reference Serum CDC1992. ClinDiagn Lab

2. Giardina, P. C., R. E. Evans, D. J. Sikkema, D. Madore, and S. W.Hildreth. 2003. Effect of antigen coating conditions on enzyme-linked immunosorbent assay for detection of immunoglobulin Gantibody to Neisseria meningitidis serogroup Y and W135 caps

Serum Neopterin for Early Assessment of Severity of SevereAcute Respiratory SyndromeJ. W. Y. Choi Department of Microbiology, The University of Hong Kong, Pokfulam, HongKong Special Administrative Region of China.

Neopterin concentrations were determined in serum samples from129 severe acute respiratory syndrome (SARS) patients and 156healthy blood donors. In the patients with confirmed SARS, an earlyneopterin elevation was detected already at the day of onset ofsymptoms and rose to a maximum level of 122.5 nmol/L 3 days afterthe onset. The mean neopterin concentrations were 34.2 nmol/L inacute sera of SARS patients, 5.1 nmol/L in convalescent sera and 6.7nmol/L in healthy controls. Serum neopterin level in SARS patientswas associated with fever period and thus the clinical progression ofthe disease. After steroid treatment, decrease of neopterin leveldemonstrates that it is a useful marker of inflammatory activity ofSARS. Together with a 100% of sensitivity within 9 days after theonset of symptoms, serum neopterin may allow early assessment of theseverity of SARS and evaluation of the treatment efficacy.

References:1. Peiris JSM, Lai ST, Poon LLM, et al. Coronavirus as a possible

cause of severe acute respiratory syndrome. Lancet 2003;361:1319-1325

2. Fahey JL, Taylor JM, Detels R, et al. The prognostic value ofcellular and serologic markers in infection with humanimmunodeficiency virus type 1. N Engl J Med 1990;322:166-172

Determining Avidity for Anti-polysaccharide AntibodiesS. Harris, J. Tam, P. Fernsten;Vaccines Research, Wyeth, Pearl River, NY.

Background: Antibody avidity is an important characteristic ofprotective immunity. One advantage of polysaccharide-conjugatevaccines over polysaccharide vaccines is the ability to elicit higheravidity antibodies which may correlate with B cell memory. Achaotropic ELISA is commonly used for assessing avidity indices.However, chaotropes disrupt secondary and tertiary antibody andantigen structure as well as antibody-antigen interactions. As a result,the avidity index may not correlate to antibody avidity alone.Methods: Avidity (KD) can be expressed as the concentration ofantigen needed to decrease antibody binding by 50% (IC50) atequilibrium. Thus, IC50 data from inhibition ELISAs using solubleantigen to inhibit antibody binding to immobilized antigen can beconverted to avidity constants if the inhibitor concentration isexpressed in terms of moles of epitope. Polysaccharide epitopemolarity can be determined from binding stoichiometry meaurementsderived from microcalorimetry titration experiments. Results: We havedeveloped an inhibition ELISA to measure antibody avidity to thecapsular polysaccharide (PS) of Neisseria meningitidis group C (MnC).For two MnC PS monoclonal antibodies, binding occurs every 10.5carbohydrate repeat units. We have determined avidity constants forseveral sera from adults immunized with MnC PS vaccine and from100 children immunized with MnC PS-conjugate vaccine. The avidityconstants for the adult sera tested range from 1.70 nM to 1173 nMand for the pediatric sera from 118 nM to 1220 nM. Comparison withpublished chaotropic ELISA data suggests the inhibition ELISA has alarger dynamic range. Conclusions: Avidity constants from an

P13

P15

P14

Eighth Annual Conference

Page 73: On-Site Progr

on Vaccine ResearchABSTRACTS OF INVITED PRESENTATIONS

73

(continued)

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

Evaluating the Influences of Glycosylation on the Immunogenicityof the Ebola Virus GlycoproteinW. E. Dowling1, R. J. Hogan2, E. Thompson3, J. Paragas1, G. Bush1, J. Smith1, W. Capps1, L. Grey1, C. Badger1, C. S. Schmaljohn1

1Virology, USAMRIID, Fort Detrick, MD, 2Infectous Diseases, University ofGeorgia, Athens, GA, 3Cder, FDA, Rockville, MD.

Background: The requirements for protective immunity againstEbola virus infection are not completely understood, although theimportance of antibody and T-cell responses against the glycoprotein(GP) have been demonstrated (Reviewed in 1). Numerous studies ofother viral systems have shown that glycosylation plays a major role inthe structure, function, and antigenicity of glycoproteins (2). Methods:We generated a series of glycosylation mutants of GP to assess theimportance of glycosylation to its expression and immunogenicity.These mutated genes were cloned into a mammalian expression plasmidand tested for expression in transfected cells. Mice were vaccinated witheach construct three times by gene gun. Sera were collected after eachinoculation and were tested for antibody titers by ELISA. Ten mice pergroup were challenged with mouse-adapted EBOV Zaire. The remainingmice were killed, and then splenocytes were harvested, stimulated withpools of overlapping GP peptides, and assessed for interferon-gammaproduction by ELISPOT. Results: The different glycosylation mutantselicited varying ELISA titers and T-cell responses in vaccinated mice. Inparticular, the absence of one of the glycosylation sites in GP2 had asignificant impact on antibody responses, T-cell responses, andprotection after challenge with EBOV. Conclusions: These resultsindicate that alterations in the glycosylation of GP can affect itsimmunogenicity and should be considered in the design of GP-basedEBOV vaccines and therapeutics.

References:1. Feldmann H, Jones S, Klenk HD and Schnittler HJ. Ebola virus:

from discovery to vaccine. Nat. Rev. Immunol. 2003; 3;677-85.2. Wei X, Decker JM, Wang S, et al. Antibody neutralization and escape

by HIV-1. Nature 2003; 422;307-12.

The Establishment of a Restriction Assorted Fragments Expression(RAFE) System for Vaccine Development of HIV-1B SubtypeR. Shi1, W. Zheng2, J. Liu1, L. Li1, W. Ma1

1Institute of Molecular Biology, NanFang Medical University, Guangzhou,CHINA, 2South China Genomics Research Center, Guangzhou, CHINA.

Background To establish a universal RAFE system which is simplerthan random peptide libraries (RPL) screening for the development ofHIV recombinant vaccines. Method The genome of HIV-1B subtypeU26942 was digested with Sau3A I into 26 restriction fragments.Specially designed universal adapters were ligated with each fragments sothat it could express all of the three possible reading frame peptides bysense and anti-sense orientations. Such fragments were cloned intoVector pFAB5C and verified for surface display endorsed by a CMVpromoter. This surface display library was screened by ELISA and wasfurther used as a pool of HIV phage-display fusion peptides, which wereprinted into a microarray. Such peptide pool was further injected intoexperimental mice, sera of experimental mice as well as negative controlswere collected and applied upon the microarray for detection. ResultsOver 1/2 among the 156 possible peptides could be expressed as fusionpeptides on phage surface. 29 antibodies against the fusion peptidescould be detected by microarray in the experimental mice, among which

P16

P17

6 were HIV-1 envelope proteins. Conclusion: A RAFE system wasestablished which could be valuable in generating vaccines for thecontrol of HIV infection.

Reference:1. Chen X, Scala G, Quinto I, et al. Protection of rhesus macaques

against disease progression from pathogenic SHIV-89.6PD byvaccination with phage-displayed HIV-1 epitopes. Nature Med 2001;7(11):1225-1231

Toll-like Receptor 4 Plays Role in Activating Dendritic Cells byNecrotic CellsC. Kang, J. Choi, S. Lee, H. Moon, S. SeongMicrobiology and Immunoogy, Seoul National University College of Medicine,Seoul, REPUBLIC OF KOREA.

It is currently thought that immune responses are initiated bypathogen-associated molecular patterns (PAMP) or by damage-associatedmolecular patterns (DAMP). However, these two groups of moleculesmight not be mutually exclusive. Many of them might be part of anevolutionarily ancient alert system in which the hydrophobic portions(Hyppos) of biological molecules act, when exposed, as universal DMAPto initiate repair and immunity.

Because life on earth originated in water, Hyppos are normally hiddenfrom the aqueous environment by conformational folding, by help fromregulatory molecules or by virtue of their insertion into a lipid bilayer.Because exposed Hyppos rapidly form non-productive or even toxicaggregates, organisms expend a great deal of metabolic energy to preventsuch exposure and maintain homeostasis. If, however, cell should dienecrotically, it will release several Hyppos into its immediateenvironment, including membrane lipids, integral membrane molecules,immature proteins, heat shock proteins, etc.

In this study, we showed that Toll like receptor 4 recognize DAMP.Lipids activated dendritic cells. The activation of DC by necrotic cells(NC) was inhibited by apolipoproteins. DCs from TLR4 knockout micewere less responsive to NC than wild type DC in terms of costimulatorymolecule expression, IL12p40 secretion and naïve T cell activation. Thechemokine expressions of DC by NC were controlled by TLR4 pathways.For these reasons, we suggest that Hyppos from NC are evolutionarilyancient DAMP and TLR4 contributes in activating DC by Hyppos.

Reference:1. Seong SY, Matzinger P. Hydrophobicity; Damage-associated

molecular pattern that initiates innate immune responses. Nature RevImmunol 2004; 4: 469-478

Construction of Recombinant BCG Based HIV-1 epitope DeliverySystem and Evaluation of its Immunogenicity in a Murine ModelA. Vivekanandhan, N. Sujatha, P. NarayananImmunology, Tuberculosis Research Centre, Chennai, INDIA.

Background: AIDS vaccine is a global emergency. Intense efforts areunder way to identify protective epitopes and novel delivery vehicles.Grafting an epitope from an immune unfriendly environment to animmune friendly environment forms the basic of ‘epitope grafting’.Methods: In this respect the present project aims at constructing an

P18

P19

Page 74: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

74

Eighth Annual Conference‘epitope trap vector’ using Mycobacterium tuberculosis chaperonin-10(Cpn10) antigen as a carrier antigen. The HIV-1 PND epitope wasselected as a test epitope.Two versions of the Cpn10-PND chimeric antigen were constructed andexpressed in M. smegmatis:

1. The replacement chimera where the PND epitope replaces theCpn10 loop and

2. The insertion chimera where the PND epitope is inserted into theCpn10 loop

Based on the expression profile p306CRC (Cpn10-PNDreplacement chimera in an episomal vector with homologous promoter)was electroporated into BCG Pasteur. The immunogenicity of therecombinant BCG was evaluated in a murine model. Results:Vaccination with rBCG expressing the PND epitope induced bothcellular and humoral immune responses as measured by lymphocyteproliferation, delayed-type hypersensitivity (DTH) reaction, cytokinesecretion, generation of memory T cells and antibody production.Conclusion: In summary heat shock protein based epitope deliveryvehicles offer novel avenues in AIDS vaccine research.

Reference:1. Honda, M., K. Matsuo, et al. (1995). “Protective immune responses

induced by secretion of a chimeric soluble protein from arecombinant Mycobacterium bovis bacillus Calmette-Guerin vectorcandidate vaccine for human immunodeficiency virus type 1 in smallanimals.” Proc Natl Acad Sci U S A 92(23): 10693-7.

Extending the Capsid Deletion Approach for Favivirus VaccineDevelopment to the N-terminal Part of the ProteinR. M. Kofler, C. W. Mandl Clinical Institute of Virology, Medical University of Vienna, Vienna, AUSTRIA.

A new genetic vaccine principle for flaviviruses was recentlydeveloped based on a non-infectious, self-replicating RNA of tick-borneencephalitis virus (TBEV). Cells expressing a replicon, which wasderived by engineering a deletion into the TBEV RNA genome codingfor the central and C-terminal parts of capsid protein C, secretedcapsidless subviral particles but not whole viral particles and induced ahighly protective immune response in adult mice. To advance thisgenetic vaccine approach, we investigated the relevance of the regionupstream of the original deletion for TBEV replication. This regioncoding for the N-terminus of protein C comprises a GC-rich predictedstem loop structure of potential functional importance and, in the caseof mosquito-borne flaviviruses, contains a so-called cyclization element(CyE) essential for RNA replication. To analyze the significance of thisregion for TBEV replication, two deletions were engineered, oneextending towards the stem-loop and the other removing approximatelyhalf of this structure and the potential CyE. Surprisingly, both mutantsshowed competence for RNA replication and protein translation anddemonstrated viability and genetic stability in cell culture and sucklingmice. Both mutants were neurovirulent in suckling mice, but the onsetof disease symptoms was delayed. In adult mice, the mutants exhibitedan attenuated phenotype and induced a protective immune response.Our results demonstrate that the N-terminal protein C coding region isnot essential for TBEV replication, but displays a further target for thegeneration of attenuated and immunogenic vaccine candidates forflaviviruses.

References:1. Kofler RM, Aberle JH, Aberle SW, Allison SL, Heinz FX, Mandl CW.

Mimicking Live Flavivirus Immunization with a Noninfectious RNAVaccine. PNAS 2004; 101: 1951-1956;

2. Kofler RM, Heinz FX, Mandl CW. Capsid Protein C of Tick-BorneEncephalitis Virus Tolerates Large Internal Deletions and is aFavorable Target for Attenuation of Virulence. J. Virol. 2002; 76:3534-3543;

Serotypes, Virulence Genes, and PFGE Patterns of Escherichia coli Strains Isolated from Piglets with Diarrhoea in SlovakiaH. Vu Khac Department of Bacteriology, Central Viet Nam Veterinary Institute, Nha trangCity, VIET NAM.

250 E.coli strains isolated form piglets were serotyped and tested forthe presence of the genes for fimbriae, intimin, enterotoxin, verotoxin,and EAST1 toxin by PCR. These 250 strains belonged to 90 differentO:H serotypes. Although 220 isolates from diarrheic piglets belonged to43 different O serogroups and 77 O:H serotypes, 60.5% of isolatesbelonged to only 10 serogroups (O2, O8, O15, O54, O84, O101,O141, O147, O149, and O157) and 57.7% belonged to only nineserotypes (O8:H-, O54:H-, O84:H7, O141:H-, O141:H4, O147:H-,O149:H10, O163:H-, and ONT:H-). PCR show that 78.6% of 220strains from diarrheic piglets carried genes for at least one of virulencefactors tested. Eighty-three (37.7%) of the 220 E. coli isolates carriedgene for F4, whereas genes for F18, F5/F41, F6, F17, and intimin weredetected in 9.1%, 3.2%, 2.7%, 1.4% and 3.2%, respectively. The geneencoding for EAST1 was the most prevalent (65.4%) followed by thoseencoding for STb (49.5%), LT (41.8%), STa (12.7%), and VT2 (4.1%).Among 30 strains from healthy piglets only two pathotypes, EAST1(26.4%) and eae (16.6%), were recovered. The results showepidemiological information about the prevalence of serogoups,serotypes, and virulence factors in porcine E. coli in Slovakia.

References:1. Blanco M, Blanco JE, Gonzalez EA, et al. Genes coding for

enterotoxins and verotoxins in porcine Escherichia coli strainsbelonging to different O:K:H serotyps: relationship with toxinphenotyes.Journal of Clinical Microbiology. 1997; 35:2958-2963.

2. Harel J, Lapointe H, Fallara A, et al. Detection of genes for fimbrialantigens and enterotoxins associated with Escherichia coli serogroupsisolated from pigs with diarrhoea. Journal of Clinical Microbiology.1991; 29:745-752.

Phase 2 & 3 Vaccine Research Agenda in the Kintampo District of Ghana.S. Owusu-AgyeiMinistry of Health, Kintampo Health Research Centre, Kintampo, GHANA.

The Kintampo Health Research Centre (KHRC), is set-up todevelop and evaluate public health interventions to influence local andinternational health policies. The district population of 150,000 live inuniquely numbered households under a continuous demographicsurveillance. An extension of active surveillance is in three adjacentdistricts, leading to 600, 000 human-population under demographicsurveillance. This area experience annual rainfall of 1280mm. Malaria is

P20

P21

P22

Page 75: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

75

(continued)

the leading cause of morbidity and mortality in the district withfalciparum malaria as the predominant species. Malaria is perennial withprevalence ranging between 45 and 84%. Parasitological resistance is35% for chloroquine and 15% for sulphadoxine-pyrimethamine.Incidence of malaria is 8 clinical attacks per child per year, andentomological inoculation rate is 268 infective bites per person peryear.Having carried out vaccine and drug trials in the past on theimmunogenicity, reactogenicity and safety of GSK Nm ACW135polysaccharide vaccine with Nm ACYW135 polysaccharide vaccine aswell as the efficacy and safety of Artesunate Combination Therapy trials,KHRC is in a position to carry out large-scale community trials onpromising malaria-vaccine candidates in the Kintampo district as theybecome available. This past experience is backed by the availability ofqualified scientists in KHRC including 3 epidemiologists, 3 publichealth physicians, 5 research fellows (demography/public health,nutrition and statistics) and about 45 assistant research officers, 7 datamanagers several fieldworkers; most of who have undergone GCPtraining. The centre is supported by a medical laboratory that conductsmicroscopy, PCR, ELISAs, hematology and biochemistry analyses.

A Multiplex Real-Time PCR Assay with An Internal Control forQuantitative Detection of Streptococcus pneumoniaeA. Hu, F. Li, P. Zhao, J. S. Tam, R. Rappaport, S. Cheng Wyeth, Pearl River, NY.

Background. A multiplex PCR assay was developed to permit timelydiagnosis of S. pneumonia in clinical specimens that often containinhibitors leading to misinterpretation of PCR results. To address thisissue, we included an internal control (IC) in our PCR procedure (1) toenable assessment of interference, if present, in clinical specimens.Methods. The green fluorescence gene (GFP) was selected as the IC anda primer/probe pair was designed for detection of the GFP usingTaqMan-based PCR. Three mono-specific PCRs, one for detection ofGFP and one each for the S. pneumoniae genes, autolysin (lytA) andpneumolysin (ply), were combined into a single tube triplex assay.Results. The limit of detection for GFP was 2.5 fg similar to that for thelytA and ply genes. More importantly, no significant differences in thedetection limit for all 3 genes were found in the triplex versus individualmono-specific PCRs. However, when nasal specimens from 12 healthysubjects were spiked with GFP and S. pneumoniae, about 10 fold fewercopies of GFP, lytA and ply were observed. This level of interference wasovercome by pre-treating specimens with Proteinase K before PCR.Conclusion. We developed a rapid and cost-effective single tube triplexreal-time PCR that includes GFP as IC and permits quantitativedetection of S. pneumoniae even in the presence of inhibitors.

Reference. 1. Li F, et al. Rapid, sensitive and quantitiative detection of streptococcus

pneumoniae using triplex real-time PCR. 105th ASM Meeting,Atlanta, Georgia.

Cloning and Characterization of the Polysaccharide BiosyntheticGenes for Shigella dysenteriae Serotype 1 into Salmonellaenterica serovar Typhi Ty21a D. Xu1, J. O. Cisar2, D. J. Kopecko1

1FDA-CBER, Bethesda, MD, 2NIH-NIDCR, Bethesda, MD.

Shigella dysenteriae type 1 causes a severe form of shigellosis and isclassified as a Category B bioterrorist threat agent. Thelipopolysaccharide (LPS) of S. dysenteriae 1 is essential for virulence, andthere is indirect evidence that antibodies against this O-specificpolysaccharide (O-Ps) are protective to the host. Thus, there isconsiderable interest in the development of an O-Ps-based vaccine toprotect against S. dysenteriae. Previous studies showed that thedeterminants for the production of O antigen LPS in S. dysenteriae type1 are distributed on the chromosome (i.e. rfb/rfc genes) and on a small9-kb plasmid (i.e. rfp gene). The current studies were aimed at cloningthe Rfb/Rfc region from strain 1617 to define all essential genes anddevelop a biosynthetic pathway for O-Ps biosynthesis. The plasmid-carried gene (i.e. the rfp-encoded galactosyl transferase) was also clonedfrom strain 1617; it’s 1.2 kb sequence was found to be >99%homologous to rfp previously analyzed from a different S. dysenteriae 1strain. Additionally, the chromosomal Rfb/Rfc region of 9 kb wascloned and sequenced, and found to contain 9 ORFs. Preliminaryanalysis suggests that all 9 ORFs plus rfp are necessary for biosynthesisof serotype 1 LPS, which is presented as core-linked LPS in the Ty21avaccine vector. We anticipate that the use of these characterized O-Psgenes in a live, attenuated Salmonella delivery system will lead to a safe,oral vaccine for protection against this severe form of shigellosis

Reference:1. Xu DQ, Cisar JO, Ambulos Jr N Jr, Burr DH, Kopecko DJ.

Molecular cloning and characterization of genes for Shigella sonneiform I O polysaccharide: proposed biosynthetic pathway and stableexpression in a live salmonella vaccine vector. Infect Imm

Pneumococcal Disease and Influenza Vaccination Acceptanceamong Health Care WorkersJ. Wallenfels1, J. Rames2

1Ministry of Health of the Czech Republic, Praha, CZECH REPUBLIC, 2Instituteof Hygiene and Epidemiology, First Faculty of Medicine, Charles University inPrague, Praha, CZECH REPUBLIC.

Background: Health care workers (HCW) may be at increased riskfor contracting influenza, especially during nosocomial outbreaks; theyalso may serve as vectors for transmitting influenza to others, includinghigh-risk patients. On that account vaccination of all HCW employedin long-term care facilities against influenza and diseases caused byStreptococcus pneumoniae as well is obligatory and free of charge in theCzech Republic (according to valid legislation) since 2000. In spite ofthat many of them fail to receive the vaccines. In the hope thatknowledge of factors inhibiting HCWs‘ participation in vaccineprogrammes could help to improve vaccination rates, we conducted inlong-term care facilities in 2003 a cross-sectional survey of HCWs‘knowledge, attitudes, and influenza and pneumococcal diseasevaccination status. Methods: An anonymous, self-administeredquestionnaire (49 questions). Results: There were 1 296 respondents.The response rate was 69.2%. The mean age of the respondents was45.8 years. 86.3% were females. 52.3% were nurses. The influenza

P23

P24

P25

Page 76: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

76

Eighth Annual Conferencevaccination rate was 52.7%, the pneumococcal disease vaccination ratewas 40.3%. Avoiding influenza and pneumococcal diseases was quotedas the most important motive for acceptance of vaccination. Believingthat immune system will protect against influenza even if notvaccinated, was identified as the most important factor associated withrefusal of influenza vaccine. Fear of side effects was identified as themost important factor associated with refusal of pneumococcal diseasevaccine. Conclusions: Many HCW fail to receive influenza (andpneumococcal disease) vaccine. Strategies to improve immunizationlevels should address concerns about vaccine safety and efficacy,misconception about prevention of influenza, barriers to vaccination,and the reasons for targeting healthcare workers.

Burden of Invasive Disease Caused by Haemophilus influenzae Typeb and Streptococcus pneumoniae Among Infants in Bamako, MaliS. O. Sow1, J. Campbell2, M. Tapia2, S. Diallo3, K. Kotloff2, M. Levine2

1Centre pour le Developpement des Vaccins, Bamako, MALI, 2University ofMaryland School of Medicine, Center for Vaccine Development, Baltimore,MD, 3Clinical Bacteriology Laboratory and the Pediatric Service, Gabriel TouréHospital, Bamako, MALI.

Background: Immunization with H. influenzae type b (Hib)conjugate is routine in industrialized countries and pneumococcalconjugate vaccines are increasingly used. In developing countries thepaucity of quantitative data on invasive Hib and pneumococcal diseasehas impeded decisions about whether to introduce routineimmunization against these infections. Methods: Children age 0-15years with fever > 39o C or syndromes compatible with invasivebacterial disease were eligible. Blood and relevant body fluids (e.g.cerebrospinal fluid (CSF)) were cultured. Bacteria were identified bystandard microbiologic techniques. Results: From June, 2002 to May,2004, 3,381 Bamako infants age 0-11 months were admitted to HôpitalGabriel Touré. 1,728 infants were eligible and 1,547 (89.5%) wereenrolled. Among these, 185 (11.9%) died. Hib was isolated from 159infants (10.3%) including 104 positive CSF cultures, 17 infants died(10.7%). S. pneumoniae was isolated from 117 (7.6%) infants including85 from CSF, 22 of these died (18.8%). Most infections (148 Hib and62 S. pneumoniae) occurred in infants age 4-11 months; 17.6% of alladmissions in this age group were due to invasive Hib andpneumococcal infections. All Hib related deaths and 11 of thoseassociated with S. pneumoniae occurred among infants age 4-11 months,accounting for 35% of all deaths observed among enrollees in this agegroup. Conclusions: Hib and pneumococcal infections were majorcauses of hospitalization and death among infants in Bamako andindicate a need to introduce immunization against these infections.Since most invasive Hib and pneumococcal disease occurs beyond 4months of age, high coverage through routine immunization at 6, 10and 14 weeks of age should prevent most of these infections.

References:1. Campbell JD, Kotloff KL, Sow SO, et al. Invasive Pneumococcal

infections among hospitalized children in Bamako, Mali. PediatrInfect. Dis. J 2004; 23 (7):642-9.

2. Sow S, Diallo S, Campbell J, et al. Burden of Invasive Disease Causedby Haemophilus influenzae type b in Bamako, Mali: Impetus forRoutine Infant Immunization with Conjugate Vaccine. Accepted forpublication.

Standardized Informed Consent for Low-literacy Audiences inAfrican Research EnvironmentB. E. Bekan Projet San Francisco, Kigali, RWANDA.

Background Some Institutional Review Boards recommend a 6-8thgrade level for informed consent processes in research involving humansubjects (1). “Grades” are not applicable to Africa where the literacylevel is below 50%. Most research is often developed in non-Africanlanguages. It is important to standardize the consent form process toensure that the meaning is understood at all literacy levels. Methods:Projet San Francisco (PSF) in Rwanda translates the consents intoKinyarwanda, the main language. Videos of counselors reading theconsent are shown during an interactive group session, written copiesgiven to participants to consult. Counselors invite questions thatenhance comprehension. An “ understanding of informed consent” isadministered to volunteers’. Results are provided using descriptivefigures. Results: More than 100 volunteers go through the informedconsent process each day for different studies at PSF’s research sites. Tenare potential volunteers for a protocol establishing normal laboratoryvalues for Rwanda as part of preparedness activities for HIV vaccinetrials. During the first two weeks of that study, 48 volunteers werescreened, 85% were enrolled. Less than 0.05 % was eliminated becausethey did not understand the consent. Conclusions: The informedconsent is particularly challenging in African research settings. Thestandardization results in time saving, and prevents subtleties inlanguage from hindering comprehension.

Reference:1. University of Chicago, Institutional Review Board Social and

Behavioral Sciences. Policies: Informed Consent. Access on December27th 2004.http://humansubjects.uchicago.edu/sbsirb/manual/consent_policies.shtml

Compatibility of Co-administered 7-valent PneumococcalConjugate, DTaP.IPV/PRP-T Hib and Hepatitis B Vaccines inInfantsD. W. Scheifele1, S. Halperin2, B. Smith2, K. Meloff3, D. Duarte-Monteiro3; 1University of British Columbia, Vancouver, BC, CANADA, 2Dalhousie University,Halifax, NS, CANADA, 3Wyeth Pharmaceuticals, Toronto, ON, CANADA.

Current immunization schedules for infants recommendsimultaneous administration of several vaccines. This study assessed thecompatibility of concurrently administered 7-valent pneumococcalconjugate (PCV7), hepatitis B (HB) and DTaP.IPV/Hib (P5) vaccines.Healthy infants were enrolled at 2 months of age and randomly assigned(2:1) to receive P5 and HB at 2,4,6 months, with PCV7 givenconcurrently or sequentially (at 3,5,7 months). Adverse events weremonitored for 3 days after each vaccination. Blood was obtained beforedose 1 and after dose 3 to measure antibody responses to each antigen.376 infants were enrolled and 368 completed the full protocol,including 246 concurrent and 122 sequential vaccinees. Responses toPCV7 were unaltered by concurrent P5 and HB vaccinations. Responsesto P5 were generally unaltered by concurrent PCV7 vaccination exceptHib anti-PRP responses were enhanced (final GMC 1.11 vs 0.64 ug/mL[concurrent vs sequential], p=0.008). Sequential vaccination increasedresponses to diphtheria as a result of repetitive stimulation by toxoid andCRM197. Responses to HB vaccine were greater with sequential (when

P26

P27

P28

Page 77: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

77

(continued)

HB and P5 were injected in opposite thighs) than concurrentvaccinations ( HB and P5 given adjacent, same thigh) . Concurrentvaccinations were well tolerated, with no increase in the rate of fever.We conclude that PCV7 and DTaP.IPV/Hib are compatible whenadministered concurrently to infants in opposite thighs. Injecting HBvaccine adjacent to DTaP.IPV/Hib may decrease responses to HB.“Adjacency” effects on responses warrant further study.

References:1. Halperin SA et al. Safety and immunogenicity of two acellular

pertussis vaccines with different pertussis toxoid and FHA content ininfants 2-6 months old. Scand J Infect Dis 1995;27:279-87

2. Heubner RE et al. Immunogenicity after1,2or 3 doses and impact onthe antibody response to co-administered antigens of a nonavalentpneumococcal conjugate vaccine in infants in Soweto, South Africa.Pediatr Infect Dis J 2002;21:1004-7

Research Subject Satisfaction Assessment: a Missing Element of“Good Clinical Practices”C. LaJeunesse, A. Kallos, K. Marty, M. Mozel, D. W. ScheifeleVaccine Evaluation Center, University of British Columbia, Vancouver, BC,CANADA.

Regulatory agencies such as Health Canada and the FDA shapeclinical practice for conducting research in humans. Their regulationsare based on Good Clinical Practice Guidelines ( GCP) from theInternational Conference on Harmonization. GCPs require clinicalresearch professionals to ensure that subjects’ rights, dignity and safetyare respected from the time of recruitment to study completion. Whilemost researchers believe that they adhere to accepted guidelines, anevaluation of their success by study participants is not required. Wedeveloped an assessment tool and anonymously surveyed thesatisfaction of parents of 698 children as they completed studyparticipation. The 3 vaccine trials ( 2 sponsored, one grant-funded)included 169 toddlers, 155 pre-kindergarten and 374 grade 6 children,respectively, and involved 1-3 vaccination visits and two venipuncturesper subject. Compliance with survey completion averaged 66% ,ranging from 41% by mail to 98% when done at the final visit. Parentsrated their level of satisfaction with several aspects of the study relatedto GCPs using a Likert scale. For example, 438 ( 96.7 %) felt theyunderstood the study at entry and 433 (95.4%) felt well informedduring the study. Overall, 94% of parents felt participation wasbeneficial and 91% would consider participating in a future study. Theresults indicate that our team provided an explicit, respectful and safeexperience for participants, as intended. We believe that end of studyevaluations from subjects should be a standard practice, added to GCPcodes.

References:1. Reynolds SM ORI findings of scientific misconduct in clinical trials

and publicly funded research, 1992-2002. Clinical Trials2004;1:509-516

2. Knatterrud GL et al Guidelines for quality assurance in multicentertrials: a position paper. Control Clin Trials 1998;19:477-93

Comparison of the Safety and Immunogenicity of Simultaneous orSequential Administration of an Adult Formulation Tdap Vaccineand Influenza VaccineS. McNeil1, F. Noya2, M. Dionne3, G. Predy4, W. Meekison5, C. Ojah6, S. Ferro7, E. Mills7, J. Langley1, S. Halperin1; 1Dalhousie University, Halifax, NS, CANADA, 2Montreal Children’s Hospital,Montreal, PQ, CANADA, 3INSPQ, Quebec City, PQ, CANADA, 4Capital-Health,Edmonton, AB, CANADA, 5Westcoast Clinical Research, Vancouver, BC,CANADA, 6Saint John Regional Hospital, Saint John, NB, CANADA, 7Sanofi-Pasteur, Toronto, ON, CANADA.

Background: The annual contact for influenza vaccination providesan opportunity to ensure that adults have received other recommendedvaccines such as Tdap. Methods: Healthy 19-64-year-olds wererandomized to concurrent administration of Tdap and influenza vaccine(GroupA) or influenza vaccine followed in 4 to 6 weeks by Tdap(GroupB). Results: 720 participants were enrolled. No differences wereobserved in the rates or severities of injection-site (Tdap-site) erythema,swelling, or pain. Injection-site pain was the most commonly reportedadverse event (66.6% GroupA v. 60.8% GroupB); most pain was gradedas mild and resolved by Day 3. Seroprotection and seroresponse rates forall influenza strains were comparable between the 2 groups. Fordiphtheria and tetanus, seroprotection rates and post-vaccination GMTswere non-inferior in GroupA compared to GroupB. A trend for lowerantibody responses to pertussis antigens PT, FHA, and FIM wasobserved after concomitant administration and, for PRN, this differencemet statistical significance. However, GMTs to all antigens in bothgroups exceeded levels achieved by infants in pre-licensure, effacy-trials.Conclusion: While there is a small diminution in antibody response,concomitant administration of Tdap and influenza vaccine was well-tolerated and immunogenic, and may offer practical advantagesincluding convenience, compliance, and cost-savings.

References:1. De Serres G, Shadmani R, Duval B, et al. Morbidity of pertussis in

adolescents and adults. J Infect Dis 2000;182:174-92. Gustafsson L, Hallander HO, Olin P, et al. A controlled trial of a

two-component acellular, a five-component acellular, and a whole-cellpertussis vaccine. N Engl J Med 1996. 334(6): 349-55.

Measurement of Tetanus Antitoxin in Oral Fluid: A Novel Methodto Evaluate Vaccination ProgramsM. D. Tapia1, L. Cuberos1, S. O. Sow2, M. N. Doumbia2, M. Bagayogo2, M.Pasetti1, K. Kotloff1, M. Levine1

1University of Maryland School of Medicine, Baltimore, MD, 2Centre pour leDeveloppement des Vaccins - Mali, Bamako, MALI.

Background: Oral fluid has been used in surveys and detection ofresponses to vaccination. Measurement of tetanus antitoxin may serve asan objective means to assess the effectiveness of vaccination programs. Asurvey was conducted to determine whether IgG tetanus antitoxin couldbe measured in oral fluid. Methods: Serum and oral fluid were collectedfrom infants, toddlers and adults (males without history of vaccinationagainst tetanus), residing in rural Mali. IgG tetanus antitoxin wasmeasured by standard enzyme linked immunosorbent assay in serum (S-ELISA) and oral fluid (OF-ELISA). Results: 215 pairs of serum andoral fluid samples were collected. There was approximately 10-fold lessIgG tetanus antitoxin measured by OF-ELISA than S-ELISA. There was

P29

P30

P31

Page 78: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

78

Eighth Annual Conferencean excellent correlation between anti-tetanus IgG measured by S-ELISAand OF-ELISA (r=0.83, p < 0.001). OF-ELISA was 82% sensitive and100% specific compared to S-ELISA for the detection of protectivetiters (0.001 IU/ml in oral fluid corresponded with 0.01 IU/ml inserum). Infants and toddlers who had received 1, 2, or 3 doses ofdiphtheria-tetanus-pertussis (DTP) vaccine had increasing geometricmean concentrations of tetanus IgG detected by S-ELISA (1.3, 2.8 and3.3 IU/ml, respectively) and OF-ELISA (0.006, 0.0171, and 0.0175IU/ml). Conclusions: Levels of IgG tetanus antitoxin in oral fluidcorrelate well with levels in serum. OF-ELISA offers a non-invasiveobjective tool to supplement information collected in coverage surveys.

References:1. Morris-Cunnington MC, Edmunds WJ, Miller E, Brown DW. A

population-based seroprevalence study of hepatitis A virus using oralfluid in England and Wales. Am J Epidemiol. 2004 Apr15;159(8):786-94.

2. Zhang Q, Lakshman R, Burkinshaw R, et al. Primary and boostermucosal immune responses to meningococcal group A and Cconjugate and polysaccharide vaccines administered to universitystudents in the United Kingdom. Infect Immun. 2001Jul;69(7):4337-4

Studies on a Live Oral Attenuated Cholera Vaccine, Peru-15 inBangladeshF. Qadri1, M. I. Chowdhury1, M. A. Salam1, S. M. Faruque1, T. Ahmed1, Y. A. Begum1, A. Saha1, L. V. Seidlein2, R. F. Breiman1, J. J. Mekalanos3, J. D. Clemens2, K. P. Killeen4, D. A. Sack1

1ICDDR,B: Centre for Health and Population Research, Dhaka, BANGLADESH,2International Vaccine Institute, Seoul, DEMOCRATIC PEOPLE’S REPUBLIC OFKOREA, 3Harvard Medical School, Boston, MA, 4AVANT Immunotherapeutics,Needham, MA.

Background: The objective was to facilitate the development of a newlive oral cholera vaccine that is safe, immunogenic and protective againstcholera caused by the V. cholerae O1 El Tor biotype, the pandemic strainnow causing cholera. Progress has been made on Peru-15a, derived froma V. cholerae O1 El Tor strain which was previously demonstrated to beimmunogenicb and efficacious against O1 El Tor cholera in clinical trials.Methods: A randomized double-blind placebo controlled study wascarried out by the ICDDR,B in Dhaka in collaboration withInternational Vaccine Institute and AVANT Immunotherapeutics Inc.The study was carried out in Dhaka in adults (n=70) and children(n=120) using Peru-15 at a dose of 2x108CFU. Results: Peru-15 wasfound to be safe no significant increase in symptoms attributed to thevaccine.Over 75% seroconversion rates were documented in vibriocidalantibodies with _ 256-fold increases in titers over baseline. It alsostimulated antibody responses to lipopolysaccharide.The strain remainedunchanged in genetic and phenotypic properties on excretion.Conclusions: Based on these encouraging results, the study hasprogressed to the infant phase(9-23m).We hope the informationobtained from these studies will be useful in planning future studies withit in Bangladesh.

References:1. Kenner JR, Coster TS, Taylor DN, et al. Peru-15, an improved live

attenuated oral vaccine candidate for Vibrio cholerae O1. J Infect Dis1995;172:1126-9

2. Sack DA, Sack RB, Shimko J, et al. Evaluation of Peru-15, a new liveoral vaccine for cholera, in volunteers. J Infect Dis 1997;176:201-5

A Novel Combination DNA and Inactivated Rabies Virus VaccineP. N. Rangarajan1, V. A. Srinivasan2

1Indian Institute of Science, Bangalore, INDIA, 2Indian Immunologicals Limited,Hyderabad, INDIA.

Background: The high cost of production of cell culture-derivedrabies vaccines render them unaffordable to a sizeable population in thedeveloping countries where rabies remains a serious public healthproblem. DNA vaccines can be produced at a low cost and are storableat room temperature and thus is an ideal choice for rabies eradicationprogrammes in developing countries. Methods: A novel combinationrabies vaccine (CRV) formulation containing an eukaryotic expressionplasmid encoding rabies virus surface glycoprotein and a small quantityof cell culture-derived inactivated rabies virus was developed. Thepotency of CRV was examined in mice and cattle by examining therabies virus neutralizing antibody titres as well as protection againstvirus challenge. Results: Mice immunized with CRV develop higherlevels of rabies virus neutralizing antibodies (RVNA) than thoseimmunized with DNA Rabies vaccine (DRV) and are completelyprotected against peripheral as well as intracerebral rabies viruschallenge. The quantity of inactivated rabies virus vaccine required forenhancing the potency of DRV can be 625 fold lower than that of astandard dose of inactivated rabies virus vaccine. CRV induces higherlevels of RVNA than DRV in cattle as well. Conclusions:Coinoculation of DNA vaccine and a low dose of inactivated virusvaccine can be developed into a novel cost-effective vaccination strategyfor combating rabies in particular and infectious diseases in general.

Reference:1. Biswas S, Reddy, GS, Srinivasan VA, Rangarajan PN. Pre-exposure

efficacy of a novel combination DNA and inactivated rabies virusvaccine. Hum. Gene Ther. 2001;12:1917-1922

Minicircle DNA Immobilized in Bacterial Ghost: A Novel Non-livingBacterial DNA-vaccine Carrier SystemC. Azimpour Tabrizi1, W. Jechlinger2, P. Becker3, T. Ebensen3, C. Guzman3, W. Lubitz1

1Microbiology and Genetic, Vienna, AUSTRIA, 2Inst. Bacteriology, Mycology andHygiene, Vienna, AUSTRIA, 3German Research Centre for Biotechnology,Braunschweig, GERMANY.

Despite the exponential rate of discovery of DNA vaccines resultingfrom modern molecular biology, the lack of the effective deliverytechnology is a major limiting factor in their applications. We hadrecently reported a new platform technology based on theimmobilization of plasmid DNA in the inner membrane of bacterialghosts (SIP)1. Furthermore, this system was combined with an in vivosite-specific recombination technology, in which an origin plasmiddivides into a minicircle and a miniplasmid. The replicativeminiplasmid containing the origin of replication and the antibioticresistance gene was lost during the bacterial ghost production2, whereasthe minicircle DNA containing only the eukaryotic expression cassettewas retained in the bacterial ghosts via the SIP technology. In vivostudies showed that immunization of C57Bl6 mice with bacterial ghostsharbouring immobilized minicircle encoding ovalbumin, results inhumoral and cellular immune responses against the target antigen.Immune responses stimulated by ghost-mediated DNA delivery wereevaluated by ELISA and ELISPOT assay to show the capacity of T-cellsto produce Antibodies and cytokines, like IL-2, IL-4 and IFN-_. The

P32

P33

P34

Page 79: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

79

(continued)

significant amount of the IgG and IL-4 in the immunized mice showedthe bias of the immune response toward Th2 responses and proliferationof the B-cells. In addition, the efficient IFN-_ production indicated thatthe bacterial ghosts as DNA delivery vehicles are also able to effectivelystimulate the cellular arm of the immune response.

References:1. Mayrhofer P, Azimpour T.C, Walcher P, Haidinger W, Jechlinger W,

Lubitz W. Immobilization of plasmid DNA in bacterial ghosts. JControl release 2004; available online.

2. Azimpour T.C, Walcher P, Mayr U.B, Stiedl T, Binder M, McGrathJ.F, Lubitz W. Bacterial ghosts-Biological particles as delivery systemsfor antigens, nucleic acids and drugs. Curr Opinn Biotech 2004,15(6): 530-537.

Multiple DNA Vaccine Plasmids Protect Mice from AcutePulmonary Infection of Pseudomonas aeruginosaS. Saha1, F. Takeshita1, S. Sasaki1, T. Matsuda1, T. Matsumoto2, K. Yamaguchi2, K. Okuda1

1Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 2Toho University School of Medicine, Tokyo, JAPAN.

We studied the immunogenicity of three DNA vaccine plasmidsencoding P. aeruginosa antigens, a fusion protein of outer membraneprotein F and outer membrane protein I (OprF/I), type III translocationprotein (PcrV), and pilin protein (PilA). BALB/c mice were immunizedtwice with single (OprF/I, PcrV, or PilA alone) or mixture of threeplasmids (OprF/I+PcrV+PilA) by using intramuscular electroporation(imEP) technology. Two weeks after 2nd immunization, mice wereadministered intarnasally with P. aeruginosa D4. The higher number ofthe lung bacteria was recovered in groups of mice vaccinated with eachsingle plasmid than OprF/I+PcrV+PilA group. Bacteremia was presentin all groups except OprF/I+PcrV+PilA group. The expression of MIP-2,TNF_, and IFN_ mRNA in broncho alveolar lavage cells weresignificantly upregulated in OprF/I+PcrV+PilA group. Finally >10 dafter bacteria challenge, the superior survival rate was observed inOprF/I+PcrV+PilA group compared with that in the group of micevaccinated with each single plasmid.The method of vaccine delivery was also examined. Multiple plasmidDNA vaccine by imEP method provided potent protection against P.aeruginosa challenge over that by gene gun method although serum IgGlevels were comparably induced by both methods.In conclusion, multiple DNA vaccine targeting three differentcomponents, OprF/I, PcrV, and PilA, delivered by imEP elicited thestrongest protection ability. These data would assist the development ofP. aeruginosa vaccine for pulmonary infections.

References:1. Sawa T, Yahr TL, Ohara M, Kurahashi K, Gropper MA, Wiener-

Kronish JP, and Frank DW. Active and passive immunization with thePseudomonas V antigen protects against type III intoxication andlung injury. Nature Medicine 1999; 5:392-398

2. Grifantini R, Finco O, Bartolini E, Draghi M, Giudice GD, KockenC, Thomas A, Abrignani S and Grandi G. Multi plasmid DNAvaccination avoids antigenic competition and enhancesimmunogenicity of a poorly immunogenic plasmid. Eur. J. Immunol1998; 28:1

Infanrix™-IPV-Hib (GSK) is Safe and Immunogenic Compared toPentacel™ (Sanofi Pasteur) as a 4th Dose in 15-20 Month OldsS. A. Halperin1, B. Tapiero2, B. Law3, B. Duval4, F. Diaz-Mitoma5, D. Elrick6; 1Pediatrics, Dalhousie University, Halifax, NS, CANADA, 2Pediatrics, Ste JustineHospital, University of Montreal, Montreal, PQ, CANADA, 3Pediatrics,University of Manitoba, Winnipeg, MB, CANADA, 4Institut National de SantéPublique, Quebec City, PQ, CANADA, 5Pediatrics, Children’s Hospital of EasternOntario, Ottawa, ON, CANADA, 6GlaxoSmithKline, Inc, Mississauga, ON,CANADA.

Background: In Canada, children are immunized using Pentacel™.GSK also produces a pentavalent DTPa-IPV-Hib (Infanrix™-IPV-Hib).Methods: 15-20 month olds previously immunized with 3 doses ofPentacel™ were randomised to receive 1 dose of Pentacel™ orInfanrix™-IPV-Hib. Results: 433 participants were enrolled. Redness>20 mm was reported after 11.5% of Pentacel™ and 5.6% ofInfanrix™-IPV-Hib (p=.038). Injection site pain was more commonafter Pentacel™ (52.1%) than Infanrix™-IPV-Hib (39.4%; p=.009).Moderate or greater drowsiness was more common after Pentacel™(13.8%) than Infanrix™-IPV-Hib (7.4%; p=.042). The proportion ofparticipants that were seroprotected and GMTs were similar fordiphtheria, tetanus, and polio. Similar proportions responded topertussis antigens; GMTs were higher in the Infanrix™-IPV-Hib thanthe Pentacel™ group against pertussis toxoid (88.5 vs. 65.6 EU/mL),filamentous hemagglutinin (207.3 vs. 132.1) and pertactin (251.9 vs.166.9). The proportion seroprotected against Hib was similar (99.5%vs. 98.4%); GMTs were higher in the Pentacel™ (29 _g/mL) than theInfanrix™-IPV-Hib (19 _g/mL) group. Conclusion: A booster dose ofInfanrix™-IPV-Hib after 3 priming doses of Pentacel™ is safe andimmunogenic in 15-20 month old infants. Infanrix™-IPV-Hib can beused as an alternative to Pentacel™ for the 4th dose in infants primedwith 3 doses of Pentacel™.

References:1. Halperin SA, Davies HD, Barreto L, et al. Safety and

immunogenicity of two inactivated poliovirus vaccines incombination with an acellular pertussis vaccine and diphtheria andtetanus toxoids in seventeen month old infants. J Pediatr.1997;130:525-3

2. Halperin SA, King J, Law B, Mills E, Willems P. Safety andimmunogenicity of Haemophilus influenzae-tetanus toxoid conjugatevaccine given separately or in combination with a three-componentacellular pertussisvaccine...Clin Infect Dis. 1999;28:995-1

Immunization with Recombinant Adenovirus SynthesizingSecretory Form of Japanese Encephalitis Virus Envelope ProteinProtects Adenovirus-Exposed Mice Against Lethal EncephalitisS. VratiNational Institute of Immunology, New Delhi, INDIA.

Background: We have previously shown that immunization with aplasmid DNA synthesizing pre-Membrane (prM) and envelope (E)proteins of Japanese encephalitis virus (JEV) provided partial protectionto mice against lethal JEV challenge [1]. An improved delivery of DNAencoding these two proteins using recombinant adenovirus (RAd) islikely to induce enhanced immune response against JEV [2]. Methods:Replication-defective RAds were constructed that synthesized the prMand E proteins of JEV. Potential of these recombinants for vaccination

P35

P36

P37

Page 80: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

80

Eighth Annual Conferenceagainst JEV was examined in mice by studying their immunogenicityand protective efficacy. Results: Recombinant virus RAdEa,synthesizing the membrane-anchored E protein, replicated poorly inHEK 293A cells and synthesized lower amounts of E protein thanRAdEs synthesizing the secretory E protein. Oral immunization of micewith RAds generated low titers of anti-JEV antibodies with little JEVneutralizing activity. Intra-muscular (IM) immunization of mice withboth RAds generated high titers of anti-JEV antibodies. Interestingly,RAdEa induced only low titers of JEV neutralizing antibodies, whereasthese were significantly higher in case of RAdEs immunization. Naïvemice immunized IM with RAdEs showed complete protection againstlethal JEV challenge. In order to study the effect of the pre-existingAdenovirus 5 (Ad5) immunity on the outcome of RAdEs immunization,mice were exposed to Ad5 through IM or intra-nasal (IN) routes beforeimmunization with RAdEs. Mice exposed to Ad5 through the IN route,when immunized IM with RAdEs, or those exposed to Ad5 through theIM route, when immunized IN with RAdEs, showed completeprotection against lethal JEV challenge.Conclusions: High levels ofprotective immunity induced by RAdEs in Ad5-exposed mice by the IMimmunization point to its potential as a candidate JEV vaccine.

References:1. Kaur R, Sachdeva G, Vrati S. Plasmid DNA immunization against

Japanese Encephalitis Virus: immunogenicity of membrane-anchoredand secretory envelope protein. J Infect Dis 2002; 185:1-12.

2. Casimiro DR, Chen L, et al. Comparative immunogenicity in rhesusmonkeys of DNA plasmid, recombinant vaccinia virus, andreplication-defective adenovirus vectors expressing a humanimmunodeficiency virus type 1 gag gene. J Virol 2003; 77:6305-6313.

A Novel Live Adenovirus Vaccine Vector Prototype: High-level Antigen Production from the Adenoviral Major LateTranscriptional UnitM. G. Berg1, B. Falgout2, G. W. Ketner1

1Molecular Microbiology and Immunology, Johns Hopkins University,Baltimore, MD, 2Cber, Food & Drug Administration, Bethesda, MD.

Safe, effective, orally-delivered live adenovirus vaccines (Ad4 & Ad7)have been used for three decades and recombinant derivatives of thesemay prove a novel approach to vaccine development or an economicalalternative to some current vaccines. We sought to design a replicatingvector system that is inexpensive, produces high antigen levels, andprotects in a single dose. As a proof of principle, we constructed a seriesof novel Ad5 recombinants that express the major capsid protein (L1) ofcanine oral papillomavirus (COPV), a model for mucosal humanpapillomavirus (HPV) infection. Our recombinants incorporate COPVL1 into adenovirus late region 5 (Ad L5) so that it is expressed as amember of the adenoviral MLTU. COPV L1 production is influencedby Ad L5 gene order, the specific mRNA processing signals associatedwith COPV L1, and the state of a putative splicing inhibitor in theCOPV L1 gene. Recombinant COPV L1 protein assembles into VLPsand also reacts with an antibody specific for conformational epitopes onnative COPV L1 protein that correlate with protection in vivo. We havenow built similar prototype vaccines intended for the prevention ofcervical cancer as well as dengue fever. These express L1 from the highrisk serotype, HPV16, or the Dengue1 antigens, pre-M and E. Our dataillustrates the versatility of this expression system and the possibility forthe construction of recombinant adenovirus vaccines against a variety ofdiseases.

References:1. Koutsky LA, Ault KA, Wheeler CM, et al. A controlled trial of a

human papillomavirus type 16 vaccine. N Eng J Med 2002;347(21):1645-1651.

2. Suzich JA, Ghim SJ, Palmer-Hill FJ, et al. Systemic immunizationwith papillomavirus L1 protein completely prevents thedevelopment of viral mucosal papillomas. Proc Natl Acad Sci USA1995; 92: 11553-11557.

Construction of Live Attenuated Shigella Vaccine Candidates using RecombineeringR. Ranallo, S. Barnoy, S. Thakkar, M. Venkatesan Enteric Infections, WRAIR, Silver Spring, MD.

Live attenuated Shigella vaccines have shown promise in inducingprotective immune responses and as carriers of heterologous antigensfrom other mucosal pathogens. In the past, construction of Shigellavaccine strains has relied on classical allelic exchange systems to transferalleles from one strain to another. These systems require extensive invitro engineering of long homologous sequences to create recombinantreplication-defective plasmids or phage. Alternatively, the Lambda redrecombination system from bacteriophage Lambda facilitatesrecombination with as little as 50bp of homologous DNA. However,until recently this system has been restricted for use in E. coli. Theprocess, referred to as recombineering, typically uses an inducible red-gam operon on a temperature-sensitive plasmid and optimaltransformation conditions to integrate linear antibiotic resistancecassettes into bacterial genomes. Recent advances in the Lambda redrecombination system have enabled modification of genomic DNAfrom bacterial pathogens such Salmonella typhimurium, EPEC, orEHEC using short regions of homology thus eliminating the tedious invitro steps associated with plasmid or phage construction. We have usedthese advances in Lambda red recombineering to delete virulence-associated genes from Shigella creating a number of isogenic strainsfrom multiple Shigella species. These strains have been characterized forattenuation using both in vivo and in vitro assays. Based on this data,prototypic Shigella vaccine strains containing multiple deletions invirulence-associated genes have been created. Thus through protocoloptimization and newly available plasmids, we have used Lambda redrecombineering to rapidly construct several potential Shigella vaccinestrains with up to four precise genetic lesions.

References:1. Murphy, K.C. and K. G. Campellone. Lambda Red-mediated

recombinogenic engineering of enterhemorrhagic andenterpathogenic E. coli. BMC Mol Biol. 2003 Dec 13;4(1):11.

2. Court D.L., Sawitzke J.A., and L.C. Thomason. GeneticEngineering Using Homologous Recombination. Annu. Rev. Genet.2002. 36:361-88.

P38

P39

Page 81: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

81

(continued)

Oral Immunization of Dogs with Baits Containing the Recombinant Rabies Virus Glycoprotein/ Nucleoprotein-canineAdenovirus Type 2R. Hu, S. Zhang, H. li Veterinary Institute, Academy of Military Medical Science, Changchun, CHINA.

Background: Human rabies is mostly caused by bite of rabid dogs.Immunization of dogs in countryside with currently commercial rabiesvaccine is laborious and costly. To develop an oral rabies vaccine effectiveand easily used for dogs, we developed two recombinant canineadenoviruses type-2 expressing rabies virus glycoprotein andnucleoprotein, respectively. Methods: The recombinant viruses wereconstructed by cloning the rabies virus glycoprotein and nucleoproteinexpression cassette into the deleted E3 region of canine adenovirus type-2,and were packaged, propagated and produced on MDCK. Therecombinant vaccine baits, which are made by injecting 3_108.5 TCID50recombinant viruses into tampon enwrapped by fat with a T-shape metalframe attached, were fed to dogs and tested for safety and efficacy. Results:Two weeks after initial oral administration, 50% of the immunized dogsdeveloped antibodies against rabies virus and the vector adenovirus. Afterbooster with an interval of two weeks, 85% of the immunized dogsbecame seroconverted. The neutralizing antibody, tested by mouseintracerebral neutralization, showed that 5_22 diluted serum couldneutralize 300 LD50 CVS-240 intracerebral infection. The immunizeddogs showed no clinical and pathogenic changes after the inoculation.Conclusion: It has demonstrated that the recombinant rabies virusglycoprotein and nucleoprotein-canine adenovirus vaccine baits are of oralsafeness, effectiveness and ease for rabies prevention in dogs.

Reference:1. Xiang ZQ, Gao GP, Reyes-Sandoval A, et al. Oral vaccination of mice

with adenoviral vectors is not impaired by preexisting immunity tovaccine carrier. J Virol 2003; 77:10780-10789.

Immunological Properties of Conjugates Prepared from PathogenicCandida Surface Antigens - Potential VaccinesS. Bystricky Institute of Chemistry SAV, Bratislava, SLOVAKIA.

Infections caused by opportunistic pathogens, namely by Candidaspecies occur increasingly because of the wide use of broad-spectrumantibiotics, chemotherapy and immunosuppressing drugs in the recentyears. The systemic diseases caused by pathogenic yeasts lead to life-threatening conditions in immunocompromised individuals, elderlypeople and infants. There is no effective licenced vaccine yet.

In our continuing conjugate studies1,2, newly prepared yeast surfacemannan - protein conjugates were used to immunize experimentalrabbits. Induction of specific humoral and celullar immune response wasmonitored. Quantitative determination of IgG and IgM by ELISA andflow-cytometry of CD4, CD8, CD19, CD25 and CD11b expression onlymphocytes and granulocytes was performed during whole multipleimmunizations experiment.

Conjugates elicited higher immune response, (especially after firstimmunization) than mannan antigen alone or killed whole cells.According to our results we can say, that novel mannan-proteinconjugates reflect T-cell dependent character of immunogens withevident induction of long-term memory functionality.

The yeast mannan conjugate synthesized by this scheme can beconsidered as a vaccine candidate for clinical evaluation.

Reference:1. Bystrick_ S., Paulovi_ová E., Machová E. Candida albicans mannan-

protein conjugate as vaccine candidate. Immun. Letters. 2003; 85:251-255

A Novel Subunit Vaccine Protects Mice Against Systemic Diseaseand Intestinal Colonization by Salmonella entericaL. Wonderling1, D. Straub2, D. Emery2

1Syntiron, Saint Paul, MN, 2Epitopix, Willmar, MN.

Salmonella enterica is a leading cause of bacterial foodborne diseasesworld wide. In response to the lack of immunoprophylactic treatmentand the need to identify potential vaccine targets for preventinginfections by Salmonella, our research efforts focused on thedevelopment of a novel subunit vaccine for controlling Salmonella. Ironuptake proteins have been identified as potent immunogens in othergram-negative species. Many of these proteins are large, surface-exposed,and expressed during infection of a mammalian host. S. enterica alsoexpresses a number of iron uptake proteins, and in the presentinvestigation we sought to determine if these proteins may constituteeffective vaccine antigens. Membrane proteins produced by S. entericaunder iron-limiting conditions were formulated into a vaccine andtested for efficacy in the murine host. When compared to unvaccinatedmice, the intraperitoneal (i.p.) administration of the multi-proteinvaccine to mice significantly (p≤ 0.035) reduced the fecal shedding ofSalmonella following an oral dose of the pathogen. In addition, thevaccine’s effects on systemic challenge were tested by i.p. administrationof vaccine and a subsequent i.p. challenge. The results show significantly(p= 2.06 x 10-13) fewer deaths in the group of mice administered thevaccine preparation as compared with unvaccinated mice. Takentogether, these results suggest that one or more proteins present in themembranes of S. enterica grown under iron-limiting conditions provideprotection against both an oral and systemic challenge of S. enterica.Ongoing investigations seek to identify the proteins responsible forprotection against S. enterica.

References:1. Mead, P.S., Slutsker, L., Dietz, V., McCraig, L.F., Bresee, J.S.,

Shapiro, C., Griffin, P.M., and Tauxe, R.V. 1999. Food-related illnessand death in the United States. Emerg. Infect. Dis. 5:607-625.

2. Bjarnason, J., Southward, C.M., and Surette, M.G. 2003. Genomicprofiling of iron-responsive genes in Salmonella enterica serovarTyphimurium by hihg-throughput screening of a random promoterlibrary. J. Bacteriol. 185:4973-7982.

Partial Protection of Mice after DNA Vaccination AgainstStaphylococcus aureus InfectionM. C. Gaudreau1, P. Lacasse2, B. G. Talbot1

1Biologie, University of Sherbrooke, Sherbrooke, PQ, CANADA, 2Agriculture and Agri-Food Canada, Lennoxville, PQ, CANADA.

Staphylococcus aureus is an opportunistic human and animalpathogen which can cause clinical manifestations ranging from mildskin infections to endocarditis, mastitis, septic arthritis and septicaemia.Our previous work demonstrated that the duration of Staphylococcusaureus mastitis in dairy cows could be reduced by immunization with

P40

P41

P42

P43

Page 82: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

82

Eighth Annual Conferenceplasmid that expressed bacterial adhesion proteins. The present reportdescribes the murine immune response and its effects on bacterialchallenge after immunization with genetic vaccines containing up tothree genes implicated in Staphylococcus aureus adhesion.

Mice were immunized three times with 100 ug of pCI-based(Invitrogen) plasmid DNA. Antibody titers were determined by ELISA.Cytokine analysis and cell proliferation assays were carried out by FACSanalysis. Mice were challenged by I.V. infection with either a humanisolate of Staphylococcus aureus or with a less virulent laboratory strain.

Antibody titers (primarily IgG2a) of up to 20,000 were producedagainst all antigens. Pre-incubation of the bacteria with immune serumconsiderably increased in vitro phagocytosis (p 0.01). FACScan analysisdemonstrated little antigen-induced lymphocyte proliferation but astrong stimulation of IFN_ compared to IL-4 production. Non-vaccinated mice infected with a virulent bacterial strain died within 4days, whereas the multigene vaccinated animals showed no signs ofinfection until 10 days post-infection. Similarly, only non-vaccinatedmice infected with the low virulence strain showed signs of septic orreactive arthritis

The results suggest that nucleic acid vaccination against adhesionproteins can produce a protective response against Staphylococcus aureusinfection.

References:1. Shkreta l., Talbot B.G., Diarra M.S., Lacasse P., Immune responses to

a DNA/protein vaccination strategy against Staphylococcus aureusinduced mastitis in dairy cows. Vaccine 2004; 23, 114-126

2. Brouillette E., Lacasse P., Shkreta, L., Grondin G., Fournier S.,Bélanger J., Diarra M., Talbot B.G. DNA immunization againstClumping factor A (ClfA) of Staphylococcus aureus. Vaccine 2002;20(17-18) 2348-57

Strong B- and T-cell response After Protein, DNA, and DNAPrime/Protein Boost Immunisation with HBV Cores Carrying HBV PreS1 SequencesD. Skrastina, I. SominskayaProtein Engineering, Biomedical Research and Study Centre, University ofLatvia, Riga, LATVIA.

Hepatitis B virus (HBV) nucleocapsid, or core (HBc) particle,immunologically defined as Hepatitis B core antigen (HBcAg) gave firstonset to an idea of universal, self-assembling, non-infectious carriers forforeign epitopes. The major immunodominant region (MIR) of HBc (aa76-81) is accepted as a target site of choice for insertion of foreignepitopes. We have chosen the preS1 sequences of different length,containing the aa 20-47 region, which are able to elicit HBV-neutralising and protective antibodies, as model epitopes for suchinsertions. We compared induction of specific B- and T-cell responsesafter protein, DNA, and DNA prime/protein boost immunisation intoBalb/c mice using ELISA and T-cell proliferation tests. We found thatdeletions within the MIR of the full- length HBc, and of the C-terminally truncated HBcAg decrease anti-HBc and increase anti-preS1antibody response. The preS1 fragments insertions correspond to the11-60 aa, as well as preS1 11-60 aa plus 89-119 aa into the MIR withaccompanying deletions increase anti-preS1, but decrease anti-HBcantibody response. Three injections of 50 µg of plasmid DNA into MTibialis anterior of Balb/c mice induced a high-titered anti-preS1antibodies as documented 7-13 weeks after immunisation. DNAprime/protein boost immunisation (1) enhanced titer of anti-preS1

antibodies, (2) expanded fraction of the IgG2a anti-preS1, (3) decreasedanti-HBc response, and (4) switched cytokine production to the IFN- _and IL-2 synthesis, which is a sign of induction of the CTL response.

References:1. Song MK, Lee SW, Suh YS, et al. Enhancement of immunoglobulin

G2a and cytotoxic T-lymphocyte responses by a boosterImmunization with recombinant Hepatitis C Virus E2 protein in E2DNA-Primed Mice. J Virol 2000;74:2920-2925

2. Pumpens P, Grens E. HBV core particles as a carrier for Bcell/Tcellepitopes. Intervirology 2001;44:98-114

Genetic Polymorphism and Positive Selection in a ‘Concealed’ GutPotential Vaccine Antigen from Rhipicephalus appendiculatus.L. M. Kamau1, R. Skilton2, T. Musoke2, D. Wasawo2, J. Rowlands2, R. Bishop2

1Kenyatta University & International Livestock Research Institute (ILRI),Nairobi, KENYA, 2International Livestock Research Institute (ILRI), Nairobi,KENYA.

Boophilus microplus Bm86 commercial vaccines control Boophilusdecoloratus and B. annulatus from different parts of the world. Theyshowed potential for control of Hyalomma anatolicum anatolicum andH. dromedarii but insufficient cattle protection against Rhipicephalusappendiculatus, an important livestock tick in sub-saharan Africa. In thisstudy, polymorphism in the Bm86 homolog in R. appendiculatus isolatedfrom four Kenyan field populations and a laboratory stock wascharacterized in view of developing an effective vaccine against this tick.

An estimated 2 Kb cDNA encoding Bm86 homologue inRhipicephalus appendiculatus, Ra86(Muguga) was isolated from a cDNAlibrary constructed using cDNA synthesized from gut RNA extractedfrom Rhipicephalus appendiculatus Muguga Laboratory strain. Nine full-length, including predicted signal peptide and membrane anchor and 10truncated sequences were analyzed. Two allele types were demonstrated;a non-deletion type, potentially encoding 693 amino acids (aa) and adeletion 4-type (654 aa). Five of 19 (26%) sequences were deletion 4-type and the remaining 74% were non-deletion type. The 40 aminoacids deletion near C terminal and 129 (20%) amino acid substitutionsfrom single nucleotide polymorphisms (SNPs) differentiated the twoproteins. Combining Ra86(Muguga) and field isolates, additionaldeletions (39-48 aa), designated 1, 2, 3 and 4, and the non-deletion typeresulted in 5 size types. Non-deletion type was isolated only fromRa86(Muguga) while deletions 1, 2 and 3 were isolated from the field.Deletion 4-type isolated from the field and laboratory would be theappropriate vaccine candidate. The polymorphism was due to positiveselection, shown by selective neutrality and neutral evolution tests, andwas associated with antigenic or another role of the concealed antigen.WINA window analysis revealed mutation hot spots scattered over themolecule.

References:1. de Vos S, Zeinstra L, Taoufik O, Willadsen P, Jongejan F. Evidence for

the utility of the Bm86 antigen from Boophilus microplus invaccination against other tick species. Exp Appl Acarol 2001;25(3):245-261

2. Rand KN, Moore T, Sriskantha A, Spring K, Tellam R, Willadsen P,et al. Cloning and expression of a protective antigen from the cattletick Boophilus microplus. Proc Natl Acad Sci U S A 1989;86(24):9657-9661

P44

P45

Page 83: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

83

(continued)

A Recombinant 63-kilodalton Form of Bacillus anthracisProtective Antigen Produced in the Yeast Saccharomycescerevisiae Provides Protection in two Inhalational ChallengeModels of Anthrax InfectionR. W. Hepler1, R. Kelly1, T. B. McNeely1, H. Fan1, M. C. Losada1, H. A.George1, A. Woods1, L. D. Cope1, A. Bansal1, J. C. Cook1, G. Zang1, S. L. Cohen1, X. Wei1, P. M. Keller1, E. K. Leffel2, J. G. Joyce1, M. L. M. Pitt2, L. D. Schultz1, K. U. Jansen1, M. Kurtz1

1Merck Research Labs, West Point, PA, 2US Army Medical Research Institute ofInfectious Diseases, Fort Detrick, MD.

The sequence of the 63-kDa form of the Bacillus anthracisprotective antigen was codon-optimized for expression in the yeastSaccharomyces cerevisiae. Large-scale fermentation of the antigenproduced intracellularly under these conditions yielded 300 µgrPA63/mL culture medium with rPA63 comprising 12% of the totalyeast protein. The 63-kDa rPA purified under denaturing conditionsdemonstrated biological activity in a macrophage killing assay. Rabbitsand non-human primates (NHP) were immunized with the 63-kDa PAand later challenged with a lethal dose of spores of the virulent Bacillusanthracis Ames strain. Nineteen of twenty-nine rabbits receivingrecombinant yeast rPA63 survived, as did all five animals in a controlgroup immunized with the currently licensed anthrax vaccine. Five ofsix NHP that received the experimental vaccine, and two of three NHPthat received the control vaccine survived. All negative control animalsin both studies died. These results suggest that the 63-kDa form of PAexpressed in yeast is a promising candidate for a safe anthrax vaccineamenable to large-scale production.

Reference:1. Cook, J.C., et. al. Protein Expression and Purification 1998,

13:291-300

A Recombinant Leishmania Antigen Related to the SilentInformation Regulatory 2 (SIR2) Protein Family Induces a B cellActivation and Antibody Specific Immune ResponseR. Silvestre1, A. Cordeiro-da-Silva1, A. Ouaissi2

1Faculdade Farmácia and I.B.M.C. of Universidade Porto, Porto, PORTUGAL,2Institut de Recherche pour le Développement, UR008, Montpellier, FRANCE.

In previous studies, we have reported the characterization of aLeishmania major gene encoding a protein (LmSIR2) with an extensivehomology to yeast SIR2p. This protein, a parasite excreted-secretedantigen is expressed by different Leishmania species. Moreover, theimmunogenicity of LmSIR2 was supported by the high reactivity ofsera from Leishmaniasis patients and dogs against LmSIR2recombinant protein (rLmSIR2). In the present study, we have analysedthe effect of LmSIR2 on T and B murine cell populations. The resultspresented showed that LmSIR2 targeted preferentially the B cells.Indeed, in vitro assays showed that LmSIR2 activates B-cells asevidenced by increased expression of CD69 surface marker.Furthermore, we found that in vivo injection of rLmSIR2 into BALB/cmice induces a significant enhancement in the total number of spleencells mostly due to the increase in the B-cell population whencompared to the control mice, whereas the number of T cells remainedcomparable to the control. Consequently, an antibody response couldbe detected upon in vivo injection of rLmSIR2 with high levels of IgG,mainly of IgG1 and IgG2a subclasses, when compared to IgM.

Interestingly, these IgG antibodies are specific to rLmSIR2, consistingof a mixed IgG1/IgG2a isotype profile and have minimum reactivityagainst other heterologous antigens. Taken together, our data suggestthat LmSIR2, through direct or indirect action towards B cells, inducesa specific immune response.

References:1. Zemzoumi K., Sereno D., François C., Guilvard E., Lemestre J.L.,

Ouaissi A. (1998). Leishmania major: cell type dependentdistribution of a 43 KDa antigen related to silent informationregulator-2 protein family. Biol. Cell 90: 239-245

2. Cordeiro-da-Silva A., Cardoso L., Tomas A., Rodrigues M., CabralM., Vergnes B., Sereno D., Ouaissi A. (2003). Identification ofantibodies to Leishmania silent information regulatory 2 (SIR2)protein homologue during canine natural infections

DNA Vaccine Expressing D8L of Vaccinia Virus Enhanced theEfficacy of Milti-Gene Smallpox DNA Vaccine FormulationsP. V. Sakhatskyy, S. Wang, T. Chou, S. Lu University of Massachusetts Medical School, Worcester, MA.

Background: Most of the world’s population is naïve to poxvirusinfections. Live attenuated smallpox vaccines are under furtherinvestigation. The subunit vaccines (DNA or protein) is effective inanimal studies but their immunogenicity needs further improvementfor human applications (1, 2). Identifying more potent protectiveantigens will strengthen such approach. In this study, vaccinia proteinD8L has been tested for its immunogenicity and protection efficacy insmall animals. Methods: The D8L gene insert was produced fromVACV (WR strain) genome and subcloned into the DNA vaccinevector pSW3891. New Zealand White rabbits or BALB/c mice receivedfour bi-weekly DNA immunizations with a gene gun. Immunized micewere challenged ip with lethal dosage of VACV. Results: D8L DNAvaccines were highly immunogenic and elicited strong anti-D8L IgGresponses in immunized rabbits and mice shown by ELISA, WesternBlot and IMV neutralization assays. D8L vaccine protected miceagainst VACV challenge, either alone or in different multi-genesmallpox DNA vaccine formulations. Superior efficacy of a formulationcontaining the D8L was demonstrated. Vaccinia immunoglobulin andsera from mice that survived VACV challenge contained anti-D8Lantibody, supporting its role in vaccine induced immune protection.D8L DNA vaccine could induce antigen specific T cell responses withpositive IFN-gamma by ELISPOT. Conclusions: D8L is a strongimmunogen capable of eliciting protective antibody and cell mediatedimmunity in small animals. It may serve as an excellent candidate toimprove the existing subunit based smallpox vaccines.

References:1. Hooper JW, Thompson E, Wilhelmsen C, et al. Smallpox DNA

vaccine protects nonhuman primates against lethal monkeypox. JVirol 2004;78(9):4433-43.

2. Fogg C, Lustig S, Whitbeck JC, Eisenberg RJ, Cohen GH, Moss B.Protective immunity to vaccinia virus induced by vaccination withmultiple recombinant outer membrane proteins of intracellular andextracellular virions. J Virol 2004;78(19):10230-7.

P46

P47

P48

Page 84: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

84

Eighth Annual ConferenceAn Intranasal, Protollin™-RSV Subunit Vaccine Induces MucosalIgA, Serum Neutralizing Antibodies and a Type-1 Cytokine-biasedResponse by Spleen and Lung Mononuclear Cells in Mice.S. L. Cyr1, T. Jones2, I. Stoica-Popescu2, S. Chabot1, D. S. Burt2, B. J. Ward1

1Microbiology and Immunology, McGill University, Montreal, PQ, CANADA, 2ID Biomedical, Laval, PQ, CANADA.

There is currently no vaccine for respiratory syncytial virus (RSV), amajor respiratory pathogen in children. A formalin-inactivated RSVvaccine (FI-RSV) exacerbated disease in children who later became RSVinfected. This aberrant response was characterized by eosinophilia and atype-2 immune phenotype. Intranasal administration (IN) ofProtollin™, a Proteosome™-based adjuvant, was shown to favor type-1immunity in mice and to be safe in humans. Antigen enriched in RSV F(fusion) and G (attachment) proteins was prepared from RSV-infectedVero cultures by chromatography, formulated with Protollin and used toimmunize Balb/c mice (IN, 3 doses). Serum, bronchoalveolar lavage(BAL), lungs and spleens were collected at euthanasia. Mean RSV-specific serum IgG titers measured by ELISA were significantly higher inmice immunized with the Protollin-formulated antigen compared withthose given antigen alone (P ≤ 0.001). Immunization with Protollin-formulated antigen also elicited serum neutralization titers in excess oflog2 7.75 as well as significant levels of RSV-specific IgA in BAL (P ≤0.001 compared with mice given antigen alone). Spleen and lungmononuclear cells from mice immunized with Protollin-formulatedantigen spontaneously released up to 45 times more IFN_ into culturesupernatants than groups immunized with antigen alone, concomitantwith greatly reduced IL-5 production. These results suggest that anintranasally delivered Protollin-formulated RSV vaccine may induceprotective immunity without the potentially adverse type-2 cytokine-biased responses.

References:1. Jones T, Cyr S, Allard F, Bellerose N, Lowell GH, Burt DS.

Protollin™: a novel adjuvant for intranasal vaccines. Vaccine 2004;22:3691-3697.

2. Fries LF, Montemarano AD, Mallett CP, Taylor DN, Hale TL, LowellGH. Safety and Immunogenicity of a Proteosome-Shigella flexneri 2alipopolysaccharide vaccine administered intranasally to healthyadults. Infect. Immun. 2001;69:4545-4553.

Prevention of Serogroup A, C and W135 Meningococcal Disease in the Meningitis Belt of Africa by Targeting Outer MembraneProteinsE. Rosenqvist, G. Norheim, E. Fritzsønn, T. Tangen, P. Kristiansen, D. A. Caugant, A. Aase, E. A. Høiby, I. S. Aaberge Division for Infectious Disease Control, Norwegian Institute of Public Health,Oslo, NORWAY.

Epidemic meningococcal disease is recurrent in the Meningitis Beltand is mainly caused by very homogenous clones of serogroup Asubgroup III (A:4/21:P1.20,9) or serogroup W135 and C (sequencetype ST-11; 2a:P1.5,2). Although current vaccine approaches for theseserogroups target capsular antigens, non-capsular antigens such as outermembrane proteins and lipopolysaccharides are also potential vaccinecandidates.

Outer membrane vesicle (OMV) vaccines were developed fromserogroup A and W135 strains with the same technique as for theNorwegian serogroup B OMV vaccine (MenBvac®) and used for

immunizing mice, either alone or in combination. The immunogenicity(ELISA), antibody specificity (immunoblot) and the bactericidal andopsonophagocytic activities of the antibodies generated by these vaccineswere investigated.

The serogroup A OMV vaccine induced high levels of anti-OMVIgG in mice, high bactericidal titres towards serogroup A strains of bothST-5 and ST-7 from different African countries, and highopsonophagocytic activity. Serogroup W135 OMVs were also highlyimmunogenic, and sera showed high bactericidal titres towards bothserogroup W135 and C strains belonging to ST-11.

We therefore suggest that immunization with a serogroup A+W135OMV mixture vaccine, either alone or combined with polysaccharideantigens can protect against meningococcal disease caused by mostserogroup A, C or W135 strains in Africa and may be an affordable T-cell dependent alternative to conjugate vaccines.

References:1. Norheim G, Høiby EA, Caugant DA, et al. Immunogenicity and

bactericidal activity in mice of an outer membrane protein vesiclevaccine against Neisseria meningitidis serogroup A disease. Vaccine2004; 22: 2171-80.

2. Tappero JW, Lagos R, Ballesteros AM, et al. Immunogenicity of 2serogroup B outer-membrane protein meningococcal vaccines: arandomized controlled trial in Chile. JAMA 1999;281:1520-7.

Development and Use of the Outer Membrane Vesicle Concept forVaccines Against Meningococcal Group B Disease E. Rosenqvist1, G. Norheim1, L. Meyer Næss1, P. Kristiansen1, P. Costantino2, E. Wedege1, D. A. Caugant1, B. Feiring1, I. S. Aaberge1, R. Rappuoli3, J. Holst1

1Division for Infectious Disease Control, Norwegian Insitute of Public Health,Oslo, NORWAY, 2Chiron Vaccines S.r.l., Siena, ITALY, 3Chiron Vaccine S.r.l., Siena,ITALY.

Protein-based, outer membrane vesicle (OMV) vaccines have provento be efficacious against serogroup B meningococcal disease in Norwayand Cuba. Whereas such vaccines induce high levels of bactericidalantibodies against homologous strains in all age groups, they are mainlystrain-specific in infants. Currently, a public health intervention isongoing in order to control a serogroup B epidemic in New Zealand,based on a B:4:P1.7b,4 production strain (MeNZB®). The upscalingand standardization of the required vaccine production for controllingthe New Zealand epidemic has allowed the establishment of large-scaleGMP manufacturing for OMV vaccines . The outcome of this will bethe licensing of the vaccine in New Zealand and possibly in othercountries.

Pre-clinical and clinical data have been evaluated and compared forMenBvac® and MeNZB®, originally designed to control outbreaks inNorway and New Zealand, respectively. Data from these twoformulations and other experimental wild-type OMV preparationsprovide an important basis for establishing international guidelines andregulatory consensus for such vaccines in the time to come. Theavailability of licensed OMV vaccines raises the question of whether thistype of vaccines may provide the opportunity to control localizedoutbreaks of serogroup B meningococci in other areas.References:

1. Tappero JW, Lagos R, Ballesteros AM, et al. Immunogenicity of 2serogroup B outer-membrane protein meningococcal vaccines: arandomized controlled trial in Chile. JAMA 1999;281:1520-7.

P49

P50

P51

Page 85: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

85

(continued)

2. Holst J, Feiring B, Meyer Næss L et al. The concept of “tailor-made”,protein based outer membrane vesicle vaccines against meningococcaldisease. Vaccine 2005, in press.

Reducing the Cost of Manufacturing Conjugate Vaccines; Effective Alternatives to Gel FiltrationA. Lees1, D. E. Shafer2

1Biosynexus Inc, Gaithersburg, MD, 2None, Gaithersburg, MD.

Vaccines consisting of capsular polysaccharides covalently linked toproteins have been extremely effective at inducing humoral immunity.However, this class of vaccines is among the most expensive of thechildhood immunizations. Gel filtration is typically used to purify thehigh MW conjugate from free protein and lower MW polysaccharide, astep which is slow and expensive, and which reduces yield. The use ofmild, efficient linking chemistries (Lees et al., Vaccine 14:190, 1998)which minimize polysaccharide hydrolysis, and the fact that the sameprotein used as the carrier is often added back as part of the final multi-vaccine formulation, allowed us to evaluate alternatives to the gelfiltration step. In one approach, conjugate vaccines were cleaned-upusing dialysis only. Since the total amount of free protein maynevertheless be significant in multivalent vaccines such as S.pneumoniae, it may be necessary to reduce the levels of unconjugatedprotein. To address this, we developed a solid-phase method that rapidlyabsorbs free protein but not the high MW conjugate (Shafer et al.Vaccine 19:1547, 2001). Conjugate vaccines of S. pnuemoniae type 14and N. meningiditis A and C capsular polysaccharides were processed bythe usual gel filtration techniques, by simple dialysis or by the solidphase adsorption method. Yields of conjugate vaccine, based onpolysaccharide recovery, were >90% for the latter two approaches buttypically <75% after gel filtration. Comparable anti-polysaccharideantibody titers were induced in mice, regardless of the process method.

Aminooxy Reagents and Oxime Chemistry for the Preparation of Conjugate Vaccines A. Lees, A. LopezAcostaBiosynexus Inc, Gaithersburg, MD.

Covalent linkage of proteins to polysaccharides significantlyenhances the antibody response to the carbohydrate. We present aversatile and efficient conjugation chemistry based on aminooxy (AO)reagents and oxime chemistry. Proteins and polysaccharidesfunctionalized with AO groups and aldehydes, respectively, werecovalently linked via oxime formation. To increase versatility, homo andheterobifunctional aminooxy reagents, e.g., bis(AOAc)ethylenediamineand N(AOAc)cysteamine, were prepared. Bis(AOAc) (ethylenediamine)was used to functionalize protein carboxyls with AO groups.Alternatively, proteins were derivatized using a two-step protocol. In thisapproach, a limited number of amines were bromoacetylated and thenreacted with (AOAc)cysteamine via a thioether linkage. These AO-derivatized-proteins were subsequently reacted with oxidizedpolysaccharides. The reverse linkage model was also employed, in whichAO-derivatized polysaccharides were linked to aldehyde-derivatizedproteins. Glycoproteins, like ovalbumin, could be oxidized under mildconditions. Aldehydes were created on nonglycosylated proteins bycarbodiimide-mediated reaction with glyceric acid, followed by mildoxidation. Alternatively, amines were modified by bromoacetylation and

reaction with mercaptoglycerol, followed by mild oxidation of the diol.These aldehyde-modified proteins reacted with AO-derivatizedpolysaccharides to form conjugates via oxime linkages. Sera from miceimmunized with a pneumococcal type 14 conjugate vaccine made usingoxime chemistry had high anti-polysaccharide immune responses. Theprimary response could be boosted, indicating that the polysaccharideconjugate had characteristics of a T cell dependent antigen.

Development of HIV-1 Subtype C Vaccine CandidatesP. Seth1, A. Arora1, P. Chugh1, S. Kumar1, P. Aggarwal1, V. Prasad1, M. Vajpayee1, David Montefiori2

1Microbiology, All India Institute of Medical Sciences, New Delhi, INDIA,2Department of Surgery, Duke University Medical Center, Durham, NC, USA.

Since in India HIV-1 subtype C accounts for more than 95% ofHIV-1 infections, it is imperative that a vaccine based on the localcirculating subtype should be designed. We cloned two envelope(gp120) gene and one gag protease gene of Indian HIV-1 subtype Cvirus in the mammalian expression plasmid DNA vector ,pNK14, andMVA (Modified Vaccinia Ankara) and evaluated these forimmunogenicity in mice and in bonnet monkeys in ‘prime-booststrategy format’. in which priming was done with rDNA vaccineconstructs and boosting was done with rMVA constructs four weeksafter priming. Immunized animals developed strong clade specific andcross clade reactive humoral as well as cell mediated immune responses.Humoral immune response was evaluated by ELISA and neutralizingantibody assay. The prime-boost regimen induced cross reactingneutralizing antibodies in monkeys against HIV-1 subtype C isolatesfrom early sero-converts from South Africa. Cellular immune responsewas assayed by Cytotoxicity assay, ELISPOT assay andLymphoproliferative assay. Interestingly, the memory of the immuneresponse was detected even beyond 6 months in mice and beyond 73months in monkeys. Immunized monkeys showed a very strong HIV-1subtype C specific recall immune response when challenged with MVAconstructs 73 weeks after vaccination with two doses of DNA vaccine.

References:1. Chugh P and Seth P. Induction of broad based immune response

against HIV-1 subtype c gag DNA vaccine in mice. ViralImmunology, 17, 2004 423-435.

2. Kumar S and Seth P. Immunogenicity of Recombinant ModifiedVaccinia Ankara Viruses (rMVA) Expressing HIV-1 Indian Subtype Cgag-protease and env-gp120 genes in Mice. Viral Immunol. 17, 2004,574-579.

Enhancement of Cell-mediated Immunity in Mice by the Model of a Whole HIV-1 gag in Live Mycobacterium bovis BCGD. PromkhatkaewDepartment Of Medical Sciences, Nonthaburi, THAILAND.

Background: A recombinant live vector vaccine approach wasfocused to elicit a vaccine candidate of recombinant Mycobacterium bovisBCG harboring a whole HIV-1 CRF01_AE gag DNA to investigatespecific cell-mediated immunities in BALB/c mice. Methods: The wholeHIV-1 CRF01_AE gag sequence of 1,497 basepairs from Thai isolatewas inserted into BCG. This live BCG was injected subcutaneously into

P52

P53

P54

P55

Page 86: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

86

Eighth Annual ConferenceBALB/c mice once, and the spleen cells were investigated gag-specificcytotoxic T-lymphocyte (CTL) activity by Cr-55 release assay andlymphocyte proliferation. Results: Construction of the stable expressionrecombinant BCG was achieved that a cytoplasmic target protein ofapproximately 55 Kd was detected by Western-blot. By single injectionof 0.1 mg of the recombinant BCG into mice, various specific CTLresponses were exhibited either against a single gag epitope of aminoacids 294-304 after 2 weeks, or widely recognition of various peptideregions along the entire gag protein of moderate CTL activities (10-35%specific cell lysis) and relatively high (50-68%), after one month.However, after two months, despite the remaining of CTL responses,the activities were obviously 3-3.7 folds lower. These may trigger us totry for a boost immunization with other vaccine vehicles containing thesame immunogen content to revert the CTL response in further study.On the other hand, lymphocyte proliferation was also detected as 9.3folds higher than that of non-immunized mice. Conclusion: Our resultsdemonstrate that BCG may be used as a live vector to induce cellularimmunity to HIV-1 gag as introducing the vaccination in mice.

References:1. Honda M, Matsuo K, Nakasone T, et al. Protective immune responses

induced by secretion of a chimeric soluble protein from arecombinant Mycobacterium bovis BCG vector candidate vaccine forHIV-1 in small animals. Proc Natl Acad Sci USA 1995;92:1069

2. Ishii K, Ueda Y, Matsuo K, et al. Structural analysis of vaccinia virusDIs strain: Application as a new replication-deficient viral vector.Virol 2002;302:433-44.

Preclinical Development of Yeast-Based Immunotherapy forChronic Hepatitis C Virus InfectionA. A. Haller, T. King, Y. Lu, C. Kemmler, D. Bellgrau, G. Gordon, D. Apelian, A. Franzusoff, T. C. Rodell, R. C. DukeGlobeImmune Inc., Aurora, CO.

Background: Evidence suggests that control of hepatitis C infectionin humans requires effective T cell-mediated immunity. Previous studieshave demonstrated that recombinant, heat-inactivated Saccharomycescerevisiae yeast (TarmogensTM) are avidly phagocytosed by and directlyactivate dendritic cells which present yeast-associated proteins to CD4and CD8 T cells that are capable of mediating antigen-specificprotective and therapeutic anti-tumor immunity. In this study,Tarmogens that produce an HCV NS3-Core fusion protein (GI-5005)were evaluated for their ability to induce protective and therapeuticimmunity in mice. Methods: C57BL/6 and BALB/c mice were injectedsubcutaneously with GI-5005 and immunogenicity was determinedusing assays that measure antigen-specific lymphocyte proliferation,cytokine secretion, and cytotoxicity. A surrogate mouse model of HCVinfection employing HCV antigen-expressing syngeneic tumor cells wasused to assess preventative and therapeutic efficacy.Results: Immunization with GI-5005 induced dose-dependent NS3 andCore antigen-specific helper and cytotoxic T cell activities that wereassociated with secretion of IL-2, IFN-g, GM-CSF and TNF-a. Micethat were immunized either prior to or seven days after challenge withNS3-expressing tumor cells were protected against tumor formation.No significant adverse effects have been observed upon repeatedadministration of Tarmogens in mice, rats, rabbit and macaques.Conclusion: The GI-5005 Tarmogen was found to elicit protectiveHCV-specific helper and cytotoxic T cell responses in mice. A Phase 1study is being initiated to evaluate GI-5005 in chronically HCV-

infected humans.

References:1. Stubbs AC, Martin KS, Coeshott C, Skaates SV, Kuritzkes DR,

Bellgrau D, Franzusoff A, Duke RC, Wilson CC. Whole recombinantyeast vaccine activates dendritic cells and elicits protective cell-mediated immunity. Nature Med. 2001; 5:625-629.

2. Lu Y, Bellgrau D, Dwyer-Nield LD, Malkinson AM, Duke RC,Rodell TC, Franzusoff A. Mutation-selective tumor remission withRas-targeted, whole yeast-based immunotherapy. Cancer Res. 2004;64:5084-5088.

Analytical Challenges for Novel Vaccine FormulationsE. Hartwell, S. D. Sen, B. J. Roser Cambridge Biostability Ltd, Cambridge, UNITED KINGDOM.

Thermostable vaccines have been developed using mixed sugar glassmicrospheres suspended in fluorinated liquids. The microspheres areinsoluble in, and density matched with the liquid, producing physicaland chemically stable formulations. Analysis of polysaccharide vaccinessuch as conjugate Haemophilus influenzae type b vaccine in suchformulations poses analytical challenges. The high sugar content is inexcess of the minute saccharide content under analysis. One method,used by the vaccine manufacturer, involved acid hydrolysis ofpolysaccharide to generate Ribitol and Ribose and the use of HighPerformance Anion Exchange Chromatography with PulsedAmperometric Detection (HPAEC-PAD) to detect the non ionic sugar,Ribitol. This appeared as a minute shoulder of the large peak of thestabilising sugars. We have now developed a sample preparation andchromatographic method which has eliminated the interference of thestabilising sugars so that the monomeric saccharide unit can bequantified.

Commercially available Haemophilus influenzae type b conjugatevaccine with aluminium phosphate, was used. The vaccine wasformulated and spray dried to form 3 component mixed glassmicrospheres. These were suspended in an inert fluorinated liquid. Thechemical stability of the vaccine is determined as a measure of theamount of free saccharide. It is expressed as percentage of the totalsaccharide content. The method we have developed uses a differentsample preparation which results in an ionic form of free saccharide, 5-D-Ribitol-(1-1)-beta-D-Ribose-3-phosphate. An HPAEC-PAD methodcompletely separates the ionic form from neutral stabilising sugars. Thismethod makes it possible to monitor stability of our novel formulationsstressed up to 70ºC.

Production and Control of a Brazilian Meningococcal C ConjugateVaccineSilveira IAFB1, Bastos RC1, Neto MS1, Larangeira AP1, Fernandes SAR1, Leal ML1, Silva WC1, Lee C-H2, Frasch C2 & Jessouroun E1

1Laboratório de Tecnologia Bacteriana, Bio-Manguinhos, FIOCRUZ, RJ, Brazil2Center for Biologics Evaluation and Research, FDA, Bethesda, USA

Meningococcal vaccines based on capsular polysaccharides induceshort term protection in children and have been replaced by saccharide-protein conjugates. In Brazil, a conjugate vaccine has been developedagainst the serogroup C by reductive amination (Jennings HJ &Lugowski C, Immunochemistry of groups A, B, and C meningococcal

P56

P57

P58

Page 87: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

87

(continued)

polysaccharide-tetanus toxoid conjugates. J Immunol 1981; 127:1011-1018). Different vaccine lots were obtained with the C polysaccharideand tetanus toxoid. The aldehyde groups present in the activated-polysaccharides by sodium periodate were measured by the purpaldmethod. The conjugation step was followed by SEC and 1HNMRspectroscopy. The volumes were increased from 8 to 2,000 mL of finalproduct in scale-up assays showing reproducibility of the productionprocedures. One lot corresponding to 27,000 human doses(10_g/0.5mL) was used to prepare vials containing 5 lyophilizedhuman doses in sterile conditions. All quality control assays requiredwere satisfactory (WHO 2001. Recommendations for the production& control of meningococcal group C conjugate vaccines). In dose-response mice studies 1_g dose showed similar bactericidal activity and IgG titers to 10_g, but higher than the control group (plainpolysaccharide; ANOVA, p < 0.05). These results support productionof clinical lots to be tested in phase I clinical studies this year.

References:1. Jennings HJ & Lugowski C, Immunochemistry of groups A, B, and

C meningococcal polysaccharide-tetanus toxoid conjugates. JImmunol 1981; 127:1011-1018

2. WHO 2001. Recommendations for the production & control ofmeningococcal group C conjugate vaccines

Production and Control of a Brazilian Meningococcal B VaccineJessouroun E1, Larangeira AP1, Pereira S1, Fernandes SA1, Nascimento DR1, Leal ML1, Gorla MC 2, Brandão A2, Simonsen V2, Schenkmann RFP 3, TanizakiMM3, Frasch CE 4, Silveira IAFB1 1Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil2Instituto Adolfo Lutz, SP, Brazil; 3Instituto Butantan, SP, Brazil; 4Center forBiologics Evaluation and Research, FDA, Bethesda, USA.

In Brazil serogroup B accounts for 60% of total clinical isolates frommeningococcal infections (Jessouroun E et al., Vaccine 2004; 22:2617-2625). Brazilian research institutions have collaborated to develop agroup B meningococcal vaccine composed of detergent treated-OMVand dLOS. The antigens were purified from bacterial biomass harvestedfrom 100L bioreactor batches. In antigen presentation stability studies,lyophilized vaccine seems to be more stable than liquid.Immunogenicity results from studies of the vaccine in mice werepromising, showing less reactogenicity and pirogenicity than a Cubanmeningococcal B vaccine (Vamengoc®). The inflammatory potential ofboth vaccines was studied in rabbits and showed leucocytosis after eachintramuscular injections (three doses). The increase of total leukocytecount, induced by Brazilian vaccine was short lasting with reduction ofmeasured levels lasting up to 7 days while Cuban vaccine tended tomaintain somewhat elevated leukocyte levels 7 and 30 days afterimmunization. The quality control methodologies were also revised andthe product presented satisfactory results compared to similar vaccines(Fredrikssen JH et al., NIPH Annals 1991; 14:67-78). These resultssupport production of clinical lots to be tested in phase I studies thisyear.

Needle-Free Delivery of Antigens to Ultrasound Pre-treated SkinDemonstrated in a Feasibility Clinical TrialD. H. Libraty1, S. P. Barman2

1Center for Infectious Diseases & Vaccine Research, University ofMassachusetts Medical School, Worcester, MA, 2Transdermal Drug Delivery ,Vaccines, Sontra Medical, Franklin, MA.

Introduction. Current immunizations involve needle-admininistered vaccines by IM/SC/ID routes. Skin immunization canelicit robust immunity, due to the network of Langerhans cells in theepidermis.We present a method to permeate the skin using an ultrasound device(SonoPrep®), pre-topical delivery of vaccines. SonoPrep appliesultrasound to a liquid that creates channels through the stratumcorneum. We report data from a clinical trial that determined delayed-type hypersensitivity to antigens placed on skin permeated withSonoPrep. Methods. The study was approved by the UMASS IRB. 20volunteers, in two groups were enrolled in the study: (a) standard dosesof tetanus toxoid and Candida albicans antigens (ID), (b) identical dosesof the same recall antigens over ultrasound-permeated sites. The antigenwas injected intradermally, into two sites (10 subjects). Results. Thedevice was activated with a microprocessor performing conductivityanalysis to determine permeability, triggering device-turn off.<br /Theantigen was placed in the target reservoir. Indurations were measured at48 h/96 h/7 days after application. Among twenty sites with SonoPrep,one individual experienced discomfort at one site, other treated siteswere sonicated with no adverse events. Tetanus toxoid and Candidaalbicans recall antigens delivered by Sonoprep produced DTH responsesin 9/10 and 10/10 subjects. The kinetics of DTH responses were similarbetween the intradermal/Sonoprep groups. Conclusions. In conlusion,immune responses were elicited to antigens delivered to SonoPrep-treated skin.

References:1. Glenn GM et.al. Transcutaneous immunization and

immunostimulant strategies: capitalizing of the immunocompetenceof the skin. Expert Rev Vaccines 2:253-67, 2003

2. Samir Mitragotri, Joseph Kost, 2004. “Low Frequency Sonophoresis:A Review”, Advanced Drug Delivery Reviews, 56, 589-601

Anaphylaxis Following Recombinant Hepatitis B Vaccines in Yeast-Sensitive Individuals: Reports to VAERSL. DiMiceli1, V. Pool1, S. V. Shadomy2, J. Iskander1

1CDC/National Immunization Program, Atlanta, GA, 2CDC/National Center forInfectious Diseases, Atlanta, GA.

Background: Theoretical concerns exist over recombinant HepatitisB vaccination (HBV) of yeast-sensitive individuals (YSI). Per theAdvisory Committee on Immunization Practices, the presence ofallergies to baker’s yeast is listed as a contraindication for HBV. Weevaluated reports to the Vaccine Adverse Event Reporting System(VAERS) for anaphylaxis following HBV in YSI. Methods: We searchedthe entire VAERS database for reports received from July 1990 to July 1,2004 mentioning a history of allergies to yeast. We classified cases whichdescribed a history of yeast allergies as probable or possible anaphylaxisbased on previously published criteria. Probable anaphylaxis was definedas >1 dermatologic symptom(s) and >1 respiratory, gastrointestinal, orcardiovascular symptom(s) with onset <4 hours of vaccination. Possible

P59

P60

P61

Page 88: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

88

Eighth Annual Conferenceanaphylaxis was defined as >1 dermatologic or respiratory symptom(s)with onset <4 hours of vaccination, or as >1 dermatologic andrespiratory symptoms within 4-12 hours of vaccination. Results: Weidentified 107 reports which mentioned yeast allergies. Out of 11potential anaphylaxis cases identified after HBV or in combination withHBV, four met the probable case definition. The age range was 10-50years, three were classified as serious per FDA regulations, and themajority was female. No deaths were reported. Conclusions: Althoughthe number of vaccinated YSI is unknown, the number of definitereported life-threatening anaphylactic reactions in YSI following HBV isfew. Further studies investigating the possible role of yeast in thepathophysiology of vaccine-associated anaphylaxis should beundertaken.

References:1. Greenburg DP. Pediatric Experience with Recombinant Hepatitis B

Vaccines and Relevant Safety and Immunogenecity Studies. PediatrInfect Dis J 1993; 12:438-45.

2. Wiedermann G, Scheiner O, Ambrosch F, et al. Lack of Induction ofIgE and IgG Antibodies to Yeast in Humans Immunized withRecombinant Hepatitis B Vaccines. Int Archs Allergy Appl Immun1988; 85:130-132.

Histopathology Analysis of Local Reactions in Mice followingInjection of Diphtheria-Tetanus-Acellular Pertussis (DTaP) vaccines A. Honjo, T. Katsuta, S. Tateyama, C. Nagaoka, T. Tokutake, Y. Arimoto, N. Nakajima, T. Goshima, T. KatoSt.Mariannna University School of Medicine, Kawasaki-si, JAPAN.

Background: In Japan, diphtheria tetanus acellular pertussis (DTaP)vaccines have been used . The rate of severe adverse events has beenreduced. However, redness and/or swelling reactions are still presentedand the mechanisms causing these reactions have not been elucidated.Therefore, we injected separately and observed histopathology changesat the injection site of: diphtheria tetanus acellular pertussis (DPT),diphtheria tetanus toxoids (DT), tetanus toxoids (T), thimerosal,aluminum chloride, and formalin.Methods: Eight-week-old male ICR strain SPF mice were used. Micewere injected subcutaneously with 0.5 ml of each vaccine or with anadditive at their shaved abdomen. Mice were injected twice at 4-weekintervals. One week after the second injection, all the mice weresacrificed and their tissues were surgically removed from the injectionsites. Results: In macroscopic findings, we found major axis indurationsof approximately 5 mm in diameter in each of the DPT, DT, and Tgroups. There were no macroscopic changes in the three groupsreceiving additives. In histopathology examinations, we detected thecellular infiltration of neutrophils suggesting an inflammation, andeosinophils or histiocytes suggesting an allergy. The DPT grouppresented the most cellular infiltration. Conclusions: These resultssuggest that local reactions depend on various factors rather than asingle one, and have relevance to inflammation and allergy.

Reference:1. Norihisa Goto and Kiyoto Akama. Histopathological Studies of

Reactions in Mice Injected with Aluminum-Adsorbed TetanusToxoid. Microbiol.Immunol. 1982; 26: 1121-1132._______

CAP Absorbed rPA Nasal Vaccine as an Approach to MucosalImmune Protection Against Anthrax InfectionP. Nagappan1, J. Arroyo2, T. Morcol1, A. R. Mitchell1, L. Nerenbaum1, S. Billingsley1, S. J. D. Bell11BioSante Pharmaceuticals, Inc., Smyrna, GA, 2DVC LLC a CSC Company,Frederick, MD.

The spore-forming bacterium Bacillus anthracis is considered to be alikely biological warfare agent1. Anthrax spores can easily be transmittedvia the air, as evidenced by the post 9/11 terrorist attacks in the USA.The current anthrax vaccine licensed for use in the United States isAnthrax Vaccine Absorbed, an aluminum hydroxide-adjuvanatedvaccine administered subcutaneously2. Our goal is to avoid the use ofaluminum adjuvant and to develop alternatives to injectable vaccines.The BioSante proprietary calcium phosphate (CAP) nanoparticle systemis a proven non-injectable adjuvant. In the present study, recombinantProtective Antigen (rPA) adjuvanated with CAP was delivered across themucosal surfaces of the upper airways. As a reference standard, the rPAwas combined with Alhydrogel (aluminum hydroxide gel). Both CAPand Alhydrogel formulations were administered intranasally (IN) toBalb/c mice using one primary and two booster IN vaccinations.Immunogenicity was monitered over 14 weeks. No mortality ormorbidity was noted during this study. Serum was collected from bi-weekly blood samples and analyzed by ELISA. After nasal delivery, theanti-rPA IgG ELISA results indicated that the CAP-based vaccineinduced a stronger, faster and longer-lasting immune response thanAlhydrogel-adjuvanated rPA or rPA alone. Therefore, our CAP-rPAanthrax vaccine candidate appears to be a safe and potent inducer ofhumoral immunity after IN delivery.

References:1. Klemm DM, Klemm WR. A history of anthrax. J Am Vet Med Res

1959; 135: 4582. Pittman PR, Kim-Ahn G, Pifat DY, et al. Anthrax vaccine:

immunogenicity and safety of a dose-reduction, route-changecomparison study in humans. Vaccine 2002; 20: 1412-20

Influenza Nucleoprotein Conjugated to Immunostimulatory DNA as a Potential Vaccine Against Pandemic InfluenzaT. dela Cruz1, D. Higgins1, G. Ott1, I. Mbawuike2, S. Tuck1, G. Van Nest1

1Dynavax Technologies, Berkeley, CA, 2Baylor College of Medicine, Houston, TX.

Background: A highly immunogenic, conserved influenza antigencould serve as a first-line vaccine defense against divergent or pandemicstrains. Immunization of mice with influenza nucleoprotein (NP)provides protection against divergent influenza stains. Linkage ofimmunostimulatory DNA (ISS) to antigens significantly enhancesantigen-specific cellular and humoral responses. NP linked to ISS (NP-ISS) is expected to induce strong Th1 and CTL responses that canprovide cross-protection against widely divergent A strains. NP-ISS mayalso enhance responses to other co-delivered viral antigens such ashemagglutinin (HA). Methods: Mice were immunized twice with NP-ISS ± split monovalent flu vaccine and bled two weeks after eachimmunization for anti-NP and anti-HA isotype analysis. Spleens wereharvested to evaluate NP-specific and HA-specific cell-mediatedimmune responses via ELISA, ELISPOT, and CTL. Statisticalsignificance calculated using one-way ANOVA with the Kruskal-Wallisand Dunn’s Multiple Comparison tests. Results: Immunization withNP-ISS induced high antigen-specific IgG2a (p<0.01) and IFN_

P62

P63

P64

Page 89: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

89

(continued)

responses (p<0.01), while immunization with NP alone inducedpredominantly IgG1 and IL-5. IFN_ ELISPOT data showed significantNP-specific CD8+ IFN_ response (p<0.001), and Cr-51 release assaysshowed significant cytotoxic activity was induced in the conjugateimmunized mice (p<0.001). Co-delivery of NP-ISS and split vaccinesignificantly enhanced overall HA antibody titers (p<0.01).Conclusions: Immunization with NP-ISS induces potent Th1 and CTLresponses, as well as enhancing response to HA when co-delivered withsplit vaccine. This vaccine shows great potential to induce protectiveimmunity against divergent and potentially pandemic influenza strains.

References:1. Mbawuike I, et al. A baculovirus–expressed influenza A/Udorn

(H3N2) nucleoprotein induces protective T cell immunity againstinfluenza A/H3N2 and A/H1N1 in mice. Vaccine Research 1994;3:211-227.

2. Tighe H, et al. Conjugation of a protein to immunostimulatory DNAresults in rapid, long-lasting and potent induction of cell-mediatedand humoral immunity. Eur J Immunol 2000; 30(7):1939-47.

Development of SARS Vaccine Using Recombinant Vaccinia VirusDerived from LC16m8M. Kitabatake1, F. Yasui2, S. Inoue3, K. Morita3, F. Murai4, M. Kidokoro5, K. Mizuno6, H. Shida7, K. Matsushima1, M. Kohara2

1Univ. Tokyo, Tokyo, JAPAN, 2The Tokyo Metro. Inst. Med. Sci., Tokyo, JAPAN,3Inst. Tropical Med., Nagasaki Univ., Nagasaki, JAPAN, 4Post Genome Inst.,Tokyo, JAPAN, 5Natl. Inst. Infect. Dis., Tokyo, JAPAN, 6The Chemo-Sero-Therapeutic Res. Inst., Kumamoto, JAPAN, 7Inst. Gen. Med., Hokkaido Univ.,Sapporo, JAPAN.

Background. Severe acute respiratory syndrome (SARS) caused bynovel type coronavirus (SARS-CoV) first emerged in China and spreadto 29 countries, resulting in over 800 people death. Methods. Wegenerated SARS-CoV spike protein expressing recombinant vacciniavirus (RVV) derived from its highly attenuated strain LC16m8. Weperformed the in vitro neutralizing assay against SARS-CoV or vacciniavirus (VV) using serum from rabbits intradermally immunized with RVVor LC16m8. Also we confirmed the efficacy of RVV against the rabbitswhich were pre-immunized with LC16m8. Results. RVV, but notLC16m8, induced the neutralizing (NT) antibody against SARS-CoV inrabbits from one week after injection, and the NT titer increased about100 times. At two weeks after boost injection, the NT titer increased by10 times further. In the rabbits pre-immunized with LC16m8, the NTantibody against SARS-CoV could be similarly induced by inoculation ofRVV in spite of the presence of the NT antibody against VV.Conclusions. RVV highly induces the NT antibody against SARS-CoVby single injection. RVV provides the protective immunity against SARS-CoV by overcoming the immunity against VV. RVV derived fromLC16m8 can be powerful vaccine against SARS even for the people whowere previously inoculated with smallpox vaccine.

References:1. Jin NY, Funahashi S, Shida H. Constructions of vaccinia virus A-type

inclusion body protein, tandemly repeated mutant 7.5 kDa protein,and hemagglutinin gene promoters support high levels of expression.Arch Virol. 1994;138:315-30.

2. Ohishi K, Inui K, Barrett T, Yamanouchi K. Long-term protectiveimmunity to rinderpest in cattle following a single vaccination with arecombinant vaccinia virus expressing the virus haemagglutininprotein. J Gen Virol. 2000 81:1439-46.

A Novel Subunit Vaccine Protects Mice Against Yersinia InfectionL. Wonderling1, D. Straub2, D. Emery2

1Syntiron, Saint Paul, MN, 2Epitopix, Willmar, MN.

The genus Yersinia consists of three human pathogens; Y. pestis is thecausative agent of plaque, while Y. pseudotuberculosis and Y. enterocoliticaprimarily cause gastrointestinal illnesses. Due to concerns that Y. pestismay be used as a potentially deadly bio-weapon, there has beenconsiderable research aimed at identifying new antigens for a plaguevaccine. Since the three Yersinia species utilize similar pathogenicstrategies during infection of mice, the more innocuous Y. enterocoliticaspecies was used for initial investigations into Y. pestis vaccinecandidates. In the current study, membrane proteins were isolated fromY. enterocolitica grown under iron-limiting conditions and formulatedinto a subunit vaccine. These proteins were selected as candidateantigens due to their large size, surface exposure, and their documentedexpression during infection. The vaccine was then tested in a mousemodel of systemic infection. Following intraperitoneal vaccination, micewere challenged in the lateral tail vein with Y. enterocolitica. None of thecontrol mice survived challenge; however, 100% of the vaccinated micesurvived with no observed morbidity (p<0.0001). These resultssuggested that one or more of the proteins present in the vaccine werehighly protective against systemic challenge. Studies using matrix-assisted laser desorption ionization time-of-flight mass spectrometryidentified several proteins in this composition as outer membrane ironacquisition proteins that are well-conserved among the pathogenicYersinia. Taken together, the results of our investigations indicate thatprotein components of the Y. enterocolitica membrane from bacteriagrown under iron-limiting conditions may be effective as antigens forvaccination against plague.

References:1. Brubaker, R.R. 1991. Factor promoting acute and chronic diseases

caused by Yersiniae. Clin. Microbiol. Rev. 4:309-324.2. Carniel, E. 2001. The Yersinia high-pathogenicity island: an iron-

uptake island. Microbes Infect. 3:561-569.

Cysteine Proteinases Vased Vaccines for L. major and L. infantum InfectionsS. Rafati, T. Taheri, A. Zadeh Vakili, A. Nakhaee, F. Zahedifard, Y. Taslimi, F. DoustdariImmunology, Pasteur Institute of Iran, Tehran, IRAN.

Recently, we have evaluated the cysteine proteinases type I and II of L. infantum using a heterologous prime-boost regime for vaccinationagainst experimental visceral leishmaniasis in dogs. Followingvaccination and challenge, dogs were followed for 12 months. All dogsvaccinated by prime/boost with DNA/recombinant CPs (incombination with CpG ODN and Montanide 720), remained free ofinfection in their bone morrow. In contrast, three out of four dogs(75%) in the control groups had infection in their bone marrow. Theperipheral lymphocytes from protected animals had generally higherproliferation responses to F/T antigen, rCPA and rCPB than controls.Analysis of cytokine mRNA level suggested that vaccinated dogs hadelevated IFN-_ mRNA in PBMC whereas there was a consistent increasein the level of IL-10 in the control groups and some vaccinated dogs.The ratio of IgG2/IgG1 against all three antigens (F/T, rCPA and rCPB)in vaccinated dogs was always higher than in control dogs. We also

P65

P66

P67

Page 90: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

90

Eighth Annual Conferenceshowed that with the exception of one dog, all dogs in the vaccinatedgroup in compare to control dogs had strong DTH responses. Wepropose that the combination of DNA and recombinant proteinvaccination using CPs could be instrumental to control VL in dogs.

Development of Attenuated Mutants as Potential VaccineCandidates for Visceral LeishmaniasisP. Salotra1, G. Sreenivas2, R. Singh1, A. Selvapandiyan3, R. Duncan3, H. L. Nakhasi3

1Institute of Pathology (ICMR), New Delhi, INDIA, 2 Insitute of Pathology(ICMR), New Delhi, INDIA, 3CBER, FDA, Bethesda, MD.

Background: Visceral Leishmaniasis (VL), a fatal disease caused byLeishmania donovani, is endemic in parts of India and Sudan.Understanding the mechanisms of Leishmania virulence anddevelopment of vaccines is essential for eradication of the disease. Wehave employed microarray technology for a rapid discovery of virulence-related genes, with the goal of preparing gene knockouts of selectedgenes. Such mutant parasites, with known gene defects, have greatpotential to be used as live, attenuated vaccines. Methodology: Geneexpression was analysed in promastigote and amastigote stages of Indianisolates of L. donovani using genomic microarrays comprising ~ 5000clones1. The data was analysed by Acuity software. Deletion mutantswere prepared by double homologous gene replacement usinghygromycin and neomycin cassettes2. Results: Gene expression analysisled to identification of over one hundred differentially expressed clones.Sequence analysis of clones abundantly expressed in amastigotes revealedhomology to certain novel as well as known genes (NAD/FADdehydrogenase, reductase, gp46, gp63, amastin etc). One growthregulated gene, Centrin was targeted for deletion in Leishmaniaparasite2. Centrin double knockout mutants showed selective growtharrest as amastigotes but not as promastigotes. The null mutants failedto survive in the amastigote stage in vitro as well as in vivo, indicating animportant role for centrin in parasite growth. The centrin knock outmutant, generated with an Indian field isolate of VL, is under evaluationin animal model as a vaccine against VL. Conclusion: Several virulence-related genes were identified as targets for gene disruption to develop anattenuated vaccine against VL. Centrin gene knockout parasites that failto proliferate as amastigotes provide a promising candidate as a live,attenuated vaccine against VL.

References:1. Duncan R, Salotra P, Goyal N et al. The application of gene

expression microarray technology to kinetoplastid research Curr. Mol.Med.2004; 4:611-621.

2. Selvapandiyan A, Debrabant A, Duncan R, et al. Centrin genedisruption impairs stage-specific basal body duplication and cell cycleprogression in Leishmania. J. Biol. Chem.2004; 279:25703-25710.

Protection Studies with Candidate Schistosoma mansoniDNAVaccinesA. M. Karim1, N. El-Ghazali1, A. Medhat1, S. F. Ibrahim2

1Ain Shams Univ. Fac. of Science, Cairo, EGYPT, 2Cairo Univ. Fac. of Science,Cairo, EGYPT.

Schistosomiasis is a debilitating often deadly parasitic diseaseafflicting hundreds of millions of people worldwide. The development ofa vaccine offers hope for effective long term control. In animal studies,native and recombinant antigens have consistently failed to induce highlevels of protection as observed with irradiated live vaccine. In this studyantigens previously shown to be associated with the tegument of theschistosomule (1, 2) have been compared for their efficacy in inducingprotection by DNA vaccination. C57BL6 mice were immunized 3 timesat 4 week intervals with 125 µg of pcDNA encoding phosphoglyceratekinase (SmPGK), Sm20.8, Tropomyosin (SmTM1), SmCalponin, Sm23,or a cocktail of the 5 antigens. Mice were challenged with 150 S. mansonicercariae 5 weeks after the last immunization and worms were collectedby liver perfusion 8 weeks later. Compared to a control group injectedwith saline, significant protection levels of 26-33%, 32-40%, 24-31%and 16-20% were observed for mice groups immunized with SmPGK,Sm20.8, Sm 23 and cocktail respectively. In a second study mice groupswere immunized twice with 200 µg pcDNA encoding SmCyclophilin, afragment of SmFilamin or SmFilamin combined with recombinantFilamin protein. Significant protection levels of 14%, 15% and 19%respectively were observed. Those results demonstrate the potential forinduction of protective immune responses using DNA constructsexpressing schistosome tegumental antigens.

References:1. Lee KW, Shalaby KA, Thakur KA, et al. Cloning the gene for

phosphoglycerate kinase from Schistosoma mansoni andcharacterization of its gene product. Mol Biochem Parasitol 1995; 71:221-231.

2. Kiang D, El-Ghazalie NE, Medhat, AM, et al. Identification andcharacterization of Schistosoma mansoni p17.7 , a cyclophilin. MolBiochem Parasitol 1996; 76: 73-82.

Protective Immunity of Single and Multiple Recombinant DNA orProtein Vaccines Against Lymphatic Filariasis.P. Kaliraj, Sr.1, S. Anand1, V. Murugan1, K. KirthikaA2, M. Reddy2

1Anna University, Chennai, INDIA, 2MGIMS, Sevagram, INDIA.

Background: There is a need to develop suitable Immunoprophylacticagent for elimination of lymphatic filariasis.In this study we report thatthe dual DNA antigen expression construct or cocktail Proteinvaccination induces stronger immune responses and higher protectionthan those of an single antigen DNA/Protein (ALT or VAH) in jirds.Methods: B.malayi L3 expressed genes BmALT2 and BmVAH wereinserted individually in eukaryotic expression vector (pVAX) andcombinely in dual antigen expression eukaryotic vector(pBUDCE4).Jirds immunized with single recombinant DNA/Protein or withcombination of two DNA/ proteins by IM/IP injection. In vivo parasiteclearance study with individual or combinational DNA/Proteins bymicropore chamber against B.malayi larvae. spleenocyte proliferationlevel was checked by MTT assay.Cytokines response was evaluated byRT-PCR. The statically significance of group difference was assessed bystudent ‘t’ test. Results: Jirds produced higher antibody responses againstdual antigen constructs compared to single antigen construct. Similarlyin vivo micropore chamber cytotoxicity study using Jirds challenged with

P68

P69

P70

Page 91: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

91

(continued)

multiple antigen (pBUD ALT-VAH) showed higher (p<0.01) protection(63-70%) than single vaccine construct i.e pVAX ALT or pVAX VAH(50-60%). Similarly cocktail of two recombinant proteins rALT+rVAHshowed higher protection i.e., 80 % (p<0.01) than single protein vaccinerALT or rVAH (61-72%). Cytokine analysis of the immunized animalsshowed that recombinant proteins induced Th2 type of response whereasDNA vaccination biased towards Th1 type response. Conclusion: Ourresults show that dual antigen DNA vaccination with pBUD ALT-VAHor vaccination with cocktail protein elicit higher protective immuneresponse against B.malayi infection in jirds than single antigenDNA/Protein vaccination. The protective efficacy of the recombinantprotein was found to be better than that of DNA vaccine.

References:1. William.F.Gregory, Rick.M.Maizels, et al. The abundant larval

transcript 1&2 genes of brugia malayi encode stage specific candidatevaccine antigen for filariasis. Infection and immunity; July2000,p4174-4179.

2. Janice Murray, Rick.M.Maizels, et al. Expression and immunerecognition of brugia malyi VAL-1, a homologue of vespid venomallergen& ancylostoma secreted proteins. Molecular and Biochemicalparasitolgy, Nov2001;118(1):89-96

Enhancing Brucellosis Vaccines, Vaccine Delivery Systems andSurveillance Diagnostics for Bison and Elk in the GreaterYellowstone AreaG. E. Plumb, Jr.1, B. Marsh2

1Yellowstone National Park, US National Park Service, Yellowstone NationalPark, WY, 2Board of Animal Health, State of Indiana, Indianapolis, IN.

The Greater Yellowstone Area (GYA) is one of the largest intactecosystems in the world. It consists of 28,000 square miles in the statesof Montana, Idaho, and Wyoming. The GYA is also home to the largestwild elk (125,000) and bison (5,000) populations in North Americawhich are chronically infected with brucellosis (Brucella abortus). Thesewildlife are recognized as the last large reservoir of B. abortus in theUnited States (elk 2-30% seroprevalence and bison 40-80% seroprevalence) and there is strong concern about disease transmission toarea livestock. To date, wildlife brucellosis vaccination and diagnosticprograms are derivative of techniques developed for livestock (Schurig2002). The brucellosis vaccine RB51 has been demonstrated to confermoderate protection against spontaneous pathogenic abortion in bison(~70%), but little protection against fetal or maternal infection (~25%).Thus, while vaccination in bison or elk must be one part of an overallstrategy to control or eliminate B. abortus in the GYA, much research isneeded before current vaccines can be judged adequate for use in thesespecies (NRC 1998). There are strategically important gaps regardingvaccines, vaccine delivery systems, and surveillance diagnosticsdeveloped for the specific immunology of wild and free-ranging bisonand elk. This abstract reports on a Special Committee of The UnitedStates Animal Health Association that is evaluating vital data andproposing overarching research strategies for enhancing wildlifebrucellosis vaccines.

References:1. Cheville NF, McCullough DR, Paulson LR. Brucellosis in the Greater

Yellowstone Area. National Academy Press 1998.2. Schurig GG, Sriranganathan N, Corbel MJ. Brucellosis vaccines: past,

present, and future. Vet Microbiol 2002 90:479-496.

Canine Herpesvirus Bacterial Artificial Chromosome Technology ForAntifertility Vaccination of Foxes in AustraliaT. Strive, J. Wright, N. French, C. M. Hardy, G. H. Reubel Sustainable Ecosystems, CSIRO, Canberra ACT, AUSTRALIA.

European red foxes are a major pest in Australia that threaten thesurvival of native wildlife and reduce lamb production throughpredation. The most commonly used method to control foxes is bypoisoning with sodium mono-fluoroacetate (1080) baits, but this isconsidered non-specific, expensive and unsuitable for certain situations(e.g. urban areas). Antifertility vaccination on the other hand isrecognized as a humane, non-lethal approach to reduce animal numbersand thus their impact. The principle of this method is to induceautoimmune responses against proteins in the reproductive tract withthe aim to interrupt fertilization.

We have selected canine herpesvirus (CHV) as the preferred vectorto deliver contraception for three reasons. Firstly, viral vectors have beenshown to be effective at delivering immunocontraception to pestanimals. Secondly, CHV is specific to canids, so greatly increasingspecificity, and finally, the population of Australian foxes is highlysusceptible to CHV infection. We have demonstrated that infectiousrecombinant CHVs can be readily produced using BAC technology.Transgenes, including a green fluorescent marker protein gene and foxand porcine oocyte zona pellucida genes were inserted into CHV ineither the thymidine kinase gene or an intergenic region betweenglycoprotein H and UL21a. All rCHVs produced as BACs wererecovered as infectious recombinant viruses with similar cytopathiceffects as wild-type CHV following transfection of BAC DNA intocanine cells. The expression of transgenes by rCHVs was confirmed invitro by immunostaining of infected cell cultures and by Western blots.

References:1. Reubel GH, Pekin J, Venables D, Wright J, Zabar S, Leslie K, et al.

Experimental infection of European red foxes (Vulpes vulpes) withcanine herpesvirus. Vet Microbiol 2001;83(3):217-33.

2. Reubel GH, Pekin J, Webb-Wagg K, Hardy CM. Nucleotidesequence of glycoprotein genes B, C, D, G, H and I, the thymidinekinase and protein kinase genes and gene homologue UL24 of anAustralian isolate of canine herpesvirus. Virus Genes 2002;25(2):195

Molecular Cloning and Sequence Analysis of Bm86 cDNA from a Thai Strain of the Cattle Tick, Boophilus microplusS. Jittapalapong1, S. Thanasilp1, T. Sirinarukmitr2, K. Kaewmongkol3, R. W. Stich4

1Veterinary Parasitology, Kasetsart University, Bangkok, THAILAND,2Veterinary Pathology, Kasetsart University, Bangkok, THAILAND, 3VeterinaryCompanion Animal Medicine, Kasetsart University, Bangkok, THAILAND,4Veterinary Preventive Medicine, The Ohio State University, Columbus, OH.

Boophilus microplus is the most important ectoparasite of livestock inThailand, and is responsible for severe economic losses due to directeffects of feeding and through transmission of pathogens. Extensiveacaricide use to control these ticks has drawbacks such as selection forpesticide resistance and contamination of the environment and animalproducts. A Bm86-based anti-tick vaccine has shown promise for tickcontrol in other countries. The purpose of this work was to characterizeBm 86 from a Thai strain of B. microplus to determine if this vaccinecould be feasible in Thailand. Ticks were collected from dairy cattle in

P71

P72

P73

Page 92: On-Site Progr

ABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

92

Eighth Annual ConferenceThailand, mRNA was isolated from B. microplus midgut samples, cDNAwas amplified with Bm86-specific primers and amplicons were clonedinto the pFastBacHTB vector for sequencing and analysis with theWisconsin Package. The 2,194 bp insert contained a 1,953 bp openreading frame, and the Thai strain Bm86 protein sequence was 92.0 and92.8% identical to Cuban and Australian strain sequences, respectively.To our knowledge, this work represents the first report of Bm86 sequenceanalysis from an Asian strain of B. microplus. Bm86 divergence amongThai and other B. microplus strains suggests that further work iswarranted to determine if a geographic strain-specific vaccine would bemore effective in Thailand.

References:1. Willadsen P, Bird PE, Cobon GS, et al. Commercialization of a

recombinant vaccine against Boophilus microplus. Parasitol 1995;110: S43-S50

2. Rand KN, Moor T, Sriskantha A, et al. Cloning and expression of aprotective antigen from the cattle tick Boophilus microplus. Proc.Natl. Acad. Sci. USA 1989; 86: 9657-9661

Intranasal Vaccination of Mares to Protect Against StreptococcalUterine InfectionsR. C. Causey Animal and Veterinary Sciences, University of Maine, Orono, ME.

Background: Intranasal Salmonella vectors show promise instimulating immunity to streptococcal pathogens at mucosal sites. Anattenuated strain of Salmonella enteritica serotype typhimuriumexpressing the protective protein of Streptococcus zooepidemicus Mooreand Bryans serovar 9 (SzP-MB9) was used to explore the feasibility ofusing intranasal Salmonella vectors to boost equine uterine immunityagainst streptococci. Methods: Ten mares were randomly assigned intovaccinated and non-vaccinated groups. Mares in the vaccinated groupreceived 2 doses, 2 weeks apart, of 2.5 X 109 CFU of live intranasalvaccine. Serum, nasal and uterine washings and Salmonella cultures werecollected from both groups before and after vaccination. Immuneresponses to lipopolysaccharide (LPS) of S typhimurium and purifiedrecombinant SzP-MB9 were assessed by Enzyme-Linked-Immunosorbent-Assay (ELISA). Immune responses were detected usinganti-equine IgA, IgG and protein-G horseradish peroxidase conjugates.Ortho-phenylene-diamine (OPD) was used as substrate for colordevelopment. Optical density was measured by spectrophotometer, andresults analyzed by one-way, analysis of variance. Results: Salmonellawas not recovered from any horses post vaccination, and vaccinatedhorses remained normal on physical examination. Significant (P < 0.05)responses were seen post vaccination to LPS in serum, nasal and uterinewashings. Significant anti-SzP-MB9 IgA responses were seen in nasalwashings of vaccinated animals, but not in serum or uterine washings.Pre-existing levels of antibodies to SzP-MB9 were high in serum of allvaccinated horses. Conclusions: These data confirm that intranasalvaccination with attenuated S typhimurium may safely stimulate nasaland uterine immune responses in horses. However, pre-existingantibodies may have affected responses to SzP-MB9.

References:1. Sheoran AS, Timoney JF, Tinge SA, et al. Intranasal immunogenicity

of a _cya _crp-pabA mutant of Salmonella enterica serotypeTyphimurium for the horse. Vaccine 2001;19:3591-3599.

2. Sirard JC, Niedergang F, Kraehenbuhl JP. Live attenuated Salmonella:a paradigm of mucosal vaccines. Immunol Rev 1999;171:5-26

CpG Oligodeoxynucleotides Upregulate Antibacterial Systems and Induce Early, Non-specific Antiviral Protection in Fish.A. C. Carrington1, B. Collet2, C. J. Secombes1

1Department of Zoology, University of Aberdeen, Aberdeen, UNITEDKINGDOM, 2Fisheries Research Services, Marine Laboratory, Aberdeen,UNITED KINGDOM.

CpG oligodeoxynucleotides (CpG ODN) show promise asimmunostimmulatory agents and vaccine adjuvants in animal models.CpG ODN were tested for their capacity to stimulate bactericidal abilityand respiratory burst activity in rainbow trout (Oncorhynchus mykiss)head kidney (HK) leucocytes. Their ability to stimulate type 1interferon production was investigated using a stable RTG-2 cell linetransfected with a luciferase reporter vector and the rainbow trout Mxpromoter. Their capacity to enhance protection against bacterialchallenge alone and as an adjuvant was determined. These studiesrevealed different ODN acted in different ways with ODN 2133inducing significantly (p<0.05 by one way ANOVA, Tukey’s pairwisecomparison) more bactericidal activity and luciferase production thanseen in the cells incubated in medium alone or with ODN 2143.Supernatant from HK leucocytes incubated with ODN 2133 alsosignificantly enhanced luciferase production providing evidence that theMx promoter is activated indirectly. Conversely, ODN 2143significantly inhibited the ability of Poly I:C to activate the Mxpromoter and significantly reduced the respiratory burst in HKmacrophages in response to PMA. ODN 2143 did however,significantly enhance protection against bacterial challenge when fishwere injected with CpG alone (p<0.001 Peto test) or as an adjuvant to acommercially available vaccine (p< 0.05). These results suggest thatCpG ODN are capable of upregulating antibacterial systems, enhancingprotection against bacterial challenge and inducing early, nonspecificantiviral protection in fish but that these responses are sequence specific.

Reference:1. Jørgensen JB, Johansen L-H, Steiro K, Johansen A. CpG DNA

Induces Protective Antiviral Immune Responses in Atlantic Salmon(Salmo salar L.). Journal of Virology 2003; 77:11471-11479.

Summary of the Research of Recombinant HEV VaccineJ. LinLanzhou Institute of Biological Products, Gansu, CHINA

Background: Open reading frame (ORF-2) of the China strain ofhepatitis E virus ’s capsid protein gene (5816~7128nt;224~660aa) wasexpressed in E. Coli, a protein with a molecular mass of approximately54kDa was derived from and the purification technics of the expressedprotein was established .After having gained a great deal of purifiedpathogen, we produced HEV vaccine and carried out the test of security,immunogenicity and protection efficacy against the challenge of HEVect, all got the satisfied results. Method: To evaluate theimmunogenicity and protective efficacy of the recombinant HEVvaccine, we vaccinated several groups of Rhesus monkeys with therecombinant vaccine and challenged them with a homologous wild-typehepatitis E virus. Neither of the immunized animals showed anyelevation of alanine aminotransferase activity after intravenous challengewith wild-type HEV stool isolate, in marked contrast with theunimmunized (control) cynos. Microscopic examination of liver biopsyspecimens from these immunized cyno failed to detect histopathologicevidence of viral hepatitis. All primates which were vaccinated thrice

P74

P75

P76

Page 93: On-Site Progr

on Vaccine ResearchABSTRACTS OF SUBMITTED POSTER PRESENTATIONS

93

(continued)

with the recombinant protein were protected from hepatitis when theywere challenged with very high doses of the homologous strain of HEV.The immune process was three times injection in 0, 2, 4 weeks. Primatesvaccinated thrice with a 40µg dose of the recombinant vaccineformulated with Aluminium Hydroxide Gel adjuvant gave high andequivalent levels of antibody and were completely protected fromhepatitis after challenge with wild-type hepatitis E virus .Results: The results indicated that the rhesus immunized byprokaryotic expressed recombinant HEV structural protein couldeffectively defend wild HEV virus attacking.

Page 94: On-Site Progr

AUTHOR INDEX

94

Author Presentation Number

Aaberge, I. . . . . . . . . . . . . . . . . .P50, P51

Aase, A. . . . . . . . . . . . . . . . . . . . . . . .P50

Acevedo, J. . . . . . . . . . . . . . . . . . . . . .S1

Ahlers, N. . . . . . . . . . . . . . . . . . . . . . . .S4

Ahmed, T. . . . . . . . . . . . . . . . . . . . . .P32

Alexander, J. . . . . . . . . . . . . . . . . . . . .S16

Alexander, N. . . . . . . . . . . . . . . . . . . . .S8

Anand, S. . . . . . . . . . . . . . . . . . . . . . .P70

Aparin, P. . . . . . . . . . . . . . . . . . . . . . .S10

Apelian, D. . . . . . . . . . . . . . . . . . . . . .P56

Arimoto, Y. . . . . . . . . . . . . . . . . . . . .P62

Arroyo, J. . . . . . . . . . . . . . . . . . . . . . .P63

Arsenault, G. . . . . . . . . . . . . . . . . . . .S13

Assadou, M. . . . . . . . . . . . . . . . . . . . .S12

Atmar R. . . . . . . . . . . . . . . . . . . . . . . . .8

Azimpour, C. . . . . . . . . . . . . . . . . . . .P34

Baatar, D. . . . . . . . . . . . . . . . . . . . . . . .S5

Baby, M. . . . . . . . . . . . . . . . . . . . . . . .S12

Bachmann, M. . . . . . . . . . . . . . . . . . . .P6

Badger, C. . . . . . . . . . . . . . . . . . . . . .P16

Bagayogo, M. . . . . . . . . . . . . . . . . . . .P31

Ballou, R. . . . . . . . . . . . . . . . . . . . . . . .21

Bansal, A. . . . . . . . . . . . . . . . . . . . . . .P46

Barman, S. . . . . . . . . . . . . . . . . . . . . .P60

Barnoy, S. . . . . . . . . . . . . . . . . . . . . . .P39

Barrett, A. . . . . . . . . . . . . . . . . . . . . . .14

Barry, E. . . . . . . . . . . . . . . . . . . . . . . .S11

Bastien, N. . . . . . . . . . . . . . . . . . . . . .S15

Baxendale, D. . . . . . . . . . . . . . . . . . . .S18

Becker, P. . . . . . . . . . . . . . . . . . . . . . .P34

Begum, Y. . . . . . . . . . . . . . . . . . . . . . .P32

Bekan, B. . . . . . . . . . . . . . . . . . . . . . .P27

Bell, S. . . . . . . . . . . . . . . . . . . . . . . . .P63

Bellgrau, D. . . . . . . . . . . . . . . . . . . . .P56

Bellone, C. . . . . . . . . . . . . . . . . . . . . .P12

Author Presentation Number

Belshe, R. . . . . . . . . . . . . . . . . . . . . . .P12

Berg, M. . . . . . . . . . . . . . . . . . . . . . . .P38

Berger, S. . . . . . . . . . . . . . . . . . . . . . .S15

Bethony, J. . . . . . . . . . . . . . . . . . . .S7, S8

Bhattacharyya, S. . . . . . . . . . . . . . . . .S17

Billingsley, S. . . . . . . . . . . . . . . . . . . .P63

Bilsel, P. . . . . . . . . . . . . . . . . . . . . . . .S16

Biragyn, A. . . . . . . . . . . . . . . . . . . . . . .S5

Bishop, R. . . . . . . . . . . . . . . . . . . . . .P45

Blackwelder, W. . . . . . . . . . . . . . . . . .S11

Borrow, R. . . . . . . . . . . . . . . . . . . . . .P15

Bottazzi, M. . . . . . . . . . . . . . . . . . .S7, S8

Boulianne, N. . . . . . . . . . . . . . . . . . . .P11

Bray, M. D. . . . . . . . . . . . . . . . . . . . .S17

Breiman, R. . . . . . . . . . . . . . . . . . . . .P32

Brooker, S. . . . . . . . . . . . . . . . . . . .S7, S8

Brunham, R. . . . . . . . . . . . . . . . . . . .S15

Buller, M. . . . . . . . . . . . . . . . . . . . . . .P12

Burkhard, P. . . . . . . . . . . . . . . . . . . . . .P6

Burt, D. . . . . . . . . . . . . . . . . . . . . . . .P49

Bush, G. . . . . . . . . . . . . . . . . . . . . . . .P16

Bystricky, S. . . . . . . . . . . . . . . . . . . . .P41

Campbell, J. . . . . . . . . . . . . . . . . . . . .P26

Cao, H. . . . . . . . . . . . . . . . . . . . . . . .S14

Capps, W. . . . . . . . . . . . . . . . . . . . . .P16

Carbone, K. . . . . . . . . . . . . . . . . . . . .S24

Carrington, A. . . . . . . . . . . . . . . . . . .P75

Carter, D. . . . . . . . . . . . . . . . . . . . . . . .P7

Caugant, D. . . . . . . . . . . . . . . . .P50, P51

Causey, R. . . . . . . . . . . . . . . . . . . . . .P74

Chabot, S. . . . . . . . . . . . . . . . . . . . . .P49

Cheng, S. . . . . . . . . . . . . . . . . . . . . . .P23

Choi, J. . . . . . . . . . . . . . . . . . . .P13, P18

Chou, T. . . . . . . . . . . . . . . . . . . . . . . .P48

Author Presentation Number

Chowdhury, M. . . . . . . . . . . . . . . . . .P32

Chuprinina, R. . . . . . . . . . . . . . . . . . .S10

Cisar, J. O. . . . . . . . . . . . . . . . . . . . . .P24

Clemens, J. . . . . . . . . . . . . . . . . . . . . .P32

Clymer, J. . . . . . . . . . . . . . . . . . . . . . . .18

Cohen, S. . . . . . . . . . . . . . . . . . . . . . .P46

Collet, B. . . . . . . . . . . . . . . . . . . . . . .P75

Cook, J. . . . . . . . . . . . . . . . . . . . . . . .P46

Cope, L. . . . . . . . . . . . . . . . . . . . . . . .P46

Cordeiro-da-Silva . . . . . . . . . . . . . . . .P47

Costantino, P. . . . . . . . . . . . . . . . . . . .P51

Crowe, J. . . . . . . . . . . . . . . . . . . . . . . .28

Cuberos, L. . . . . . . . . . . . . . . . . . . . .P31

Cutts F. . . . . . . . . . . . . . . . . . . . . . . . . . .5

Cyr, S. . . . . . . . . . . . . . . . . . . . . . . . .P49

Daigneault, J. . . . . . . . . . . . . . . . . . . .S13

Dalloul, R. . . . . . . . . . . . . . . . . . . . . .S20

Daly, P. . . . . . . . . . . . . . . . . . . . . . . . .S13

de Roux, A. . . . . . . . . . . . . . . . . . . . . .S4

Decker, M. . . . . . . . . . . . . . . . . . . . . .S18

Del Giudice, G. . . . . . . . . . . . . . . . . . .11

dela Cruz, T. . . . . . . . . . . . . . . . . . . . .P64

Dhiman, N. . . . . . . . . . . . . . . . . . . . .S23

Diallo, D. . . . . . . . . . . . . . . . . . .S12, P26

Diaz-Mitoma, F. . . . . . . . . . . . . . .S2, P36

Dicko, A. . . . . . . . . . . . . . . . . . . . . . .S12

Diemert, D. . . . . . . . . . . . . . . . . . . . .S12

DiMiceli, L. . . . . . . . . . . . . . . . . . . . .P61

Ding, X. . . . . . . . . . . . . . . . . . . . . . . .S20

Dion, R. . . . . . . . . . . . . . . . . . . . . . . .P11

Dionne, M. . . . . . . . . . . . . . . . . .S2, P30

Doherty, C. . . . . . . . . . . . . . . . . . . . . .P8

Dolo, A. . . . . . . . . . . . . . . . . . . . . . . .S12

AUTHOR INDEX

Eighth Annual Conference

Page 95: On-Site Progr

on Vaccine ResearchAUTHOR INDEX

95

Author Presentation Number

Doumbia, M. . . . . . . . . . . . . . . . . . . .P31

Doumbo, O. . . . . . . . . . . . . . . . . . . .S12

Doustdari, F. . . . . . . . . . . . . . . . . . . .P67

Dowling, W. . . . . . . . . . . . . . . . . . . . .P16

Du, X. . . . . . . . . . . . . . . . . . . . . . . . . .S6

Dubovsky, F. . . . . . . . . . . . . . . . . . . . . .19

Duarte-Monteiro . . . . . . . . . . . . . . . .P28

Duke, R. . . . . . . . . . . . . . . . . . . . . . .P56

Duncan, R. . . . . . . . . . . . . . . . . . . . .P68

Dunning, A. . . . . . . . . . . . . . . . . . . . . .P4

Duval, B. . . . . . . . . . . . . . . . . . .P11, P36

Ebensen, T. . . . . . . . . . . . . . . . . . . . . .P34

Edwards, S. . . . . . . . . . . . . . . . . . . . . .P4

El-Ghazali, N. . . . . . . . . . . . . . . . . . .P69

Elias, F. . . . . . . . . . . . . . . . . . . . . . . . . .P5

Elkina, S. . . . . . . . . . . . . . . . . . . . . . .S10

Elrick, D. . . . . . . . . . . . . . . . . . . . . . .P36

Emery, D. . . . . . . . . . . . . . . . . .P42, P66

Falgout, B. . . . . . . . . . . . . . . . . . . . . .P38

Fan, H. . . . . . . . . . . . . . . . . . . . . . . . .P46

Faruque, S. . . . . . . . . . . . . . . . . . . . . .P32

Feiring, B. . . . . . . . . . . . . . . . . . . . . .P51

Fernsten, P. . . . . . . . . . . . . . . . . .P14, P15

Ferro, S. . . . . . . . . . . . . . . . . . . . . . . .P30

Finlay, B. . . . . . . . . . . . . . . . . . . . . . .S19

Fletcher, P. . . . . . . . . . . . . . . . . . . . . . .P4

Flo, J. . . . . . . . . . . . . . . . . . . . . . . . . . .P5

Franzusoff, A. . . . . . . . . . . . . . . . . . . .P56

French, N. . . . . . . . . . . . . . . . . . . . . .P72

Frey, S. . . . . . . . . . . . . . . . . . . . . . . . .P12

Fritzsønn, E. . . . . . . . . . . . . . . . . . . . .P50

Frolushkina, T. . . . . . . . . . . . . . . . . . .S10

Fulford, T. . . . . . . . . . . . . . . . . . . . . . .P8

Gahan, M. . . . . . . . . . . . . . . . . . . . . .S19

Author Presentation Number

Galanis, E. . . . . . . . . . . . . . . . . . . . . .S13

Gantcho, T. . . . . . . . . . . . . . . . . . . . .S10

García-Sastre, A. . . . . . . . . . . . . . . . . .S14

Gaudreau, M. . . . . . . . . . . . . . . . . . . .P43

Geiger S . . . . . . . . . . . . . . . . . . . . . . . .S8

George, H. . . . . . . . . . . . . . . . . . . . . .P46

Giardina, P. . . . . . . . . . . . . . . . . . . . .P15

Gilca, V. . . . . . . . . . . . . . . . . . . . . . . .P11

Goldblatt D. . . . . . . . . . . . . . . . . . . . . . .3

Golovina, M. . . . . . . . . . . . . . . . . . . .S10

Gordon, G. . . . . . . . . . . . . . . . . . . . .P56

Goshima, T. . . . . . . . . . . . . . . . . . . . .P62

Goud, G. . . . . . . . . . . . . . . . . . . . . . . .S7

Graff, A. . . . . . . . . . . . . . . . . . . . . . . . .P6

Grey, L. . . . . . . . . . . . . . . . . . . . . . . .P16

Gruber,W. . . . . . . . . . . . . . . . . . . . . . .S4

Guasparini, R. . . . . . . . . . . . . . . . . . . .S2

Guindo, O. . . . . . . . . . . . . . . . . . . . .S12

Guttieri M . . . . . . . . . . . . . . . . . . . . .S21

Guzman, C. . . . . . . . . . . . . . . . . . . . .P34

Hall, A. . . . . . . . . . . . . . . . . . . . . . . . .P8

Haller, A. . . . . . . . . . . . . . . . . . . . . . .P56

Halperin, S. . . . . .S2, S18, P28, P30, P36

Haran, J. . . . . . . . . . . . . . . . . . . . . . .S14

Hardy, C. . . . . . . . . . . . . . . . . . . . . . .P72

Harris, S. . . . . . . . . . . . . . . . . . . . . . .P14

Hartwell, E. . . . . . . . . . . . . . . . . . . . .P57

Hayney, M. . . . . . . . . . . . . . . . . .P9, P10

Heckert, R. . . . . . . . . . . . . . . . . . . . . .S20

Hepler, R. . . . . . . . . . . . . . . . . . . . . .P46

Heppner, D. . . . . . . . . . . . . . . . . . . . . .22

Heymann D. . . . . . . . . . . . . . . . . . . . . .1

Higgins, D. . . . . . . . . . . . . . . . . . . . .P64

Hill, R. . . . . . . . . . . . . . . . . . . . . . . . . .15

Author Presentation Number

Hogan, R. . . . . . . . . . . . . . . . . . . . . .P16

Høiby, E. . . . . . . . . . . . . . . . . . . . . . .P50

Holst, J. . . . . . . . . . . . . . . . . . . . . . . .P51

Honjo, A. . . . . . . . . . . . . . . . . . . . . . .P62

Honko, A. N. . . . . . . . . . . . . . . . . . . . .P2

Hooper J . . . . . . . . . . . . . . . . . . . . . .S21

Hotez, P. . . . . . . . . . . . . . . . . . . . . .S7, S8

Houghton, M. . . . . . . . . . . . . . . . . . . .24

Hu, A. . . . . . . . . . . . . . . . . . . . . . . . .P23

Hu, R. . . . . . . . . . . . . . . . . . . . . . . . .P40

Huang, X. . . . . . . . . . . . . . . . . . . . . . .S14

Hussell, T. . . . . . . . . . . . . . . . . . . . . . .29

Hutt, P. . . . . . . . . . . . . . . . . . . . . . . . .P8

Ibrahim, S. . . . . . . . . . . . . . . . . . . . . .P69

Inoue, S. . . . . . . . . . . . . . . . . . . . . . . .P65

Insel, R. . . . . . . . . . . . . . . . . . . . . . . . .26

Iskander, J. . . . . . . . . . . . . . . . . . . . . .P61

Jacobsen, S. . . . . . . . . . . . . . . . . . . . .S23

Jacobson, R. . . . . . . . . . . . . . . . .S22, S23

Jansen, K. . . . . . . . . . . . . . . . . . . . . . .P46

Jechlinger, W. . . . . . . . . . . . . . . . . . . .P34

Jefferies, D. . . . . . . . . . . . . . . . . . . . . .P8

Jin, H. . . . . . . . . . . . . . . . . . . . . . . . . .P3

Jittapalapong, S. . . . . . . . . . . . . . . . . .P73

Johnson, D. . . . . . . . . . . . . . . . . . . . . .17

Jones, T. . . . . . . . . . . . . . . . . . . . . . . .P49

Joyce, J. . . . . . . . . . . . . . . . . . . . . . . .P46

Kaewmongkol, K. . . . . . . . . . . . . . . .P73

Kaliraj, P. . . . . . . . . . . . . . . . . . . . . . .P70

Kallos, A. . . . . . . . . . . . . . . . . . . . . . .P29

Kamate, B. . . . . . . . . . . . . . . . . . . . . .S12

Kamau, L. . . . . . . . . . . . . . . . . . . . . .P45

Kang, C. . . . . . . . . . . . . . . . . . . . . . . .P18

Kang, Y. . . . . . . . . . . . . . . . . . . . . . . . .P3

Page 96: On-Site Progr

AUTHOR INDEX

96

Author Presentation Number

Kappe, S. . . . . . . . . . . . . . . . . . . . . . . .20

Karim, A. . . . . . . . . . . . . . . . . . . . . . .P69

Kast, W. . . . . . . . . . . . . . . . . . . . . . . . .23

Kato, T. . . . . . . . . . . . . . . . . . . . . . . .P62

Katsuta, T. . . . . . . . . . . . . . . . . . . . . .P62

Katz, J. . . . . . . . . . . . . . . . . . . . . . . . .S16

Keller, P. . . . . . . . . . . . . . . . . . . . . . . .P46

Kelly, R. . . . . . . . . . . . . . . . . . . . . . . .P46

Kemmler, C. . . . . . . . . . . . . . . . . . . . .P56

Ketner, G. . . . . . . . . . . . . . . . . . . . . .P38

Kidokoro, M. . . . . . . . . . . . . . . . . . . .P65

Killeen, K. . . . . . . . . . . . . . . . . . . . . .P32

King, A. . . . . . . . . . . . . . . . . . . . . . . .S15

King, T. . . . . . . . . . . . . . . . . . . . . . . .P56

Kirithika, K. . . . . . . . . . . . . . . . . . . . .P70

Kitabatake, M. . . . . . . . . . . . . . . . . . .P65

Klein, J. . . . . . . . . . . . . . . . . . . . . . . . .16

Klinman, D. . . . . . . . . . . . . . . . . . . . .S20

Klugman K. . . . . . . . . . . . . . . . . . . . . . .2

Koehm, S. . . . . . . . . . . . . . . . . . . . . .P12

Kofler, R. . . . . . . . . . . . . . . . . . . . . . .P20

Kohara, M. . . . . . . . . . . . . . . . . . . . . .P65

KoKai-kun, J. . . . . . . . . . . . . . . . . . . . .S1

Kopecko, D. J. . . . . . . . . . . . . . .P24, S17

Kotloff, K. . . . . . . . . . . .9, S11, P26, P31,

Kristiansen, P. . . . . . . . . . . . . . .P50, P51

Kurbatova, I. Yu. . . . . . . . . . . . . . . . .S10

Kurtz, M. . . . . . . . . . . . . . . . . . . . . . .P46

Lacasse, P. . . . . . . . . . . . . . . . . . . . . . .P43

LaJeunesse, C. . . . . . . . . . . . . . . . . . .P29

Langley, J. . . . . . . . . . . . . . . . . . .S2, P30

Lanzavecchia, A. . . . . . . . . . . . . . . . . . .30

Larder, A. . . . . . . . . . . . . . . . . . . . . . .S15

Lavigne, P. . . . . . . . . . . . . . . . . . . . . .S18

Author Presentation Number

Law, B. . . . . . . . . . . . . . . . . . . . . .S2, P36

Lawrence, S. . . . . . . . . . . . . . . . . . . . .P12

Lee, S. . . . . . . . . . . . . . . . . . . . . . . . .P18

Leeper, A. . . . . . . . . . . . . . . . . . . . . . . .P4

Lees, A. . . . . . . . . . . . . . . . .P52, P53, S1

Leffel, E. . . . . . . . . . . . . . . . . . . . . . .P46

Levine, M. . . . . . . . . . . . . .S11, P26, P31

Li, C. . . . . . . . . . . . . . . . . . . . . . . . . . .P7

Li, F. . . . . . . . . . . . . . . . . . . . . . . . . . .P23

Li, H. . . . . . . . . . . . . . . . . . . . . . . . . .P40

Li, L. . . . . . . . . . . . . . . . . . . . . . . . . .P17

Li, Y. . . . . . . . . . . . . . . . . . . . . . . . . . .S15

Libraty, D. . . . . . . . . . . . . . . . . . . . . .P60

Lillehoj, E. . . . . . . . . . . . . . . . . . . . . .S20

Lillehoj, H. . . . . . . . . . . . . . . . . . . . . .S20

Lin, J. . . . . . . . . . . . . . . . . . . . . . . . . .P76

Liu, J. . . . . . . . . . . . . . . . . . . . . . . . . .P17

Locher, C. . . . . . . . . . . . . . . . . . . . . . .33

Lockhart, S. . . . . . . . . . . . . . . . . . . . . .P4

Lode, H. . . . . . . . . . . . . . . . . . . . . . . . .S4

Long, C. . . . . . . . . . . . . . . . . . . . . . . .S12

Longworth, E. . . . . . . . . . . . . . . . . . .P15

Lopez, R. . . . . . . . . . . . . . . . . . . . . . . .P5

Lopez A. . . . . . . . . . . . . . . . . . . . .P53, S1

Losada, M. . . . . . . . . . . . . . . . . . . . . .P46

Lottenbach, K. . . . . . . . . . . . . . . . . . .P12

Loukas, A. . . . . . . . . . . . . . . . . . . . . . .S7

Lu, S. . . . . . . . . . . . . . . . . . . . . .P48, S14

Lu, Y. . . . . . . . . . . . . . . . . . . . . . . . . .P56

Lubitz, W. . . . . . . . . . . . . . . . . . . . . .P34

Lvov, V. L. . . . . . . . . . . . . . . . . . . . . .S10

Ma, W. . . . . . . . . . . . . . . . . . . . . . . . .P17

Machaidze, G. . . . . . . . . . . . . . . . . . . .P6

Malkin, E. . . . . . . . . . . . . . . . . . . . . .S12

Author Presentation Number

Mandl, C. . . . . . . . . . . . . . . . . . . . . .P20

Marsh, B. . . . . . . . . . . . . . . . . . . . . . .P71

Marty, K. . . . . . . . . . . . . . . . . . . . . . .P29

Matsuda, T. . . . . . . . . . . . . . . . . . . . .P35

Matsumoto, T. . . . . . . . . . . . . . . . . . .P35

Matsushima, K. . . . . . . . . . . . . . . . . .P65

Maus, D. . . . . . . . . . . . . . . . . . . . . . .S18

Mbawuike, I. . . . . . . . . . . . . . . . . . . .P64

Mboujka, I. . . . . . . . . . . . . . . . . . . . .S14

McCaughey, M. . . . . . . . . . . . . . . . . . .P4

McConkey, S. . . . . . . . . . . . . . . . . . . . .P8

McElroy A. . . . . . . . . . . . . . . . . . . . . .S21

McNeely, T. . . . . . . . . . . . . . . . . . . . .P46

McNeil, S. . . . . . . . . . . . . . . . . . . . . .P30

Medhat, A. . . . . . . . . . . . . . . . . . . . . .P69

Meekison, W. . . . . . . . . . . . . . . . . . . .P30

Mekalanos, J. . . . . . . . . . . . . . . . . . . .P32

Meloff, K. . . . . . . . . . . . . . . . . . . . . .P28

Mendez, S. . . . . . . . . . . . . . . . . . . . . . .S7

Mendy, M. . . . . . . . . . . . . . . . . . . . . . .P8

Meyer Næss, L. . . . . . . . . . . . . . . . . .P51

Michalek S. . . . . . . . . . . . . . . . . . . . . .10

Miller E. . . . . . . . . . . . . . . . . . . . . . . . . .4

Miller, L. . . . . . . . . . . . . . . . . . . . . . .S12

Mills, E. . . . . . . . . . . . . . . . . . . . .S2, P30

Min, W. . . . . . . . . . . . . . . . . . . . . . . .S20

Mitchell, A. . . . . . . . . . . . . . . . . . . . .P63

Mizel, S. . . . . . . . . . . . . . . . . . . . . . . . .P2

Mizuno, K. . . . . . . . . . . . . . . . . . . . . .P65

Monath, T. . . . . . . . . . . . . . . . . . . . . . .34

Mond, J. . . . . . . . . . . . . . . . . . . . . . . . .S1

Montaner, A. . . . . . . . . . . . . . . . . . . . .P5

Moon, H. . . . . . . . . . . . . . . . . . . . . . .P18

Morcol, T. . . . . . . . . . . . . . . . . . . . . .P63

AUTHOR INDEX

Eighth Annual Conference

Page 97: On-Site Progr

on Vaccine ResearchAUTHOR INDEX

97

Author Presentation Number

Morita, K. . . . . . . . . . . . . . . . . . . . . .P65

Mozel, M. . . . . . . . . . . . . . . . . . . . . .P29

Mullen, G. . . . . . . . . . . . . . . . . . . . . .S12

Murai, F. . . . . . . . . . . . . . . . . . . . . . .P65

Murugan, V. . . . . . . . . . . . . . . . . . . . .P70

Musoke, T. . . . . . . . . . . . . . . . . . . . . .P45

Nagaoka, C. . . . . . . . . . . . . . . . . . . . .P62

Nagappan, P. . . . . . . . . . . . . . . . . . . .P63

Nakajima, N. . . . . . . . . . . . . . . . . . . .P62

Nakhaee, A. . . . . . . . . . . . . . . . . . . . .P67

Nakhasi, H. . . . . . . . . . . . . . . . . . . . .P68

Narayanan, P. . . . . . . . . . . . . . . . . . . .P19

Nataro, J. . . . . . . . . . . . . . . . . . . . . . .S11

Neatby, A. . . . . . . . . . . . . . . . . . . . . .S18

Nerenbaum, L. . . . . . . . . . . . . . . . . . .P63

Newman, F. . . . . . . . . . . . . . . . . . . . .P12

Newman, M. . . . . . . . . . . . . . . . . . . .S16

Niambele, M. . . . . . . . . . . . . . . . . . . .S12

Norheim, G. . . . . . . . . . . . . . . .P50, P51

Noya, F. . . . . . . . . . . . . . . . . . . . . . . .P30

Ojah, C. . . . . . . . . . . . . . . . . . . . . . . .P30

Okuda, K. . . . . . . . . . . . . . . . . . . . . .P35

Osorio, M. . . . . . . . . . . . . . . . . . . . . .S17

Osterhaus, A. . . . . . . . . . . . . . . . . . . . .12

Ott, G. . . . . . . . . . . . . . . . . . . . . . . .P64

Ouaissi, A. . . . . . . . . . . . . . . . . . . . . .P47

Ovsyannikova, I. . . . . . . . . . . . .S22, S23

Owusu-Agyei, S . . . . . . . . . . . . . . . . .P22

Pankratz, S. V. . . . . . . . . . . . . . .S22, S23

Paragas, J. . . . . . . . . . . . . . . . . . . . . . .P16

Pasetti, M. . . . . . . . . . . . . . . . . .P31, S11

Pavlova, L. . . . . . . . . . . . . . . . . . . . . .S10

Petric, M. . . . . . . . . . . . . . . . . . . . . . .S15

Pielak, K. . . . . . . . . . . . . . . . . . . . . . .S13

Author Presentation Number

Pitt, M. . . . . . . . . . . . . . . . . . . . . . . .P46

Plumb, G. . . . . . . . . . . . . . . . . . . . . .P71

Poland, G. . . . . . . . . . . . . . . . . .S22, S23

Pool, V. . . . . . . . . . . . . . . . . . . . . . . .P61

Predy, G. . . . . . . . . . . . . . . . . . . .S2, P30

Promkhatkaew, D. . . . . . . . . . . . . . . .P55

Protodiakonov, A. . . . . . . . . . . . . . . . .S10

Qadri, F. . . . . . . . . . . . . . . . . . . . .P32, S9

Rafati, S. . . . . . . . . . . . . . . . . . . . . . .P67

Raman, S. . . . . . . . . . . . . . . . . . . . . . . .P6

Rames, J. . . . . . . . . . . . . . . . . . . . . . .P25

Ranallo, R. . . . . . . . . . . . . . . . . . . . . .P39

Rangarajan, P. . . . . . . . . . . . . . . . . . . .P33

Rappaport, R. . . . . . . . . . . . . . . . . . .P23

Rappuoli, R. . . . . . . . . . . . . . . . . . . . .P51

Rayco-Solon, P. . . . . . . . . . . . . . . . . . .P8

Reddy, M. . . . . . . . . . . . . . . . . . . . . .P70

Remple, V. . . . . . . . . . . . . . . . . . . . . .S13

Reubel, G. . . . . . . . . . . . . . . . . . . . . .P72

Robbins J . . . . . . . . . . . . . . . . . . . . . . . .6

Robinson, H. . . . . . . . . . . . . . . . . . . . .31

Rodell, T. . . . . . . . . . . . . . . . . . . . . . .P56

Rodrigues, L. . . . . . . . . . . . . . . . . . . . .S8

Rodriguez, J. . . . . . . . . . . . . . . . . . . . .P5

Rosenqvist, E. . . . . . . . . . . . . . .P50, P51

Roser, B. . . . . . . . . . . . . . . . . . . . . . . .P57

Rowlands, J. . . . . . . . . . . . . . . . . . . . .P45

Rubin, S. . . . . . . . . . . . . . . . . . . . . . .S24

Ryan, J. . . . . . . . . . . . . . . . . . . . . . . .S23

Rykers, P. . . . . . . . . . . . . . . . . . . . . . .S18

Sack, D. . . . . . . . . . . . . . . . . . . . .P32, S9

Sagara, I. . . . . . . . . . . . . . . . . . . . . . .S12

Saha, A. . . . . . . . . . . . . . . . . . . . . . . .P32

Saha, S. . . . . . . . . . . . . . . . . . . . . . . .P35

Author Presentation Number

Sakhatskyy, P. . . . . . . . . . . . . . . . . . . .P48

Salam, M. . . . . . . . . . . . . . . . . . . . . . .P32

Salotra, P. . . . . . . . . . . . . . . . . . . . . . .P68

Sasaki, S. . . . . . . . . . . . . . . . . . . . . . .P35

Sauder, C. . . . . . . . . . . . . . . . . . . . . . .S24

Saul, A. . . . . . . . . . . . . . . . . . . . . . . . .S12

Scheifele, D. . . . . . . . . . . . . . . . .P28, P29

Schellekens, C. . . . . . . . . . . . . . . . . . . .P6

Schenk, D. . . . . . . . . . . . . . . . . . . . . . .25

Schiavo, R. . . . . . . . . . . . . . . . . . . . . . .S5

Schmaljohn, C. . . . . . . . . . . . . .P16, S21

Schmoele-Thoma . . . . . . . . . . . . . . . . .S4

Schultz, L. . . . . . . . . . . . . . . . . . . . . .P46

Secombes, C. . . . . . . . . . . . . . . . . . . .P75

Seidlein, L. . . . . . . . . . . . . . . . . . . . . .P32

Selvapandiyan, A . . . . . . . . . . . . . . . .P68

Sen, S. . . . . . . . . . . . . . . . . . . . . . . . .P57

Seong, S. . . . . . . . . . . . . . . . . . . . . . .P18

Serres, G. De. . . . . . . . . . . . . . . . . . . .P11

Seth, P. . . . . . . . . . . . . . . . . . . . . . . . .P54

Shadomy, S. . . . . . . . . . . . . . . . . . . . .P61

Shafer, D. . . . . . . . . . . . . . . . . . . . . . .P52

Shi, R. . . . . . . . . . . . . . . . . . . . . . . . .P17

Shida, H. . . . . . . . . . . . . . . . . . . . . . .P65

Shiver, J. . . . . . . . . . . . . . . . . . . . . . . . .32

Shmigol, V. . . . . . . . . . . . . . . . . . . . . .S10

Shurtleft, A. . . . . . . . . . . . . . . . . . . . .S21

Siber, G. . . . . . . . . . . . . . . . . . . . . .S4, P4

Siegrist, C. . . . . . . . . . . . . . . . . . . . . . .27

Sikkema, D. . . . . . . . . . . . . . . . . . .S4, P4

Silveria, I. . . . . . . . . . . . . . . . . . .P58, P59

Silvestre, R. . . . . . . . . . . . . . . . . . . . .P47

Simon, J. K. . . . . . . . . . . . . . . . . . . . .S11

Singh, R. . . . . . . . . . . . . . . . . . . . . . .P68

Page 98: On-Site Progr

Author Presentation Number

Sirinarukmitr, T. . . . . . . . . . . . . . . . . .P73

Sissoko, M. . . . . . . . . . . . . . . . . . . . . .S12

Skilton, R. . . . . . . . . . . . . . . . . . . . . .P45

Skowronski, D. . . . . . . . . . . . . .S13, S15

Skrastina, D. . . . . . . . . . . . . . . . . . . .P44

Smith, B. . . . . . . . . . . . . . . . . . .S18, P28

Smith, J. . . . . . . . . . . . . . . . . . . . . . . .P16

Sogoba, M. . . . . . . . . . . . . . . . . . . . . .S12

Soistman, E. . . . . . . . . . . . . . . . . . . . . .P7

Solórzano, A. . . . . . . . . . . . . . . . . . . .S14

Sominskaya, I. . . . . . . . . . . . . . . . . . .P44

Sow, S. . . . . . . . . . . . . . . . . . . . .P26, P31

Spik, K. . . . . . . . . . . . . . . . . . . . . . . .S21

Sreenivas, G. . . . . . . . . . . . . . . . . . . .P68

Srinivasan, V. . . . . . . . . . . . . . . . . . . .P33

Stanley, S. . . . . . . . . . . . . . . . . . . . . . .P12

Steele, D. . . . . . . . . . . . . . . . . . . . . . . . .7

Stewart, B. . . . . . . . . . . . . . . . . . . . . .S16

Stich, R. . . . . . . . . . . . . . . . . . . . . . . .P73

Stoever, K. . . . . . . . . . . . . . . . . . . .S7, S8

Stoica-Popescu . . . . . . . . . . . . . . . . . .P49

Straub, D. . . . . . . . . . . . . . . . . .P42, P66

Strive, T. . . . . . . . . . . . . . . . . . . . . . . .P72

Strugnell, R. . . . . . . . . . . . . . . . . . . . .S19

Sujatha, N. . . . . . . . . . . . . . . . . . . . . .P19

Swayne, D. . . . . . . . . . . . . . . . . . . . . . .13

Sweet, L. . . . . . . . . . . . . . . . . . . . . . . .S18

Sztein, M. . . . . . . . . . . . . . . . . . . . . . .S11

Taaffe, J. . . . . . . . . . . . . . . . . . . . . . . .S14

Taheri, T. . . . . . . . . . . . . . . . . . . . . . .P67

Takeshita, F. . . . . . . . . . . . . . . . . . . . .P35

Talbot, B. . . . . . . . . . . . . . . . . . . . . . .P43

Tam, J. . . . . . . . . . . . .S13, S15, P14, P23

Tangen, T. . . . . . . . . . . . . . . . . . . . . .P50

Author Presentation Number

Tapia, M. . . . . . . . . . . . . . . . . . .P26, P31

Tapiero, B. . . . . . . . . . . . . . . . . . .S2, P36

Taslimi, Y. . . . . . . . . . . . . . . . . . . . . .P67

Tateyama, S. . . . . . . . . . . . . . . . . . . . .P62

Thakkar, S. . . . . . . . . . . . . . . . . . . . . .P39

Thanasilp, S. . . . . . . . . . . . . . . . . . . .P73

Thera, M. . . . . . . . . . . . . . . . . . . . . . .S12

Thompson, E. . . . . . . . . . . . . . . . . . .P16

Tissot, A. . . . . . . . . . . . . . . . . . . . . . . .P6

Tokutake, T. . . . . . . . . . . . . . . . . . . . .P62

Tomovici, A. . . . . . . . . . . . . . . . . . . . . .S2

Tropel, D. . . . . . . . . . . . . . . . . . . . . . .P6

Tuck, S. . . . . . . . . . . . . . . . . . . . . . . .P64

Tweed, S. . . . . . . . . . . . . . . . . . .S13, S15

van der Sande, M. . . . . . . . . . . . . . . . .P8

Van Nest, G. . . . . . . . . . . . . . . . . . . .P64

Vandenburgh, K. . . . . . . . . . . . . . . . .S24

Venkatesan, M. . . . . . . . . . . . . . . . . .P39

Vierkant, R. . . . . . . . . . . . . . . . .S22, S23

Vivekanandhan, A . . . . . . . . . . . . . . .P19

Vrati, S. . . . . . . . . . . . . . . . . . . . . . . .P37

Vu Khac, H. . . . . . . . . . . . . . . . . . . . .P21

Waight, P. . . . . . . . . . . . . . . . . . . . . . . .P8

Walker, R. . . . . . . . . . . . . . . . . . . . . .S17

Wallenfels, J. . . . . . . . . . . . . . . . . . . .P25

Wang, B. . . . . . . . . . . . . . . . . . . . . . . .P3

Wang, S. . . . . . . . . . . . . . . . . . . .P48, S14

Ward, B. . . . . . . . . . . . . . . . . . . . . . . .P49

Wasawo, D. . . . . . . . . . . . . . . . . . . . .P45

Wasserman, S. . . . . . . . . . . . . . . . . . .S11

Watson, W. . . . . . . . . . . . . . . . . . . . . . .P4

Webster, D. . . . . . . . . . . . . . . . . . . . .S19

Wedege, E. . . . . . . . . . . . . . . . . . . . . .P51

Wei, X. . . . . . . . . . . . . . . . . . . . . . . . .P46

Author Presentation Number

Welte, T. . . . . . . . . . . . . . . . . . . . . . . . .S4

Wen, Y. . . . . . . . . . . . . . . . . . . . . .S3, P1

Wesselingh, S. . . . . . . . . . . . . . . . . . .S19

Whalen, R. . . . . . . . . . . . . . . . . . . . . . .S6

Whitsitt, P. . . . . . . . . . . . . . . . . . . . . . .S2

Whittle, H. . . . . . . . . . . . . . . . . . . . . .P8

Wiegert, N. . . . . . . . . . . . . . . . . .P9, P10

Wonderling, L. . . . . . . . . . . . . . .P42, P66

Woods, A. . . . . . . . . . . . . . . . . . . . . .P46

Wright, J. . . . . . . . . . . . . . . . . . . . . . .P72

Wu, Y. . . . . . . . . . . . . . . . . . . . . . . . .S17

Xu, D. . . . . . . . . . . . . . . . . . . . . .S3, P24

Xu, L. . . . . . . . . . . . . . . . . . . . . . . . . . .S6

Yamaguchi, K. . . . . . . . . . . . . . . . . . .P35

Yao, X. . . . . . . . . . . . . . . . . . . . . . . . . .P1

Yasui, F. . . . . . . . . . . . . . . . . . . . . . . .P65

Yuan, Z. . . . . . . . . . . . . . . . . . . . . . . . .S3

Zadeh Vakili, A. . . . . . . . . . . . . . . . . .P67

Zahedifard, F. . . . . . . . . . . . . . . . . . . .P67

Zang, G. . . . . . . . . . . . . . . . . . . . . . .P46

Zhan, B. . . . . . . . . . . . . . . . . . . . . . . . .S7

Zhang, S. . . . . . . . . . . . . . . . . . . . . . .P40

Zhao, K. . . . . . . . . . . . . . . . . . . . . . . . .S3

Zhao, P. . . . . . . . . . . . . . . . . . . . . . . .P23

Zheng, W. . . . . . . . . . . . . . . . . . . . . .P17

Zelman, M. . . . . . . . . . . . . . . . . . . . .S18

Zorzopulos, J. . . . . . . . . . . . . . . . . . . .P5

AUTHOR INDEX

98

Eighth Annual Conference

Page 99: On-Site Progr

on Vaccine ResearchAUTHOR INDEX

99

DISCLOSURE INDEX

As a sponsor accredited by the Accreditation Council of Continuing Medical Education (ACCME) the NationalFoundation for Infectious Diseases must insure balance, independence, objectivity, and scientific rigor in all itsindividually sponsored or jointly sponsored educational activities. All faculty participating in a sponsored activity and allScientific Program Committee Members are expected to disclose to the activity audience: (1) any significant financialinterest or other relationship (a) with the manufacturer(s) of any commercial product(s) and/or provider(s) of commercialservices discussed in an educational presentation and and/or (b) with any commercial supporters of the activity.(Significant financial interest or other relationship can include such things as grants or research support, employee,consultant, major stock holder, member of speakers bureau, etc.); and (2) any intention to discuss off-label uses ofregulated substances or devices.

The intent of this disclosure is not to prevent a speaker, nor a Scientific Program Committee member, with a significantfinancial or other relationship from making a presentation, or assisting in conference organization, but rather to providelisteners with information on which they can make their own judgments. It remains for the audience to determinewhether the speaker's interests or relationships may influence the presentation with regard to exposition or conclusion.

The following Presenters have no relationships to disclose:

C. Azimpour TabriziB. BekanM. BergA. BiragynM. BottazziS. BystrickyR. CauseyJ. ChoiR. DalloulA. de RouxN. DhimanA. DickoL. DiMiceliW. DowlingM. EbrahimiM. Gahan

S. SowD. SteeleB. TalbotM. TapiaD. TropelA. VivekanandhanL. YuexiH. Vu khacS. VratiK. WaliaJ. WallenfelsB. WangY. WenR. WhalenX. YaoD. Xu

V. GilcaE. HartwellM. HayneyA. HonjoA. HonkoR. HuE. HwangB. ImoukhuedeS. JittapalapongT. KaihuaP. KalirajL. KamauC. KangA. KarimM. KitabatakeR. Kofler

F. QadriS. RafatiR. RanalloG. ReubelJ. RobbinsE. RosenqvistR. SadriS. SahaP. SakhataskyyP. SalotraC. SauderP. SethR. ShiI. SilveriaJ. SimonD. Skrastina

C. LaJeunesseJ. LangleyS. LawrenceD. LibratyJ. LinS. LuS. McNeilM. MendyA. MontanerV. MuzioA. OlagunM. OsorioS. Owusu-AgyeiI. OvsyannikovaG. PlumbD. Promkhatkaew

L. Babiuk R. RabinovichR. Duma P. McInnesH. GoldingP. Nara

The following Program Committee Members have no relationship to disclose:

The remaining Presenters have disclosed the following:

H. Robinson C. Schmaljohn

Presenter Company Relationship*

J. Alexander Epimmune A,CR. Atmar Corixa, Vaxgen EJ. Crowe MedImmune A

sanofi aventis, MedImmune, Vaxgen BMedImmune, Morphotek, Symphogen, Syngenta, VaxGen, Vaxin Esanofi pasteur G

S. Cyr ID Biomedical AT. dela Cruz Dynavax Technologies A,CG. Del Giudice Chiron CP. Giardina Wyeth A,C

(continued)

Page 100: On-Site Progr

AUTHOR INDEX

100

AUTHOR INDEXAUTHOR INDEXDISCLOSURE INDEX

A. Haller GlobeImmune A,CS. Halperin GlaxoSmithKline BS. Harris Wyeth CR. Hepler Merck A,CD. Heppner GlaxoSmithKline BA. Hu Wyeth A,CA. Lees Biosynexus, Inc. A,C,GC. Li New Century Pharmaceuticals, Inc. A,CP. Nagappan DynPort Vaccine BP. Rangarajan Indian Immunologicals Ltd BD. Scheifele Wyeth BG. Siber Wyeth A,CD. Skowronski sanofi Pasteur BL. Wonderling Syntiron C

The remaining Program Committee Members disclosed the following:

Presenter Company Relationship*

C. Baker Chiron ED. Griffin Chiron, Vical Collaboration

MedImmune FElan, Wyeth DSMB

R. Lambert Chiron, GlaxoSmithKline, Ortho McNeil EM. Levine VaxGen A

GlaxoSmithKline, ID Medical Group BS. Plotkin sanofi Pasteur A,C

Dynport FR. Rappuoli Chiron CS. Rehm sanofi aventis, Wyeth, Pfizer, Cubist, Glaxo E

Pfizer ACubist B

A. Shaw Merck & Company CG. Siber Wyeth Vaccines A,CB. Weniger VaxGen A

(Remaining Presenters continued)

*Please refer to the following relationship table

Label Relationship

A I have stocks, stock options, and/or bond holdings in this company B I have a research grant, stipend, and/or fellowship from this company C I am employed by this company, or it employs a member of my immediate family D I or a member of my immediate family own or is a partner in this company E I or a member of my immediate family receive consulting fees, honoraria, paid meeting

registration fees, paid travel, speaking fees, or other financial compensation from this companyF I or a member of my immediate family hold a nonrenumerative position of influence with this

company such as officer, board member, trustee, or public spokesperson.

G I or a member of my immediate hold a patent for and/or receive royalties from this company’s product

Eighth Annual Conference