Abstract Book - United Scientific Group

advertisement
A New Era in Vaccine Discovery
International Conference on
Vaccines
Research & Development
November 02 - 04, 2015
Baltimore, USA
Vaccines R&D-2015
Sponsors
c
Group
UNITED Scientifi
Index
About United Scientific Group
4
USG Editors Association
4
USGEA Founding Members
5
Sponsors6
Keynote Presentations
10
Symposium15
Key Concepts
16
Featured Presentations
17
Speaker Sessions
Track-A20
Track-B29
Track-C43
Track-D56
Track-E66
Poster Session
77
About United Scientific Group
United Scientific Group (USG), an expert-driven initiative led by the editors association and the advisory
board which includes academicians, researchers, and industry leaders across various fields of research.
USG provides broad range of services in the fields of science and technology including publishing,
conducting world class scientific events, and holding highly interactive and proficient world forums.
USG Editors Association
Founding President
Founding Vice-President
Kenneth Blum, PhD, DHL
University of Florida, USA
Sayon Roy, PhD, FARVO
Boston University, USA
The scientific industry involved in networking, organizing meetings and publishing scholarly journals is increasing constantly
in order to meet the ever changing demands of emerging new concepts and subjects in different fields of science. Rigorous,
meticulous policies and guidelines are essential to maintain the highest standards of scientific excellence. United Scientific
Group (USG) is fortunate to have the United Scientific Group Editors Association (USGEA) that serves this role. USGEA
is an association of United Scientific Group Journals editors with diverse backgrounds and professional experience, who seek
to foster cooperation and communication among editors, improve editorial standards, promotes the concept of self-criticism,
self-regulation in scholarly publishing, and encourage research on the editorial principles and practices of publishing.
United Scientific Group Editors Association Meeting
July 07, 2015 • San Francisco, USA
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
4
Founding Members
Ahmad Salehi, MD, PhD
Stanford Medical School
USA
Jin Jun Luo, MD, PhD
Temple University
USA
Rajendra Badgaiyan, MD Claudio Ando Nicolini, PhD
University of Minnesota
USA
Fondazione ELBA Nicolini
Italy
Dawei Li, PhD
Shanghai Jiao Tong
University, China
Mun Yhung Jung, PhD
Reza Hakkak, PhD
University of Arkansas for
Medical Sciences, USA
Woosuk University
South Korea
Anil K Mandal, MB, BS
Mandal Diabetes Research
Foundation, USA
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
5
Sanofi Pasteur
w w w . s a n o fi p a s t e u r . c o m
Partnership opportunities with Sano Pasteur
Sano Pasteur, the vaccines division of Sano , is seeking partners who will share the company’s pursuit of innovation
and drive for excellence. Our diverse culture and geography allow us to collaborate effectively with a broad array of
partners around the world.
W
e are not only at the forefront of
conquering newly targeted infectious
diseases, but also leading the way in
expanding immunization across all age groups,
including adolescents and the elderly. This
leadership has translated into outstanding success
in the industry, and our company has experienced
a recent period of unprecedented growth.
Sano Pasteur is interested in partners who
will share in the pursuit of innovation and our
drive for excellence, while becoming a part of
our market success story. “We welcome the
opportunity to evaluate technologies related
Sanofi Pasteur recently signed an agreement
with SK Chemical Co. of South Korea to
co-develop an innovative pneumococcal
conjugate vaccine
Sano Pasteur is interested in potential partnering opportunities in the eld of active and
passive human immunization, as well as technologies supporting product development and
industrial performance, including:
Vaccines, monoclonal antibodies and supporting technologies for prevention and treatment
of infectious diseases
New ways to administer vaccines
Novel antigens and methods for antigen
discovery and characterization
Carrier proteins and proteinpolysaccharide conjugation methods or
Vaccine vectors suitable for nasal or oral
alternative technologies
use
Agents to enhance vaccine immune responses
Adjuvants and immunomodulators
Biological and immunological studies
to further characterize adjuvants and
Vaccine vectors and delivery systems
immunomodulators
intended to enhance or modify immune
responses
Characterization and assay of immune responses and disease markers
Animal models of human diseases
In vitro models of human tissues,
including the immune system
Biological markers for evaluating the
Epidemiological studies relevant to the
use of vaccines and immunotherapeutics
interventions
Tools for improving vaccine and monoclonal antibody research, development and production
Development and application of new
technologies in the areas of genomics
and proteomics
Process automation
Prokaryotic or eukaryotic cell lines for
Preservatives and stabilizers
antigen production
Bioinformatics techniques for modeling,
Fermentor and bioreactor technology
data handling and analysis
Disposable systems
Anti-counterfeiting technology
Online testing
to the development and production of human
vaccines, both prophylactic and therapeutic,
including vaccines for infectious and chronic
diseases,” said Roman Chicz, PhD.
Sano Pasteur has a strong commitment to
Research and Development partnerships with
major universities, research institutes, government bodies, biotechnology companies and contract research organizations. The company's collaborations cover virtually all aspects of vaccine
development, including early-stage research.
Examples of current partnerships and technology investments include: vaccines for dengue
fever, Japanese encephalitis, S. pneumoniae,
and M. tuberculosis; pediatric combination
vaccines; rabies monoclonal antibodies;
large-scale, cell culture-based virus production;
adjuvants and immunomodulators; conjugate
vaccine production; and vaccine delivery
systems.
A company partnering with Sano Pasteur
interacts with a multidisciplinary team that has
years of experience in working to ensure that
partnerships are executed succesfully and nurtured for the mutual bene t of all parties.
This approach utilizes the value-added Sano
Pasteur alliance management capability, which
focuses on the relationship by facilitating
open communication, trust, understanding and
clear expectations across the project lifespan.
Combined with the technical competency of the
alliance, this balance provides a well rounded
environment for your technology to ourish.
CONTACT DETAILS
Sano Pasteur welcomes information about new
partnership opportunities. Each opportunity is
carefully evaluated and reviewed by our dedicated
team.
Please contact:
Roman Chicz, PhD
Head, External Research and Development
Tel: +1 617 866 4562
Email: roman.chicz@sanofipasteur.com
Jean-Marc Renard, MS, MBA
Vice President, Corporate Development
Tel: +33 4 3737 3480
Email: jean-marc.renard@sano pasteur.com
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
6
InDevR, Inc., is a leader in providing progressive analytical technologies that enhance and accelerate
vaccine characterization and production. The company is also committed to developing nextgeneration diagnostic products that will enable unprecedented tracking of seasonal and emerging
influenza viruses around the globe.
InDevR provides the Cypher One™ product for automated hemagglutination analysis and the
VaxArray™ Influenza potency assay for rapid quantification of hemagglutinin protein, the key
ingredient in flu vaccines. The company also offers a range of other microarray-related products,
including the Vidia™ Microarray Imaging System. New tools in advanced stages of R&D include the
FluChip-8G™, a multiplexed influenza diagnostic providing deep genotyping influenza viruses in a
simple cost-effective format. All of our products support vaccine developers and manufactures by
providing faster turnaround time and improved results at an affordable price.
In 2005, InDevR purchased a majority ownership share of 2B Technologies, a leader in the field of
analytical instruments for atmospheric and environmental measurements. 2B Technologies has
produced and sold more than 2,000 ozone monitors and related instruments. The two companies
share a facility to promote collaboration for high quality research, manufacturing, sales, and customer
service.
In 2012, InDevR raised venture capital to create a new company, ViroCyt, Inc. ViroCyt is focused
on expanding the commercial market for the Virus Counter technology originally developed and
marketed by InDevR. The Virus Counter provides rapid quantification of viruses resulting in faster,
more accurate decision-making in settings that utilize viruses to create lifesaving products, such as
viral vaccine production, protein expression, antiviral drug development, and viral diagnostics. For
more information visit ViroCyt.com.
InDevR continues to grow its network of partners in order to expand into new markets, develop
innovative technologies and applications, and collaborate with scientists for the evaluation of new
products. Due to its dedication to addressing public health issues identified as high priority for our
Nation, InDevR has received the majority of its non-dilutive funding from agencies such as the
National Institute of Allergy and Infectious Diseases and BARDA. Founded in 2003, InDevR is a
privately held, women-owned business based in Boulder, Colorado.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
7
Cellular Technology Limited (CTL) proudly serves the “who’s who” in the biomedical field with
over 20 years of excellence in three industry segments which encompass complete solutions to
advance the frontiers of biomedical research: instrumention, reagents, and laboratory services.
Instrumention:
Scientifically-validated, intelligent analyzers and software that provide accurate, transparent, and
objective results.
Reagents:
Cryopreserved PBMC, Standardized ELISPOT Kits, Serum-free Media, and CEF Peptide Pools that
have pre-defined reactivity for the CEF peptide pools and for the individual peptides of the pool as
well as HLA class I and II high-resolution haplotype characterization.
Laboratory Services:
High-throughput immune monitoring with an emphasis on T cells. Our expertise in conducting
ELISPOT assays in a GLP-compliant manner is unparalleled. This permits identification of antigenspecific T cell effector classes secreting different cytokines or exerting cytotoxic functions. ELISPOT
is also uniquely suited for its clients in various test systems including, but not limited to, human,
monkey, mouse, rat, rabbit, and pig. CTL’s Specimen Management department is set up to serve your
needs through the entire process from loss-free shipment and cryopreservation to batch testing of
the PBMC.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
8
Aeras is a nonprofit, global biotech organization developing new tuberculosis vaccines for the world,
in partnership with other biotech, pharmaceutical, and academic organizations.
Aeras has an international staff of more than 100 people, with offices in the U.S., Africa, and Asia
representing a critical mass of expertise in TB and vaccine development. With our extensive network
of collaborators and funders, we and our partners effectively manage and prioritize the many
vaccine candidates to accelerate progress in TB vaccine research and development. As a nonprofit, we
champion TB vaccines based on global need and science and work to ensure that the final product is
affordable and accessible to those who need it most.
Serving as a critical translational bridge from the bench to the field Aeras has sponsored and conducted
over 25 clinical vaccine trials enrolling thousands of subjects, and is a key partner in 6 active clinical
development programs.
LumaCyte is a biotechnology instrumentation company headquartered in Charlottesville, VA.
LumaCyte’s novel single cell analysis and sorting technology has tremendous application for vaccine
development in both R&D and manufacturing including viral infectivity assays. The ability to
automate data collection on the viral infectivity of cells while improving results, reducing cost and
time is invaluable for developing and producing vaccines.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
9
Keynote Session
Cancer Vaccines: Translation from Mice to Human Clinical Trials
Jay A. Berzofsky
Vaccine Branch, Center for Cancer Research, National Cancer
Institute, National Institutes of Health, USA
Abstract
Both the humoral and cellular immune systems can be elicited
by cancer vaccines to target cancer, but antibodies can detect
only surface antigens on tumor cells whereas T cells can
detect fragments of any protein made in the cell, presented
by HLA molecules. I will describe examples of both types of
vaccines that we have translated from preclinical discovery
in mice to human clinical trials. For T cell recognition, many
cancer epitopes may be of too low affinity to induce a T cell response, so we developed an epitope
enhancement approach to increase affinity for HLA molecules. TARP is an antigen discovered by Ira
Pastan’s lab expressed in about 95% of prostate cancers of all types and stages as well as 50% of breast
cancers. We mapped epitopes presented by HLA-A2, the most common human class I HLA molecule,
and then modified the sequence of one with intermediate affinity to make it more immunogenic
(“epitope enhancement”). The enhanced epitope induced T cells in HLA-A2 transgenic mice reactive
with the natural sequence more effectively than the natural sequence itself, and induced human T
cells that killed human tumor cells expressing TARP and HLA-A2. Two peptides were translated into
a clinical trial in stage D0 prostate cancer, the stage in which the primary tumor has been removed
but a rising PSA indicates microscopic recurrence. In this setting, in which no tumor can be measured
radiographically, the rate of change of PSA has been validated as a measure of clinical outcome. In
patients receiving the TARP peptide vaccine, nearly three-fourths had a statistically significantly
decreased PSA slope at 6 and 12 months, and the median tumor growth rate constant was reduced
in half. A follow-up randomized, placebo-controlled phase II trial is underway. The vaccine targeting
antibodies is a recombinant adenovirus expressing the extracellular and transmembrane domains of
HER2. In mice, such a vaccine cured virtually 100% of large established murine mammary carcinomas,
and cured mice of large established lung metastases. In mice, the therapeutic effect was independent
of CD8 T cells but completely dependent on antibodies. However, the mechanism was different from
that of trastuzumab in that it was not FcR-dependent. A human version was used to transduce
autologous dendritic cells to treat patients with any type of HER2-positive tumor not previously
treated with HER2-directed therapy. Early results show several objective clinical responses. Thus,
cancer vaccines eliciting either T cells or antibodies have great promise to treat cancer and may be
amplified by synergy with other agents such as checkpoint inhibitors.
Biography
Dr. Jay A. Berzofsky, National Cancer Institute, USA, Chief of the Vaccine Branch, CCR, NCI, former
president of the American Society for Clinical Investigation and former Chair of the Medical Sciences
Section of AAAS, who won the NIH Director’s Award and NCI Merit Award in 2008 and a Merit
Award in 2011.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
10
Ebola Vaccine in 12 months, Global Village, and the Need for Speed
Robert W. Malone
RW Malone MD, LLC, USA and Harvard Medical School Global
Clinical Scholars Research Training Program, USA
Abstract
The story of the amazingly rapid advancement of the PHAC/
Health Canada/NewLink/Merck rVSV ZEBOV vaccine to clinical
proof of concept/efficacy within 12 months demonstrates
what we, as a community of vaccine developers, are capable of.
But it is also a rich source of “lessons learned” and cautionary
warnings concerning current world health and the infectious
disease threat response capability. It took a global village to
enable this achievement. But how many times will we, the
community of public health specialists and vaccine developers, be able to protect the world from
this type of infectious disease outbreak when each time we end up having to cobble together an adhoc response? A more comprehensive and sustainable global surveillance, detection, and diagnosis
capability, integrated into a rapid response vaccine development system is required. Advances in
ID surveillance, computational immunology, biological polymer delivery, conjugation chemistry,
information processing/data fusion (including social media tools), disease tracking and predictive
modeling, regulatory science, innovative clinical trial designs and public health politics must be
aligned to enable this goal. The amazing global response to the Ebola threat shows that we can do
this. Now is the time to build an integrated system in anticipation of the next outbreak, not after the
inevitable has yet again occurred.
Biography
One of three original inventors of “naked” polynucleotide delivery and original inventor of
polynucleotide (RNA, DNA) vaccine technology (1989), with seminal contributions in in-vivo nonviral delivery (cationic lipids, pulsed electrical fields), Dr. Malone received his BS (Biochemistry)
from UC Davis, MS (Biology) from UC San Diego while training under Inder Verma (Salk Institute
Molecular Biology and Virology Laboratories), MD from Northwestern University Feinberg School
of Medicine, and completed his internship and post graduate fellowships at UC Davis (Pathology).
He has served as an academic (to Associate Professor) at UC Davis, U Maryland, and USUHS, played
a key role in Biotechnology start-ups (including Vical, Inovio), Non-Governmental Organizations
(Aeras Global TB Vaccine Foundation), large government contractors (DynCorp/Dynport Vaccine
Company, TASC), Pharma (Solvay Biologicals), and currently operates a vaccine and biologics-focused
consulting company serving a wide range of clients.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
11
Regulatory Perspective on the Evaluation of New Vaccine Technologies
Norman W. Baylor
Biologics Consulting Group, Inc., USA
Abstract
The pathway from discovery to marketing of a new vaccine can
be long, complex and costly. Regulatory procedures impact
all stages of the vaccine development process; moreover, a
detailed safety review during research and development stages,
regulatory approval, and legal registration are necessary
stages of product development. Successful development
of a marketable product requires that vaccine developers,
especially those using new technologies, are not only aware
of, but have an understanding of regulatory obligations and
opportunities to expedite the development of safe and effective products in a cost effective manner.
In order to minimize the risks and costs involved in the vaccine product development process a wellfocused regulatory strategy that can be presented to regulatory authorities should be developed. It is
imperative that clear planning is done during the early stages of product development, with regular
assessment of chemistry, manufacturing and control (CMC), non-clinical and clinical data as it is
generated. Further, a comprehensive understanding of the FDA regulatory process, regulations and
policies is essential when developing an effective product life cycle management strategy, seeking
product approval, and assuring that products are marketed in compliance with federal requirements.
The aim of this presentation is to provide an overview of the FDA regulatory product approval process
and discuss considerations for successfully developing a regulatory strategy that leads to product
marketing authorization from national regulatory authorities.
Biography
Dr. Norman W. Baylor is currently the President and CEO of Biologics Consulting Group, Inc.,
headquartered in Alexandria, Virginia. Prior to joining BCG, Inc., Dr. Baylor was the Director of
the Office of Vaccines Research and Review in the US Food and Drug Administration’s Center for
Biologics Evaluation and Research. During his 20 year tenure with FDA, he evaluated and facilitated
the development and licensure of numerous new vaccines such as acellular pertussis, varicella,
pneumococcal conjugate, human papillomavirus (HPV), influenza (seasonal and pandemic), Zoster
and all of the new generation combination vaccines. Dr. Baylor served as FDA’s liaison to the Centers for
Disease Control’s Advisory Committee on Immunization Practices, the US Department of Health and
Human Services National Vaccine Advisory Committee, and the Advisory Commission on Childhood
Vaccines. Dr. Baylor continues to serve as an expert advisor to the World Health Organization on
global vaccine initiatives. He is also a member of the NIH NIAID Advisory Council, and a member of
PATH Malaria Vaccine Initiative’s Vaccine Science Portfolio Advisory Council.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
12
Vaccines Against Invasive Salmonella Infections
Myron M. Levine
Center for Vaccine Development, University of Maryland
School of Medicine, USA
Abstract
Certain serovars of Salmonella can cause invasive disease in
humans. The human-restricted Salmonella enterica serovar
Typhi (S. Typhi), S. Paratyphi A and S. Parayphi B cause enteric
fever. Clones of these serovars carrying multiple antibiotic
resistance have emerged and disseminated, particularly in
South and SE Asia, thereby making it increasingly difficult
to treat these invasive infections. S. Paratyphi C and closely
related S. Choleraesuis cause septicemia and focal metastatic
infections, albeit uncommonly. More recently, in sub-Saharan Africa variants of S. Typhimurium
and S. Enteritidis have emerged as major agents associated with invasive infections (bacteremia,
septicemia, meningitis, septic arthritis, etc.) in infants, toddlers and young children accompanied by
distressingly high case fatality. The rationale for vaccines against some of these serovars, the target
populations and the status of various licensed vaccines and vaccine candidates will be discussed.
Biography
Dr. Myron M. Levine is the Bessie & Simon Grollman Distinguished Professor at the University of
Maryland School of Medicine, Associate Dean for Global Health, Vaccinology and Infectious Diseases,
and the Founder/Former Director of the Center for Vaccine Development (1974-2014). He has
extensive experience in design and evaluation of vaccines to prevent bacterial enteric infections, and
has made substantial contributions in basic vaccinology, bacterial pathogenesis, clinical research,
field epidemiology and public health. He has published over 591 peer reviewed journal articles, is an
inventor/co-inventor on numerous patents and is Senior Editor of New Generation Vaccines, 4th ed.,
a textbook of research vaccinology.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
13
Immune Protection Inducing Synthetic Protein Structures (IMPIPS): The New Vaccines
Manuel Elkin Patarroyo M.
Fundacion Instituto de Inmunologia de Colombia –FIDIC;
Bogota D.C., Colombia
Abstract
Physicochemical rules must be followed for the development
of fully-protective, chemically-synthesized vaccines. We have
identified most of these principles or rules for the prototype
disease (malaria) for developing a panel of functionallyrelevant, specifically modified, high activity binding peptides
(mHABPs) attaching to host cells, thereby inducing sterile
protective immunity in the experimental Aotus monkey
model. These principles include a polypropylene II - left
handed-like (PPIIL) 26.5 ± 3.5 A° distance between amino acids fitting into Pockets 1 to 9 of Class II
molecules’ peptide binding region (PBR), an appropriate charge and volume of residues fitting into
these molecules’ Pockets 1, 4, 6 and 9, as well as specific rotamer orientation of residues contacting
the T-cell receptor. These principles or rules have led to developing long-lasting, protection-inducing
multi-epitope, multistage, minimal subunit-based, chemically-synthesized vaccines or effective
immune protection-inducing, synthetic protein structures.
Biography
Professor Manuel Elkin Patarroyo M. (1946) received his M.D. degree from the Universidad Nacional
de Colombia in 1971 where he is Full Professor of Molecular Pathology. He conducted post-doctoral
studies at the Yale University and at Rockefeller University, U.S.A. with Professor Henry Kunkel, and
on Tumor Immunology at Karolinska Institutet, Sweden, with Professor George Klein. In 1976, he
founded the Instituto de Inmunología at the San Juan de Dios Hospital, devoted to the development of
chemically synthesized vaccines, among them the antimalarial vaccine, with the advice of Professors
Bruce Merrifield (Rockefeller University), Professor Richard Lerner (Scripps Research Institute) and
Professor David Andreu (Pompeu Fabra University).
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
14
Symposium by Sanaria, USA
PfSPZ-Based Malaria Vaccines from Concept to Licensure
The development of an effective malaria vaccine has proven elusive, despite decades of effort. The most advanced
candidate, RTS,S/AS01B is being considered for licensure in Africa, but provides only partial protection against
clinical disease and little or no lasting protection against infection. Moreover, intensive use of currently available
tools for malaria control, including insecticide-impregnated bed nets, indoor residual spraying of insecticides,
intermittent treatment of vulnerable populations such as infants and pregnant women, and rapid case finding
and treatment, have achieved only modest reductions in malaria-related morbidity in endemic areas, and are
insufficient to halt transmission. A highly effective vaccine is urgently needed. In this setting, Sanaria Inc.
has developed the world's first whole sporozoite (SPZ)-based malaria vaccines for parenteral injection, and
demonstrated 100% protection against the vaccine-strain Plasmodium falciparum (Pf) parasite by controlled
human malaria infection (CHMI), and the highest protection ever recorded against infection in the field. Eight
trials of two related PfSPZ vaccine candidates are completed or ongoing, and 11 more are scheduled to begin
in 9 countries in the next few months. These studies, designed to optimize immunization regimens for these
vaccines, aim for long-lasting (e.g., 6 month) high-level protection against heterologous CHMI and against
naturally acquired infection in the field. Manufactured according to GMPs, extremely safe and well tolerated,
causing essentially no adverse events at current doses, and suitable for transport to remote locations, these
vaccines are poised to fill the gap in the malaria control toolkit, and enable regional malaria elimination
through mass administration campaigns. In this symposium, Sanaria staff will describe the fascinating story
of the development of these award-winning vaccine candidates, including manufacturing, cryostabilization,
cold chain distribution, regulatory strategies, clinical development, immunological correlates of protection,
and our international partnerships and strategy for licensure to prevent malaria in travelers and in persons
living in endemic areas, and eventually to achieve regional malaria elimination and worldwide eradication.
1. Introduction: Stephen L. Hoffman, MD, Chief Executive Officer and Chief Scientific Officer
2. Challenges and Successes in Developing a New GMP Manufacturing Process: Aseptic, Purified,
Cryopreserved Malaria Sporozoites from Mosquitoes. B. Kim Lee Sim, PhD, Executive Vice President for
Process Development and Manufacturing
3. Creating a New, Improved, Cost-Effective Distribution System for a Live, Whole Eukaryotic Cell
Vaccine: Liquid Nitrogen Vapor Phase Storage for Human Vaccines. Eric R. James, PhD, Head, Vaccine
Stabilization and Logistics
4. The Road Less Traveled: the Pathway to Licensure of a Malaria Vaccine for Travelers and
Elimination Campaigns, Regulatory Obstacles and Opportunities Encountered on the Way: Tooba
Murshedkar, MS, Associate Director Regulatory Affairs
5. Clinical Development of Plasmodium falciparum Sporozoite (PfSPZ)-Based Products: A Multistage
Clinical Development Plan spanning 15 sites in the USA and 10 Countries in Africa and Europe. Thomas L
Richie, MD PhD, Chief Medical Officer
6. Using Immunology and Systems Biology to Optimize PfSPZ-based Vaccine Regimens: Sumana
Chakravarty, PhD, Senior Director, Sporozoite Extraction, Immunology and Model Systems
7. A Collaborative Approach to Success: The International PfSPZ Consortium (I-PfSPZ-C). Peter F.
Billingsley, PhD, Vice President International Projects and Strategy
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
15
Key Concepts
• Cancer Vaccines
• Clinical Trials
• Novel Vaccines
• Vaccines Discovery, Development & Formulation
• HIV/AIDS Vaccines
• Influenza Vaccines/Virus
• HPV Vaccines & Ebola Outbreak
• Basic Vaccinology
• Passive Vaccines-Therapeutic Antibodies Against Infectious Diseases
• Emerging Infectious Diseases
• Therapeutic Vaccination for Auto Immune Diseases
• Immunoinformatics
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
16
Featured Presentations
Nanoemulsion Delivery/Adjuvant for Eliciting Mucosal Immunity a Key in Protection
against Respiratory and Sexually Transmitted Infections
Ali I. Fattom
Nano Bio, USA
Abstract
The majority of pathogens colonize mucosal surfaces and invade the body via these surfaces. Almost all vaccines available
today are administered parenterally: either intramuscularly or intradermally. Mucosal immunity may prove to be useful
in blocking invasion and preventing colonization and subsequently transmission. Considering that most infections
have a mucosal port of entry, development of safe and efficacious mucosal vaccines is a major unmet medical need.
Mucosal immunity have been kept out of the spotlight due to lack of suitable delivery and adjuvant technologies. We
developed an oil-in-water nanoemulsion that showed efficiency in delivering antigens intranasal and inducing immunity
against infections including prevention of viral and bacterial shedding. Safety, compatibility, and immunogenicity of
this technology was demonstrated with multiple antigens in various animal models and was also shown to be safe and
immunogenic in human clinical trials. Data will be presented to demonstrate applicability of this approach to respiratory
(RSV) and sexually transmitted (HSV2) infections.
Histologic Regression of Cervical Intraepithelial Neoplasia (CIN) after Treatment with
VGX-3100, Therapeutic HPV16/18 DNA Vaccine
M. Bagarazzi1*, C. Trimble2, M. P. Morrow1, X. Shen1, M. Dallas1, J. Boyer3, J. Yan1, K. Kraynyak1, A. Sylvester1,
M.Giffear1, K. Marcozzi-Pierce1, D.Shah1, D. Weiner3, and the HPV-003 protocol team
Inovio Pharmaceuticals, USA
Johns Hopkins School of Medicine, USA
3
University of Pennsylvania Perelman School of Medicine, USA
1
2
Abstract
Objective: Evaluate the histopathologic response to three 6 mg doses of VGX-3100 administered by IM injection followed
by electroporation (EP) with Inovio’s CELLECTRA-5P in adult females with biopsy-proven HPV16 or 18 associated
CIN2/3.
Methods: Histologic regression of high grade cervical intraepithelial neoplasia (CIN2/3) was evaluated in a randomized,
placebo-controlled, double-blind phase 2b study of HPV16/18 E6/E7 DNA vaccine, VGX-3100 followed by EP with
Inovio’s CELLECTRA®2000 device.
Results: The study met its primary efficacy endpoint; the percentage of patients who had regression of CIN2/3 to CIN1
or no disease at 6 months post third dose was significantly higher in the VGX-3100 group compared to placebo (p=0.017).
In addition, the trial demonstrated the ability of VGX-3100 to clear HPV infection concurrent with regression of CIN
lesions. The study also explored cell mediated immune responses to VGX-3100 in blood samples taken prior to the first
vaccine dose and periodically thereafter. IFNγ ELISpot revealed higher responses in the VGX-3100 treated group than in
placebo, suggesting that VGX-3100 was able to robustly engage the cellular arm of the patients’ immune system.
Conclusion: The successful phase 2b results represent a significant milestone in the development of active
immunotherapies to treat cancer and infectious diseases and have the potential to provide physicians an important
alternative to surgery to treat CIN2/3. The results illustrate the highly promising potential of therapeutic immunization
with DNA followed by EP for the treatment of HPV-related precancerous cervical disease in women and present the
possibility of treating HPV-associated cervical, head and neck, and anogenital cancers.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
17
A Replication Defective Human Cytomegalovirus (CMV) Vaccine
Tong-Ming Fu
Merck Research Laboratories, USA
Abstract
Congenital CMV infection is one of the leading causes of birth defects; developing a prophylactic vaccine is an unmet
need for public health. Naturally acquired CMV immunity in women prior to conception is effective in preventing CMV
transmission to fetus during pregnancy; both humoral and cellular immunity to CMV are likely playing a role in blocking
the transmission. However, a live attenuate CMV vaccine is difficult to develop since the CMV is known to establish
persistent and life-long infection in host. Thus, to develop a safe CMV vaccine incapable of establishing productive and
persistent infection, we first restored expression of viral gH/gL/pUL128-131 pentameric gH complex, and then applied
a genetic/chemical switch to two viral proteins essential for replication. This design would allow regulation of viral
replication with a small synthetic molecule; the vaccine virus cannot replicate without the chemical in culture. Vaccine is
effective in induction of durable neutralizing Abs, mostly due to the restored pentameric gH complex, in rhesus monkeys.
Furthermore, it can induce balanced CD4 and CD8 T-cell responses. The vaccine is under Phase I evaluation.
Biography
Dr. Tong-Ming Fu obtained his medical degree at Peking University Health Science Center, formerly Beijing Medical
University, and Ph.D. at Pennsylvania State University, Hershey Medical Center. He joined Merck Research Laboratories
after his academic trainings in virology and immunology, and has been working at Merck Vaccines Research for 20-years.
He conducted research to support many novel vaccine programs, including Merck HIV-1 and influenza M2 peptideconjugate vaccines. He also studied immune mechanisms of novel adjuvants and immune modulations by PD-1 blocking
mAbs. He currently is the research lead for Merck CMV vaccine. He is a member of the council of 100 of VACCINE
editorial board, and has co-authored 60 manuscripts and 10 issued patents or applications.
The Use of the TLR 7/8 Agonist 3M-052 as a Potent Vaccine Adjuvant
Mark Tomai1*, John Vasilakos1, and Bali Pulendran2
3M/Drug Delivery Systems, USA
Emory Vaccine Center, Emory University, USA
1
2
Abstract
As the vaccine industry moves away from live or attenuated vaccines towards recombinant vaccines the need for novel
adjuvants is increasing. A major problem with most protein and peptide based vaccines is that they are poorly immunogenic
or elicit an inappropriate immune responses and therefore novel adjuvants that boost the immune responses will be
required to make these vaccines effective. Toll-like receptors (TLRs) agonists are one class of adjuvant that has shown
great promise for enhancing adaptive immunity. In fact, the TLR4 agonist monophosphoryl lipid A has been incorporated
into a number of recently approved vaccines. 3M is a leader in the discovery and development of TLR 7/8 agonists and
its lead compound 3M-052 is being developed as a vaccine adjuvant. The compound shows enhanced adjuvant activity
and less systemic toxicity than first generation TLR7/8 compounds like resiquimod. In most vaccine models, 3M-052
leads to enhanced antibody and T cell responses and skews immunity towards a Th1 response. Data has been generated
using 3M-052 with a variety of bacterial, viral and parasitic antigens, using a variety of different formulations in both
mouse and more recently primate models. Other potential advantages of 3M-052 over other TLR agonists is its simple
manufacturing process, its long term stability profile both as an API and when formulated, and its inherent potency in
animal models. Future studies with this adjuvant will focus on moving 3M-052 into the clinic.
Biography
Dr. Mark Tomai has a PhD in immunology from the University of Minnesota and has worked in the Pharmaceutical
Industry for 24 years. Most recently, he has been Head of Toll-like Receptor (TLR) Business Development at 3M Drug
Delivery Systems. He has been involved with numerous licensing deals involving TLR agonists. During his first 16 years he
held various positions in Drug Discovery and Development focused on TLR 7/8 agonists. He was the lead immunologist for
development of the TLR7 agonist, AldaraTM for the treatment of warts and skin cancer. He has authored 80+ publications
and is an inventor on 12 issued patents.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
18
Nanoparticle Vaccine: Antigen Protein Delivery to Dendritic Cells Using Amphiphilic Poly
(Amino Acid) Derivatives
Mitsuru Akashi1*, Fumiaki Shima1, Takami Akagi1, and Masanori Baba2
Graduate School of Frontier Biosciences, Osaka University, Japan
Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
1
2
Abstract
Vaccination to induce adaptive immune responses is expected for the protection and/or treatment against various
infectious diseases and cancers. Polymeric nanoparticles (NPs) with entrapped vaccine antigens, represent an exciting
approach to control the release of antigens and to optimize the desired immune response via selective targeting of the
antigen to antigen-presenting cells (APCs). Dendritic cells (DCs) are highly specialized APCs that can activate naive T
cells and hence initiate primary immune responses. Therefore, the delivery of antigens to DCs and the activation of
DCs are one of the most important issues in the development of effective vaccines. To design optimal vaccine carriers,
polymeric NPs have the advantage of being able to regulate their physicochemical properties, such as particle size, shape,
and polymer composition. We have designed a novel antigen delivery system with self-assembled amphiphilic polymeric
NPs. We selected poly (γ-glutamic acid) (γ-PGA) as the biodegradable hydrophilic polymer and hydrophobic amino acid
as the hydrophobic side chain. Hydrophobically-modified γ-PGA with L-phenylalanine (γ-PGA-Phe) formed NPs due to
its amphiphilic characteristics in water. The size of γ-PGA-Phe NPs was easily controlled from 30 to 300 nm by regulating
the aggregated number of γ-PGA-Phe copolymers. Antigen-encapsulated γ-PGA-Phe NPs were efficiently taken up by DCs
and activated them. Interestingly, γ-PGA-Phe NPs possessed the adjuvant activity, and could maturate DCs. Moreover,
immunization with antigen-encapsulated γ-PGA-Phe NPs dramatically enhanced the antigen-specific cellular immunity.
Our study clearly demonstrates that γ-PGA-Phe NPs can provide safe and efficient nanoparticle-based vaccine adjuvants.
Biography
Dr. Mitsuru Akashi is the Specially Appointed Professor of Graduate School of Frontier Biosciences, Osaka University. He
received his PhD degree in Engineering from Osaka University in 1978. He was a post-doc at NIH, Gerontology Research
Center (USA) and the University of Waterloo (Canada) in 1978-1980. He joined the Department of Applied Chemistry
and Chemical Engineering, Faculty of Engineering, Kagoshima University as an assistant professor in 1981. He was
promoted to associate professor in 1984 and a full professor in 1989. He was a full professor for the Department of
Applied Chemistry, Graduate School of Engineering, Osaka University in 2003-2015.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
19
Track-A
Speaker Sessions
Oncogenic Viruses and Tumor Associated Antigens Vaccines
Giulio Filippo Tarro
Foundation de Beaumont Bonelli for Cancer Research, Italy
Abstract
There are at least two antiviral cancer vaccines: 1) anti-HBV vaccine was found to prevent some liver tumors. 2) anti-HPV vaccine
is able to prevent most of the cervical cancers, the second cause of cancer death for woman in the world. Tumor associated
antigens (TAA) were studied in SV40 and Polyoma viruses that were patterns for human cancer. Cancer immunotherapy, or
the manipulation of the naturally occurring oncolytic immune reaction, is based on the observation that both in animals and
humans neoplastic cell antigens stimulate the onset of specific humoral and cellular antibodies. Antigens, from autologous and
homologous human cancer cells, were obtained by various purification procedures. Tumor Liberated Protein (TLP) is a protein
extracted from tumors and from cultured transformed cells, detectable in blood as well as in cancer tissue. It behaves as a
TAA of 50KD monomer, overexpressed in lung tumor and other epithelial adenocarcinomas. TLP is immunogenic in humans
as evidenced by serum antibodies. When TLP is extracted from a tumor, purified in the laboratory, and reintroduced into
the patient’s body, it boosts the immune system’s cancer responsive capabilities. The perspectives are that TLP may have the
potential to greatly improve the cure rate and/or serve as a cancer vaccine.
Biography
Dr. Giulio Tarro graduated from Medicine School, Naples University (1962). Research Associate, Division of Virology and
Cancer Research, Children’s Hospital (1965-1968), Assistant Professor of Research Pediatrics, College Medicine (1968-1969),
Cincinnati University, Ohio. Oncological Virology Professor, Naples University (1972-1985). Chief Division Virology (19732003), Head Department Diagnostic Laboratories, (2003-2006), D. Cotugno Hospital for Infectious Diseases, Naples; Emeritus,
2006 - present. Since 2007 Chairman Committee of Biotechnologies and Virus Sphere, World Academy Biomedical Technologies,
UNESCO, Adjunct Professor Department Biology, Temple University, College of Science and Technology, Philadelphia, recipient
of the Sbarro Health Research Organization lifetime achievement award (2010). President Foundation de Beaumont Bonelli for
Cancer Research.
The Provocative Issue of Tumor Genomic Heterogeneity in Immunotherapy
Michael G. Hanna
Vaccinogen, Inc., USA
Abstract
While it has always been presumed that neoplasia is a consequence of somatic cell mutations, only in the last few years has
the magnitude and diversity of these mutations been elucidated by modern DNA sequencing technology. Immunotherapy
is the premier biological approach to targeted therapy. Target therapies require targets. In this case the targets are tumor
specific or associated antigens, the proteins expressed from these somatic cell mutations. While the immunotherapeutic
approach to eliminating cancer was launched with the assumption that cancer cells were homogeneous, the recent genomic
understanding of tumor cells indicates that there is both inter- and intra-tumoral heterogeneity. This presentation will discuss
the consequences of this new knowledge of tumor cell biology to the immunotherapeutic approach to treating cancer. What
is more, this presentation will discuss the translational development of an active specific immunotherapeutic approach from
preclinical to beneficial clinical benefit.
Biography
Dr. Michael G. Hanna, Jr. received his PhD in experimental pathology and immunology from the University of Tennessee in
1964. He was on staff of the Oak Ridge National Laboratory, biology division from 1964-75. Dr. Hanna served during 1975–83
as Director of the National Cancer Institute, Frederick Cancer Research Center (MD, USA). He was Chief Operating Officer during
1984–94 of Organon Teknika/Biotechnology Research Institute and Senior Vice President of Organon Teknika Corporation, a
subsidiary of Akzo Nobel, The Netherlands. Dr. Hanna founded PerImmune Inc., for which he served as President and Chief
Executive Officer before it merged with Intracel Corp. in1998. He continued to work for Intracel Resources as Chief Scientific
Officer and Chairman. In 2007, Dr. Hanna founded Vaccinogen Inc., where he served as Chairman and CEO. Currently, Dr.
Hanna is Chairman Emeritus. Dr. Hanna’s research resulted in over 225 publications in international peer-reviewed journals
and book chapters, and he holds 10 patents related to immunotherapy. Dr. Hanna has been the recipient of numerous honors
and awards and has served on many editorial boards; he currently serves on two editorial boards.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
20
Utility of CpG Oligonucleotides as Adjuvants for Cancer Vaccines
Dennis M. Klinman
Cancer and Inflammation Program, National Cancer Institute, USA
Abstract
The ability of tumor-specific NK and cytotoxic T cells to eliminate cancers is hindered by the large number of immunosuppressive
cells present in the tumor microenvironment. Monocytic myeloid-derived suppressor cells constitute the majority of these
tumor infiltrating leukocytes and are key contributors to the immuno-suppressive milieu. Murine mMDSC express Toll-like
receptors (TLR) and respond to TLR stimulation by differentiating into tumoricidal macrophages. In vivo administration of the
TLR9 agonist CpG ODN (alone and in combination with other TLR agonists) slows/prevents tumor growth. This outcome is
linked to reduced immune suppression in the tumor microenvironment allowing for increased activity of tumoricidal T cells.
Human mMDSC also express TLRs and are induced to differentiate into tumoricidal macrophages when stimulated via relevant
Toll-like receptors.
Generation of Donor Specific Customized Anti-Tumor T Cells and Antigen Presenting Cells
for Personalized
Arvind Chhabra
University of Connecticut Health Center, USA
Abstract
Human pluripotent stem cells [hPS, i.e. human embryonic stem cells (hES) and induced pluripotent stem cells (iPS)] can be
used to derive a cell lineage of choice, under defined differentiation conditions. We will discuss findings from our ongoing
study on generating donor specific iPS lines, derivation of donor specific antigen presenting cells (APC) from these lines and
their functional characterization. In addition, T cell engineering approaches with transgenic T cell receptor (TCR) or chimeric
antigen receptor (CAR) have generated encouraging results in cancer patients, however, generation of customized anti-tumor T
cells with predefined functional characteristics could improve the clinical outcomes significantly. We have recently shown that
TCR engineering approach can not only be used to generate CD8+ customized anti-tumor cytolytic T lymphocytes (CTL), but
also to program human CD4 T cells to function as MHC class I restricted anti-tumor effectors that can simultaneously exhibit
a helper response as well as cytolytic effector response (Chhabra et al., JI, 2008 and Ray et al., J. Clin. Immunol., 2010). We
will share our recent findings on biology of MHC class I restricted CD4 T cells, such as their effector function profile, ability to
mitigate Treg-mediated immune suppression, and their susceptibility to undergo AICD. We will also discuss molecular pathways
involved in development of multifunctional effector profile of MHC class I restricted CD4 T cells, and strategies for generating
customized anti-tumor T cells with predefined functional profiles. Generation of Customized Anti-Tumor T Cells and antigen
presenting cells are highly desirable for developing patient specific personalized cancer immunotherapy. Therefore, our findings
bear significant translational implications for cancer immunotherapy field and beyond.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
21
A Novel HPV-16 Immunotherapeutic Vaccine Plus PD-1 Checkpoint Inhibition Results in
Induction of Heightened Cell Mediated Immunity and Anti-Tumor Responses
Frank R. Jones1*, Adrian E. Rice1, Yvette Latchman1, John H. Lee2, Joseph P, Balint1, and Elizabeth S. Gabitzsch1
Etubics Corporation, USA
Sanford Healthcare, USA
1
2
Abstract
We have reported on a novel adenovirus serotype 5 (Ad5) vector gene delivery platform (Ad5 [E1-, E2b-]), in which regions
of the early 1 (E1), early 2 (E2b), and early 3 (E3) genes have been deleted. The unique deletions in this platform result in
a dramatic decrease in late gene expression, leading to a marked reduction in host immune response to the vector. Human
papilloma virus 16 (HPV) E6 and E7 are tumor-associated antigens (TAA) expressed on a wide range of HPV induced human
tumors such as cervical, head & neck and anal.
We report here the development of a novel Ad5 [E1-, E2b-]-E6/E7 vaccine construct for immunotherapy of HPV induced cancers.
Our results show that this construct is capable of activation, as well as generation of antigen specific T cells in vaccinated
mice. We have investigated Ad5 [E1-, E2b-]-E6/E7 combined with programmed death-ligand 1 (PD-1) blockade to determine
if combined treatment could increase therapeutic effect as compared to the vaccine alone. Ad5 [E1-, E2b-]-E6/E7 as a single
agent induced HPV-E6/E7 cell-mediated immunity in treated tumor bearing mice. Immunotherapy alone using Ad5 [E1-, E2b]-E6/E7 resulted in clearance of small tumors and an overall survival benefit in mice with larger established tumors. When
immunotherapy was combined with immune checkpoint blockade, an increased level of anti-tumor activity against large tumors
was observed. Analysis of the tumor microenvironment in Ad5 [E1-, E2b-]-E6/E7 treated mice revealed elevated CD8+ tumor
infiltrating lymphocytes (TIL); however, we observed induction of suppressive mechanisms such as programmed death-ligand 1
(PD-L1) expression on tumor cells and an increase in PD-1+ TIL. When Ad5 [E1-, E2b-]-E6/E7 immunotherapy was combined
with anti-PD-1 antibody, we observed CD8+ TILs at the same level but a reduction in tumor PD-L1 expression on tumor cells
and reduced PD-1+ TILs providing a mechanism by which combination therapy favors a tumor clearance state and a rationale for
pairing antigen-specific vaccines with checkpoint inhibitors in future clinical trials.
Epstein - Barr virus in the Breast Cancer Tissue: Friend or Foe?
Emmanuel Drouet1*, Gina Marao2, Artur Paiva2, Mohammed Habib1, Patrice Morand1, and Carlos Freire de Oliveira3
Unit of Virus Host-Cell Interactions, Université de Grenoble-Alpes, France
Portuguese Institute for Blood and Transplantation, University Hospital, Coimbra, Portugal
3
Department of Gynecology, University Hospital, Coimbra, & Faculty of Medicine, University of Coimbra, Coimbra, Portugal
1
2
Abstract
During the past decade, insights have been gained regarding mechanisms underlying the dynamic interplay between immune
cells and tumor progression. Regarding the breast cancer (BC), various studies have reported detecting the Epstein-Barr virus
(EBV) in BC cases. Yet the results are unconvincing, and their interpretation has remained a matter of debate. We have now
presented prospective data on the effect of EBV infection combined with survival in patients enrolled in a prospective study
(Marrao et al. BMC Cancer 2014). We assessed 85 BC patients over an 87-month follow-up period to determine whether EBV
infection interacted with host cell components that modulate the evolution parameters of BC. Immunological studies were
performed on a series of 35 patients, involving IFN-γ and TNF-α intracellular immunostaining tests in peripheral NK and T
cells, in parallel with EBV signature. The patients who recovered from their disease were found to have a measurable EBV DNA
load, together with a high frequency of IFN-γ and TNF-α producing PBMCs (p = 0.04), which indicates the existence of a Th1type polarized immune response in both the tumor and its surrounding tissue. Our study represents the first ever report of
the positive impact of EBV on the clinical outcome of BC patients, regardless of tumor histology or treatment regimen. The
durability of this reaction led us to hypothesize that endogenous EBV infection could be considered a “friendly” setting by
means of a heightened innate immune activation. These new findings could offer clinicians attractive targets for anticancer
immune-based therapies.
Biography
Dr. Emmanuel Drouet is currently professor of Virology at Grenoble University (Faculty of Pharmacy) and focused on the
interplay between virus and immunity. He received his PhD in Clinical Virology from the Université Claude-Bernard (Lyon) in
1991 and has been leading diagnosis activities at Institut Pasteur (Lyon) in the area of viral diseases (herpes viruses). Dr. Drouet
has an excellent track record in herpes virus field, and his group is engaged in clinical research in the field of EBV diseases. Current
research included the field of Hepatitis C Virus research, leading to elucidation of some aspects of its epidemiology and control.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
22
Sequential Intralesional Administration with Low Dose GM-CSF and IL-2 in Melanoma
Lesions can Induce Immense Antitumor Response that Reflects on Patient Survival
E. George Elias
University of Maryland St. Joseph Medical Center, USA
Abstract
Background: Cutaneous melanoma is an immunogenic tumor, but it seems to be very heterogeneous. Utilizing patient own
tumor as the source for tumor-specific antigens and activating the autologous immune system within the tumor by intralesional
injection of low dose GM-CSF and IL-2 can overcome such heterogeneity.
Methods: 10 patients with over 100 dermal and subdermal metastases were studied: 4 had in-transit metastases, 3 had large
sclerotic skin lesions and 2 had distant metastases with palpable subdermal lesions. Each received intralesional GM-CSF 500
µg once/week and if no complete response (CR) was achieved, intralesional IL-2 was substituted at 11 million IU weekly. In
addition, a patient with an invasive primary melanoma with a satellite metastasis received both cytokines on 2 consecutive
days, at both sites, one week before the surgical resection.
Results: Three patients with in-transit metastases had CR to GM-CSF and one failed but had CR to IL-2. The three large
sclerotic skin lesions failed to respond to either cytokine. The two patients with distant metastases had CR only at the injection
sites; one to GM-CSF and the other to IL-2. The resected tissue, one week after intralesional therapy with both cytokines,
showed complete tumor necrosis with massive histiocytosis and overexpression of CD4+ and CD+ cells at the injection sites and
in some regional lymph nodes. The duration of CR ranged from 31-60 months.
Conclusions: In vivo intralesional administration of two cytokines in melanoma lesions seemed to induce an immense
antitumor immune response that was transmitted via the lymphatics.
Development of Dendritic Cell-Based Cancer Vaccine in Japan
Masato Okamoto
Department of Advanced Immunotherapeutics, Kitasato University School of Pharmacy, Japan
Oncology Cencer, Kitasato Institute Hospital, Japan
Abstract
We have developed an effective dendritic cell (DC)-based cancer vaccine named “Vaccell”, and large-scale retrospective studies and
small-scale prospective trials in several types of malignancies such as pancreatic cancer and lung cancer. We have retrospectively
analyzed 255 patients with inoperable pancreatic cancer who received standard chemotherapy in combination with DC vaccines
pulsed with WT1 peptide etc. Survival time of the patients with positive DTH skin reaction was significantly prolonged as
compared with that of the patients with negative DTH. The findings strongly suggest that there may be “Responders” for the
DC vaccine in advanced pancreatic cancer patients. We next conducted a small-scale prospective clinical trial. In this trial,
we pulsed HLA class II-restricted WT1 peptide (WT1-II) in addition to HLA class I-restricted peptide (WT1-I) into the DCs.
Survival of the inoperable pancreatic cancer patients received DC vaccine pulsed with both WT1-I and -II was significantly
extended as compared to that of the patients received DC vaccines pulsed with WT1-I or WT1-II alone. Furthermore, WT1specific DTH positive patients showed significantly improved the survival rate as compared to the DTH negative patients. The
activation of antigen-specific immunity by DC vaccine in combination with standard chemotherapy may be associated with a
good clinical outcome in advanced pancreatic cancer. Similar results were observed in non-small cell lung cancer patients. We
are now planning pivotal studies of the “Vaccell” in appropriate protocols in Japan.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
23
DNA Vaccines for Cancer Immunotherapy
B. Ferraro1*, M. Bagarazzi1, J. Walters1, M. P. Morrow1, X. Shen1, J. Boyer1, J. Yan1, K. Kraynyak1, K.Broderick1, A.S.
Khan1, D.B. Weiner2, N. Y. Sardesai1, and L. Humeau1
Inovio Pharmaceuticals, Inc., USA
University of Pennsylvania Perelman School of Medicine, USA
1
2
Abstract
Immunotherapy of cancer through induction of anti-tumor cellular immunity has recently reemerged as an important therapy
for the treatment of cancers. Current treatment modalities for cancer can be useful, but may be accompanied by deleterious
side effects and often do not confer long-term control. Accordingly, additional modalities, such as immunotherapy, may
represent an important approach for the treatment of a variety of cancers. Delivery of DNA vaccines with electroporation
(EP) has shown promising results for therapeutic targets in a variety of species, and recently in humans. Specifically, Phase I
studies of VGX-3100, a highly optimized DNA immunotherapy for HPV16/18 delivered using EP, drove seroconversion to at
least one HPV antigen (E6 or E7) in 100% of patients while 78% of patients mounted a detectable IFN-γ ELISpot response.
Moreover, all patients showed the presence of CD8 T-cells exhibiting full HPV-specific cytolytic functionality, a readout thought
to be informative of the ability of VGX-3100 to induce an immune response that may be important for the elimination of
HPV-infected cells. Indeed, recent data from a randomized placebo controlled double blind Phase II efficacy study in women
with untreated HPV 16/18 associated high grade cervical intraepithelial neoplasia demonstrated that VGX-3100 treatment
led to the statistically significant regression of CIN 2/3 lesions and viral clearance relative to placebo treated patients. This
presentation will encompass VGX-3100 for the treatment of HPV-associated pre-cancers and the application of the DNA/EP
immunotherapeutic platform to target additional tumor types, such as prostate cancer, in both clinical and preclinical studies.
Restoring Anti-HER-2 CD4 Th1 Responses Using DC1 Vaccines in HER-2 Expressing Breast
Cancer
Brian J. Czerniecki
University of Pennsylvania, USA
Abstract
HER-2 Expressing breast cancer remains a difficult treatment problem in breast cancer. Those with high risk disease despite
HER-2 targeted therapies still have significant risk of recurrence. We have demonstrated a progressive loss of ant-HER-2 CD4
Th1 responses during tumorigenesis that correlates with increased risk of recurrence and response to neoadjuvant therapy.
We have demonstrated that this loss of anti-HER-2 Th1 response is not a result of a fixed deficit but can be restored using
HER-2 pulsed DC1 vaccines. Administration of HER-2-pulsed DC1 vaccines in high risk Stage III or IV NED patients or those
with residual disease post neoadjuvant chemotherapy results in restoration of anti-HER-2 Th1 responses. Th1 cytokines cause
induction of tumor senescence and apoptosis especially in combination with HER-2 Targeted therapies. Given the loss of MHC
class I expression in HER-2 expressing breast cancer consideration should be given to targeting CD4 Th1 cells to increase CD8
CTL function in patients with HER-2 expressing breast cancer. Targeting ant-HER-2 Th1 responses in breast cancer may be
critical to achieve meaningful long terms survival in patients with HER-2 expressing breast cancer.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
24
Resurgence of Phase I-Phase III Global Trials Multi-Dosing BCG for Autoimmunity and
Allergies
Denise L. Faustman
Masschusetts General Hospital/Harvard Medical, USA
Abstract
The Bacillus Calmette-Guerin (BCG) vaccine, long used in tuberculosis prevention, has gained new attention in recent years
for potential immunomodulatory effects in multiple types of human autoimmunity, including type 1 diabetes and multiple
sclerosis, as well as atopic diseases. This revived attention is due in part to mechanistic studies that suggest multiple BCG
vaccinations or the related and more pathologic tuberculosis organism modulate the immune system. Similar to tuberculosis,
BCG induces tumor necrosis factor (TNF). TNF is a primary ligand for regulatory T cell (Treg) expansion and creation of potent
Treg cells. Human clinical trial data in type 1 diabetes now documents the beneficial induction of Treg cell population in multiple
autoimmune diseases after BCG vaccination. Further, although long-term beneficial effects are observed after BCG vaccination
in patients with type 1 diabetes, similar long-term benefits are not observed after exposure of type 1 diabetic humans to
other organisms that induce acute TNF and Tregs. This trait now distinguishes the BCG organism for a more durable effect in
immunoregulation. In autoimmunity, the most advanced trials examining multi-dosing of the BCG vaccine are being conducted
in multiple sclerosis (Phase III treatment studies), type 1 diabetes prevention (via childhood vaccination) and type 1 diabetes
treatment (Phase II treatment studies in long term diabetic cohorts). Trials are also beginning in the United States and Europe
to examine BCG in Sjögren’s syndrome.
Biography
Dr. Denise Faustman, MD, PhD, is Director of the Immunobiology Laboratory at the Massachusetts General Hospital (MGH)
and an Associate Professor of Medicine at Harvard Medical School. She is currently leading a clinical trial program investigating
the potential of the BCG vaccine as a disease reversal treatment for long-standing type 1 diabetes. Dr. Faustman’s research has
been highlighted in publications including Science, Nature, The Wall Street Journal, The New York Times, Los Angeles Times, The
London Financial Times and Scientific American. She earned her MD and PhD from Washington University School of Medicine,
in St. Louis, Missouri, and completed her internship, residency, and fellowships in Internal Medicine and Endocrinology at the
Massachusetts General Hospital.
A Tumor Mitochondria Vaccine Protects Against Experimental Renal Cell Carcinoma
Stefano Pierini, Chongyun Fang, Stavros Rafail, John G. Facciponte, Jialing Huang, Francesco De Sanctis, Mark A.
Morgan, Mireia Uribe-Herranz, Janos L. Tanyi and Andrea Facciabene*
Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, USA
Abstract
Mitochondria provide energy for cells via oxidative phosphorylation (OXPHOS). Reactive oxygen species (ROS), a byproduct of
this mitochondrial respiration, can damage mitochondrial DNA (mtDNA), and somatic mtDNA mutations have been found in
all colorectal, ovarian, breast, urinary bladder, kidney, lung, and pancreatic tumors studied. The resulting altered mitochondrial
proteins or tumor associated mitochondrial antigens (TAMAs) are potentially immunogenic, suggesting that they may be
targetable antigens for cancer immunotherapy. Here, we show that the RENCA tumor cell line harbor TAMAs that can drive an
antitumor immune response. We generated a cellular tumor vaccine by pulsing dendritic cells (DC) with enriched mitochondrial
proteins from RENCA cells. Our DC-based RENCA mitochondrial lysate vaccine elicited a cytotoxic T-cell response in vivo
and conferred durable protection against challenge with RENCA cells when used in a prophylactic or therapeutic setting. By
sequencing mtDNA from RENCA cells we identified two mutated molecules, COX-1 and ND-5. Peptide vaccines generated
from mitochondrial-encoded cytochrome c oxidase 1 (COX1) but not from NADH dehydrogenase 5 (ND5) had therapeutic
properties similar to RENCA mitochondrial protein preparation. Thus, TAMAs can elicit effective antitumor immune responses,
potentially providing a new immunotherapeutic strategy to treat cancer.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
25
A Novel Clinical Trials Design for Combination Therapy of Cancer Vaccines and Immune
Modulators
Osama E. Rahma1*, Emily Gammoh2, Richard M. Simon3, and Samir N. Khleif4
University of Virginia, USA
Arabian Gulf University, RCSI-Bahrain
3
Biometric Research Branch, National Cancer Institute, USA
4
Georgia Health Sciences Cancer Center, USA
1
2
Abstract
Cancer vaccines had been under investigation in clinical trials for the last few decades. However, their clinical outcomes had
been disappointing. This may be due in part to the suppressive tumor microenvironment including suppressive immune
cells and immune checkpoints. Accordingly, the combination of cancer vaccines and immune modulators, such as checkpoint
inhibitors, must be further investigated. We had previously shown that the traditional phase I dose escalation design may not
be applicable for therapeutic cancer vaccines development due to the lack of correlation between dose escalation and cancer
vaccines’ toxicities or cellular immune responses except for certain cases. Here, we present a novel design for therapeutic
cancer vaccines early development. This design may allow investigators to conduct early phase clinical trials in cancer vaccines
without the need to enroll large number of patients in order to identify the safe and immune active dose. Subsequently,
the identified safe and immune active dose could be used in combination with other agents, such as immune checkpoint
inhibitors, in phase II/III clinical trials. This would provide clinical investigators with a new tool to conduct clinical trials using
a combination of cancer vaccines and immune modulators in cancer immunotherapy.
Biography
Dr. Osama Rahma has completed his MD from Damascus University and Internal Medicine Residency from East Carolina
University. He completed a fellowship in Immunotherapy and Medical oncology from the Vaccine Branch and the Medical
oncology Branch at the National Cancer Institute. He is currently a co-leader of the hepatobiliary and pancreatic cancer program
at University of Virginia. Dr. Rahma is a Principal Investigator on few multicenter clinical trials in pancreatic cancer and has
published more than 20 papers in reputed journals.
Intravenous Immune Globulin (IGIV) Containing High Titer Neutralizing Antibodies to
Respiratory Syncytial Virus (RSV) and other Respiratory Viruses (RI-002)
James J Mond 1*, Ann Falsey2, Jordan Orange3, and Wei Du4
1
ADMA Biologics Inc. USA
2
University of Rochester School of Medicine, USA
3
Baylor School of Medicine, USA
4
Clinical Statistics Consulting, USA
Abstract
Despite the great advances in vaccine technology and novel targets that mediate protection for infectious diseases, there remains
a large segment of the population that will never benefit from these advances. Patients with serious immune deficiencies
whether caused by genetic mutations as in primary immune deficiencies (PIDD) or caused by medically induced suppression of
the immune response are unresponsive to immunogenic stimulation. Protection can be mediated only by infusion of immune
globulin or by restoration of immune responses by stem cell transplantation. ADMA Biologics has developed an IVIG containing
high titer neutralizing antibodies to RSV and other respiratory viruses (RI-002) which in preclinical studies was shown to treat
lower tract pulmonary RSV infections in chronically immune suppressed cotton rats, the ideal surrogate animal model for
RSV. Treatment with RI-002 completely reversed an RSV mediated abnormal pulmonary histology in these infected immune
suppressed cotton rats to one that is entirely normal. Unlike the available monoclonal anti RSV antibody Palivizumab, RI-002 is
also protective for escape mutant RSV that is resistant to the monoclonal antibody. We have recently completed a pivotal phase
III clinical trial in patients with PIDD with RI-002 with successful attainment of primary and secondary endpoints. There were
zero serious infections observed during the one year duration of the trial. The secondary endpoints measured reflected striking
improvement in all of the subject`s clinical endpoints. Additional studies may be warranted to explore whether the unique
antibody profile of RI-002 may be correlated to these successful outcomes.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
26
A Randomized Phase II Study of the CSF-470 Therapeutic Vaccine plus BCG plus rhGM-CSF
versus IFN-α2b in Cutaneous Melanoma Patients Stages IIB, IIC and III
María Marcela Barrio1*, María Betina Pampena1, Mariana Aris1, Paula Blanco1, Alicia Ines Bravo2, Juan O´Connor3, Ana
Gabriela Orlando4, Franco Ramello5, Estrella Mariel Levy1, and Jose Mordoh2
1
Centro de Investigaciones Oncologicas-Fundacion Cancer, Ciudad Autonoma de Buenos Aires, Argentina
2
Instituto Medico Especializado Alexander Fleming, Ciudad Autónoma de Buenos Aires, Argentina
3
Unidad de Inmunopatología, Hospital Interzonal General de Agudos Eva Peron, San Martín, Provincia de Buenos Aires, Argentina
4
Reconquista Santa Fé, Argentina
5
Centro Médico San Lucas, Gualeguaychu, Entre Ríos, Argentina
Abstract
Adjuvant treatment of high-risk cutaneous melanoma (CM) patients (pts) is still an unsolved issue, since the cost-benefit ratio
of high-dose IFN-α2b is under discussion. We have developed CSF-470 therapeutic vaccine, consisting in four lethally-irradiated
allogeneic CM cell lines+BCG + rhGM-CSF as adjuvants, currently being tested vs medium-dose IFN-α2b in post-surgical stage
IIB-III CM pts (Phase II/III trial). I will present the results of the phase II part of the study. A total of 30 pts were randomized:
19 pts to receive CSF-470 and 11 pts to IFN-α2b. During the two-year treatment, pts in the vaccine arm received a total of 13
vaccinations. Our results demonstrate a clear superiority of CSF-470 plus BCG plus GM-CSF vs IFN-α2b:a)CSF-470 was better
tolerated than IFN-α2b: CSF-470 main toxicity was reaction at the injection site(grade 2); 3/19 pts presented grade 3 allergic
reactions easily handled with anti-histamines and corticosteroids; b)with a mean follow-up of 42 m a significant benefit in
the distant metastasis-free survival (DMFS) for CSF-470 was observed: vaccine arm 68.4% vs IFN-α2b 27.2% (p=0.025);
median OS in both groups is still undefined; c)QOL was significantly superior for CSF-470 vs IFN-α2b treatment (p<0.0002).
With respect to immune monitoring in vaccinated pts, a) DTH was higher in DMF pts than in progressing pts; b) a significant
increase in peripheral blood NK cells, a decrease in Tregs and development of anti-CM cells Abs at 6 months after initiation of
treatment were observed. The phase III part of the study is currently ongoing.
Biography
Dr. María Marcela Barrio graduated in 1991 as a Biologist at the University of Buenos Aires and obtained her PhD working on monoclonal
antibodies at the Fundación Instituto Leloir. She became a member of The National Scientific and Technical Research Council (CONICET)
in 2007. Dr. Barrio has been working in the cancer immunology as part of Dr. Mordoh´s team since 1984. At present she is Subdirector
of the Centro de Investigaciones Oncológicas Fundación Cáncer. She has published more than 40 scientific papers about her specialty. Dr.
Barrio is working in translational research for the development of therapeutic vaccines for cutaneous melanoma.
Phase I Clinical Study of a Venezuelan Equine Encephalitis Virus DNA Vaccine Candidate
Delivered by Electroporation Mediated Intradermal or Intramuscular Administration
Drew Hannaman1*, Lesley C. Dupuy2, and Connie S. Schmaljohn2
Ichor Medical Systems, Inc., USA
United States Army Medical Research Institute of Infectious Diseases, USA
1
2
Abstract
Venezuelan equine encephalitis virus (VEEV) is a mosquito borne alphavirus that is a NIAID Category B pathogen. The development of
effective vaccines against VEEV is warranted due to the potential for both natural and intentional disease outbreaks. Currently, there
are no VEEV vaccines licensed in the U.S. for human use. In light of their favorable safety profile and potential logistical advantages,
DNA vaccines represent an appealing approach to the development of novel vaccines against known and emerging pathogens.
Following demonstration of protection against VEEV challenge in multiple animal models, a novel VEEV DNA vaccine candidate
was evaluated in a randomized, placebo controlled Phase I study. The study was designed to assess the safety and immunogenicity
of the candidate delivered either intradermally or intramuscularly using the electroporation-based TriGrid Delivery System. A total
of 42 subjects were enrolled in the three arm study (intramuscular VEEV DNA vaccine, intradermal VEEV DNA vaccine, or placebo).
Subjects received three injections administered as placebo (saline) or one of two DNA dose levels (0.5 mg/ml or 2.0 mg/ml) at Week 0,
4 and 8. Administrations were well tolerated with mild injection site reactions comprising the vast majority of adverse events. There
were no vaccine or device related serious adverse events observed. Analysis of serum anti-VEEV neutralization activity demonstrate
high rates of seroconversion for both the intradermal and intramuscular routes of delivery and a DNA dose dependent magnitude of
response. These data provide initial proof of concept for the vaccine candidate and have set the stage for further clinical development.
Biography
Drew Hannaman has served as the Vice President of Research & Development at Ichor Medical Systems since 2000. In that role
he has been responsible for the development of the company's electroporation based TriGrid Delivery System for DNA vaccine
administration. The technology has been incorporated into more than 20 clinical stage products addressing a range of disease
indications. He is a co-author of over 40 publications in the field of DNA based biologics. He holds a degree in Cybernetics from
the University of California, Los Angeles and has over twenty years of experience in the development of drug delivery systems.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
27
Live Attenuated Hepatitis A Vaccine – Long Term Persistence of Immunogenecity of Single
Dose in Children – Indian Perspective
Monjori Mitra1*, Nitin Shah2, MMA Faridi3, Apurba Ghosh1, VS Sankaranarayanan4, Anju Aggarwal5, Suparna
Chatterjee6, Nisha Bhattacharyya1, Ganesh Kadhe7, Gaurav Vishnoi7, and Amey Mane7
Institute of Child Health, India
Department of Pediatrics, Lion’s Tarachand Bapa Hospital, India
3
Department of Pediatrics, University College of Medical Sciences, GTB Hospital, India
4
Kanchi Kamakoti Childs Trust Hospital, India
5
Department of Pediatrics, University College of Medical Sciences, GTB Hospital, India
6
Deptartment of Pharmacology, Institute of Postgraduate Medical Education & Research, India
7
Medical Affairs, Wockhardt Limited, India
1
2
Abstract
Worldwide, viral hepatitis continues to be a cause of considerable morbidity and mortality. Mass immunization with a single
dose of live attenuated HAV has been shown to significantly reduce disease burden in the community. This was a phase IV, 5-year
follow up study carried out at 4 centers (Kolkata, Delhi, Mumbai and Chennai) across India.
The subjects with antibody titer <20 mIU/mL at baseline were evaluated for long term immunogenicity. Of the 503 subjects
enrolled, 349 subjects were baseline seronegative with an anti-HAV antibody titer <20 mIU/mL. Overall, 343 subjects could be
followed up at some point of time during this 5 y post vaccination period. In the last year (60 months) of follow-up, 108 subjects
(97.3%) of 111 subjects (who came for follow-up at the end of 5 y) had a protective antibody titer (anti-HAV antibody titer
>20 mIU/mL). The seroconversion rates considering seroprotection levels of anti-HAV antibody titer >20 mIU/mL, following
vaccination starting from 6 weeks, 6 months, 12 months, 24 months, 36 months,48 months and 60 months were 95.1%, 97.9%,
98.3%, 96.2%, 97.8%, 92.6% and 97.3%, respectively. The geometric mean concentration (GMC) over the years increased from
64.9 mIU/mL at 6 weeks to 38.1 mIU/mL and 135.2 mIU/mL at 6 months and 12 months, respectively and was maintained
at 127.1 mIU/mL at 60 months. In conclusion, the result of this 5-year follow up study showed that the single dose of live
attenuated vaccine is well tolerated and provides long term immunogenicity in healthy Indian children.
National Differences in Requirements for Ethical and Competent Authority Approval for a
Multinational Vaccine Trial under the EU Directive 2001/20/EC
Eva van Doorn*, E. Hak and B. Wilffert
University Groningen, The Netherlands
Abstract
Obtaining approval for a multinational vaccine trial from an ethics committee and the national competent authority of
different Member States of the European Union (EU) is challenging under clinical trial Directive 2001/20/EC. This because of
the differences in the implementation of the directive in national laws of Member States. The national ethical and competent
authority review procedures in Finland, Hungary, The Netherlands, Norway and Slovenia are discussed to illustrate the national
differences in requirements for ethical and competent authority approval in addition to the provisions of the trial directive
related to both review procedures. It illustrates the differences between the countries in the documents that have to be submitted
for the review procedures, the submission procedures and the language requirements of the documents, the organization of the
ethics committees and the role of the competent authority in the approval procedure.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
28
Track-B
Synthetic Virus-Like Particles (SVLPs) for the Development of Fully Synthetic Prophylactic
Vaccines
Armando Zuniga1*, Nina Geib1, Aniebrys Marrero-Nodarse1, Marco Tamborrini2, Kerstin Mohele3, Rajni Sharma4,
Kenneth McCullough4, Gerd Pluschke2, Tina Riedel3, Peter Rusert3, Alexandra Trkola3, John A. Robinson3, and Arin
Ghasparian1
Virometix AG, Switzerland
Swiss Tropical and Public Health Institute, Switzerland
3
University of Zurich, Switzerland
4
Institute of Virology and Immunoprophylaxis, Switzerland
1
2
Abstract
Synthetic Virus-Like Particles (SVLPs) represent a novel strategy for the development of next-generation prophylactic and
therapeutic vaccines. SVLPs do not require cell lines or other biological systems for their production since they are prepared fully
synthetically using well-established peptide synthesis methods and down-stream processing. These nanoparticles are in the size
of 20-30 nm and have similar chemical composition as naturally derived Virus-Like Particles (VLPs). SVLPs, when loaded with
peptide antigens of choice, resemble VLPs in their size, shape, and the way repetitive antigens on their surface are presented to
the immune system. In vivo studies of SVLPs carrying various antigens in different animal species have shown that SVLPs are an
excellent antigen delivery system, are highly immunogenic at low doses without co-administration of adjuvant, and elicit strong
peptide specific immune responses against the parental protein in the context of the native pathogen. In vitro characterization
showed that they interact very efficiently with dendritic cells and are capable of strongly activating mouse, pig and human
immune cells. The talk will provide an overview about the SVLP technology with specific examples.
Biography
Dr. Armando Zuniga is the Chief Scientific Officer of the company Virometix AG. Dr. Zuniga has been working in the field of
vaccine development since 15 years. Prior to joining Virometix in 2013 he held various positions at Redbiotec, Berna Biotech
and Crucell. He was strongly involved in the development of the measles virus vaccine vector technology that is now being
tested in clinical trials by two vaccine companies. Dr. Zuniga received his Doctoral degree in Bacteriology from the University
of Zurich, Switzerland.
Antibody Induction by Group B Streptococcal Vaccines for Protection against Fungal
Infections
John F. Kearney*, Emily K. Stefanov, R. Glenn King, and Rebekah E. Wharton
Department of Microbiology, University of Alabama at Birmingham, USA
Abstract
The morbidity associated with invasive fungal infections (IFIs) is high and IFIs are highly intractable to treatment once these
opportunistic pathogens gain a foothold. We are proposing new active vaccination and passive protection strategies that
provide more optimal prevention and treatment for personnel at risk for IFI. We are proposing a novel approach that involves
vaccination with immunogens expressing epitopes shared by Aspergillus fumigatus (A.f.) and other fungi with bacteria. We
have shown that anti-Group B streptococcal (GBS) monoclonal antibodies and a GBS bacterial vaccine protect against IFI in
mice, and that immune sera from GBS polysaccharide-tetanus toxoid conjugate human vaccinees react to A.f. very similarly to
GBS induced protective antibodies from mice. This strategy will circumvent the low responsivity of the immune response to
surface fungal antigens which normally leading to a lack of protective immunity in fungal infections. This approach, to develop
active immunity against IFI, is based on findings that (i) immunization of mice with whole heat-killed strains of streptococci
induced protection against IFI and (ii) anti-A. fumigatus Abs are induced in humans by conjugate vaccines that have been used
to vaccinate humans against streptococcal infections. Repurposing of prophylactic conjugate vaccines and availability of passive
monoclonal antibodies for protection against IFI by multiple fungal species would have a high impact and provide much needed
treatments for patients at risk for, or suffering from IFIs.
Biography
Dr. John F. Kearney completed his Ph.D. studies in immunology at the University of Melbourne, Australia in 1973 and carried
out postdoctoral studies at UAB where he is now Distinguished Professor in Microbiology. His present research is focused
on the following areas which are not mutually exclusive but sometimes overlap between human and mice. These include (i)
development of the immune system, (ii) analysis of immune responses to bacterial organisms including the pathogens and
opportunistic pathogens (Streptococcus pneumoniae, group B streptococci, Enterobacter cloacae, Aspergillus fumigates).
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
29
Pertussis Vaccine Efficacy against Old and Novel Circulating Bacteria can be Improved by
the Addition of Two New Vaccine Candidates
Jimena Alvarez Hayes1, Juan Marcos Oviedo1, Miguel Ayala2, Fabricio Maschi2, Cecilia Carbone2, and María Eugenia
Rodriguez1*
CINDEFI (UNLP-CCT CONICET) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
Catedra de Animales de Laboratorio, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, Argentina
1
2
Abstract
Bordetella pertussis (Bp) is the etiologic agent of whooping cough, the most prevalent vaccine-preventable disease. Current
acellular pertussis vaccines (aP) are formulated with different combination of Bp virulence factors such as pertactin (Prn) among
others. Probably due to vaccine immune selection, during the last few years clinical isolates not expressing Prn have been causing
epidemic outbreaks. While Prn is not critical for infection, it is the only antigen present in Pa able to raise opsonophagocytic
antibodies, a key protecting activity against Bp infection. Thus, the addition of new opsonin targets is increasingly needed to
improve current vaccines. Recently, we characterized two new antigens, namely AfuA and IRP1-3, which are already in preclinical
studies of pertussis vaccines. These two antigens are particularly interesting since they induce opsonic antibodies that increase
significantly Pa protective capacity. In the present study we investigated whether this new vaccine formulation might be
also more effective against this newly circulating clinical strain. By mean of flow cytometry and fluorescence microscopy we
found that, as compared with anti-aP anti-serum, opsonization with serum from mice vaccinated with aP+AfuA+IRP1-3 (aP*)
significantly increased PMN phagocytosis of a Prn deficient mutant of Bp (Bp∆Prn), a strain that mimics new clinical isolates
genotype. Polymyxin B protection assay showed that bacteria opsonized with anti-aP* anti-serum were efficiently killed by
PMN. Finally, we evaluated the ability of aP* vaccine to protect mice against infection with Bp∆Prn. We observed a significant
increase in the protection level when mice were immunized with aP* as compared with aP immunization (p<0,05). Altogether
these results suggest that a new generation of aP including AfuA and IRP1-3 will not only provide higher protection against the
wild type strain of Bp infection but also against new circulating Bp genotypes.
A Novel Therapy for Melanoma Developed in Mice: Transformation of Melanoma into
Dendritic Cells with Listeria monocytogenes
Carmen Alvarez-Dominguez1*, Ricardo Calderon-Gonzalez1, Lucia Bronchalo-Vicente1,2, Javier Freire3, Jose Javier
Gomez-Roman3 and Sonsoles Yañez-Diaz2
Instituto de Investigacion Marques de Valdecilla (IDIVAL), Santander, Spain
Servicio de Dermatologia, Spain
3
Servicio de Anatomía Patologica, Hospital Universitario Marques de Valdecilla, Satander, Spain
1
2
Abstract
Listeria monocytogenes is a gram-positive bacteria and human pathogen widely used in cancer immunotherapy because of its
capacity to induce a specific cytotoxic T cell response in tumours. This bacterial pathogen strongly induces innate and specific
immunity with the potential to overcome tumour induced tolerance and weak immunogenicity. Here, we propose a Listeria based
vaccination for melanoma based in its tropism for these tumour cells and its ability to transform in vitro and in vivo melanoma
cells into matured and activated dendritic cells with competent microbicidal and antigen processing abilities. This Listeria
based vaccination using low doses of the pathogen caused melanoma regression by apoptosis as well as bacterial clearance.
Vaccination efficacy is LLO dependent and implies the reduction of LLO-specific CD4+ T cell responses, strong stimulation of
innate pro-inflammatory immune cells and a prevalence of LLO-specific CD8+ T cells involved in tumour regression and Listeria
elimination. These results support the use of low doses of pathogenic Listeria as safe melanoma therapeutic vaccines that do not
require antibiotics for bacterial removal.
Biography
Dr. Carmen Alvarez-Dominguez has completed her Ph.D at the age of 29 years from Universidad Autonoma de Madrid and
postdoctoral studies from Washington University School of Medicine on Small GTPases role in Listeria monocytogenes
phagocytosis. She is the director of the Group on Genomics, Proteomics and Vaccines at the Research Institute Marqués de
Valdecilla (IDIVAL) in Santander, Spain. She has published more than 29 papers in reputed journals and serving as an editorial
board member of Microbes and Infection and OMICS. She is also a coauthor of the patent: Immunogenic peptides against
Listeria and Mycobacterium, antibodies and their uses with reference PCT/ES2007/070144.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
30
Update on the Development of Vaccines against Shigella
Mark S. Riddle
Naval Medical Research Center, USA
Abstract
Shigella remains a World Health Organization (WHO) priority for vaccine development. Emerging epidemiology has reemphasized the importance of this pathogen among developing world and traveler populations. Of particular importance is the
growing recognition of antimicrobial resistance and chronic health consequences of shigellosis which make primary prevention
a priority. Multiple vaccine strategies, mostly relying upon serotype-specific O-SPs as antigens are under development and
entering into proof of concept studies in humans. Live-attenuated approaches have advantages for ease of delivery and low cost
of goods with efforts to strike the right balance between reactogenicity and immunogenicity. Killed whole-cell approaches are
also an attractive cellular-based strategy, though as with live-attenuated, generation of a robust immune response induction
in the gut of the developing world child has been a challenge. Lipooligosaccharide protein conjugate vaccines have advanced
to the field and new generation candidates are now in early clinical trials. Other novel strategies including subcellular type III
secretion systems and outer membrane vesicle constructs also show promise. Given all the strategies and global health interest,
the outlook for a Shigella vaccine is promising.
Biography
Dr. Mark S. Riddle is a Captain in the Medical Corps of the United States Navy and boarded in General Public Health &
Preventive Medicine. He is a 1997 graduate of Tulane University where he earned both his MD and MPH&TM, and Uniformed
Services University of the Health Sciences where he earned Doctorate in Public Health. Dr. Riddle has over 15 years in the field
of vaccinology ranging from pre-clinical vaccine development and clinical vaccine and drug development, to vaccine health
economics and research related to the chronic health consequences of acute infection. He is the co-author of over 100 peer
reviewed publications, four book chapters, and is the Editor-in-Chief of Tropical Diseases, Travel Medicine and Vaccines (a
BioMed Central journal).
Single-cycle Replicating Adenovirus Vaccines
Michael A. Barry
Mayo Clinic, USA
Abstract
Replication-competent adenoviral (RC-Ad) vectors generate exceptionally strong gene expression and vaccine responses by
amplifying the transgenes they carry up to 10,000-fold. While they are potent, they also risk causing adenovirus infections in
patients or health care workers. More common replication-defective Ad (RD-Ad) vectors with deletions of E1 avoid this risk.
However, RD-Ads do not replicate transgene and generate markedly weaker expression. We recently engineered “single-cycle”
adenovirus (SC-Ad) vectors that still replicate their genomes and the transgenes they carry, but that do not generate infectious
progeny. In primary human cells, SC-Ads require 33-fold less vector to equal expression mediated by an RD-Ad. These Ads
replicate transgenes in mice, hamsters, ferrets, bovine, sheep, non-human primates, and of course humans. Tests in mouse,
hamster, and macaque models demonstrate SC-Ads mediate more robust expression and immune responses than RD-Ads.
For influenza vaccines, this potency translates into the need for 100-fold less SC-Ad than RD-Ad vaccine to generate the same
hemagglutination inhibition titers. These data suggest that single-cycle replicating Ad vectors may have utility as vaccines or
therapies against infectious agents, cancer, and other applications in an array of species.
Biography
Dr. Michael A. Barry received his Ph.D from Dartmouth working on Apoptosis with Alan Eastman. He performed his postdoctoral fellowship on Genetic Immunization and Vector targeting at UT Southwestern Medical Center with Stephen Johnston.
Dr. Barry spent 10 years with a joint appointment in the Center for Cell and Gene Therapy at Baylor College of Medicine and
in Bioengineering at Rice University prior to moving to Mayo Clinic. His laboratory currently works on gene therapy, genetic
vaccines, oncolytic viruses, and basic virology.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
31
HSV-2 Glycoprotein D Deletion Virus Induces Protective ADCC Mediated Immunity and
Provides Complete Protection against HSV Latency
Christopher Petro, Kayla Weiss, Nazanin Khajoueinejad, Natalia Cheshenko, Brian C. Weinrick, William R Jacobs Jr*,
and Betsy C. Herold*
Albert Einstein College of Medicine, USA
Abstract
Background: Previous efforts to develop prophylactic vaccines against HSV have primarily focused on subunit formulations
comprised of glycoprotein D (gD-2) alone or in combination with other HSV antigens and different adjuvants. These vaccines
induce high levels of serum neutralizing antibodies (Abs) against gD, but have failed to protect in large-scale clinical trials. We
hypothesize that a virus deleted in gD-2 will elicit a different and more protective immune response than that observed with
other vaccine candidates or natural infection.
Methods: An HSV-2 gD null virus was engineered by co-transfecting VD60 cells (gD-1 expressing Vero cells, gift from D.
Johnson) with HSV-2(G) DNA and an allelic exchange plasmid containing a CMV-GFP/Neo+ cassette flanked by regions upstream
and downstream to gD-2. The null virus was propagated on VD60 cells to yield phenotypically complemented virus (designated
ΔgD) capable of a single cycle of infection. Safety was assessed in SCID mice and efficacy by subcutaneously vaccinating (primeboost) female C57BL/6 (B6) and Balb/C mice with HSV-2 ∆gD or VD60 cell lysates (control) and challenging with HSV-2 or
HSV-1 intravaginally or via skin scarification. Passive transfers were performed with serum and T cells harvested 3 weeks post
boost. Serum and vaginal washes were collected to measure viral shedding and Ab responses, dorsal root ganglia (DRG), genital
tract tissue and skin were used to assess viral spread by PCR and culture; and spleen, lymph nodes, and skin sections to measure
cellular immune responses.
Results: The ΔgD virus caused no disease in SCID mice and no virus was detected in DRG by PCR. In initial efficacy study, primeboost subcutaneous vaccination with 10^6 pfu protected 20/20 B6 and 10/10 Balb/C mice against intravaginal viral challenge
with a clinical isolate of HSV-2 (4674), whereas none of the control-vaccinated mice were protected (p=0.0001). Complete
protection was also observed with vaccine doses as low as 10^4 pfu and against a panel of 5 HSV-1 and 5 HSV-2 clinical isolates
including 100x the LD90 of a highly virulent clinical isolate from Africa (SD90) in both the vaginal and skin scarification models.
No infectious virus or HSV DNA has been detected in the DRG at any time post challenge in ΔgD-2 vaccinated mice in either
the vaginal or skin scarification model, indicating that the vaccine prevents the establishment of latency. The vaccine induces
high levels of HSV-specific Abs (1:800,000) and HSV-specific CD4+ and CD8+ T cell responses. Passive transfer of serum from
ΔgD-vaccinated mice completely protected naïve animals from intravaginal or skin challenge. Protection was lost if serum was
transferred into FcγR and FcRn knockout mice. The HSV-specific Abs were enriched for IgG2, were rapidly transported into
vaginal mucosa and skin following viral challenge, recognized multiple HSV antigens, and mediated significantly increased
antibody-dependent-cellular-cytotoxicity and antibody-dependent phagocytosis compared to HSV infected mice, but had low
levels of neutralizing activity (50% neutralization titer 1:5). In contrast, serum Abs from infected mice primarily recognized gD
and gB by Western blot.
Conclusions: The immune responses to ΔgD differ from those observed with subunit or other attenuated HSV vaccines and
natural infection. The latter are characterized by high titers of neutralizing Abs whereas ΔgD induces robust FcγR-mediated Ab
responses that are rapidly transported into mucosa sites and recognize a broad repertoire of HSV-2 proteins. These findings
support the hypothesis that gD interferes with the development of protective responses because of immune dominance or
immune modulatory properties. The ΔgD vaccine may be more effective than previous vaccine candidates and warrants further
development.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
32
Gorilla Adenovirus Vectors for Molecular Therapeutics and Vaccines
Douglas E. Brough
GenVec Inc, USA
Abstract
Adenovirus vectors built from gorilla adenovirus are showing superior performance characteristics for molecular vaccines. These new
adenovectors (GC Ads) are built from virus isolated from gorilla, have been cloned and engineered to be replication-defective, grow
efficiently to high titer on cell lines GenVec has characterized, banked and filed with regulatory agencies for scalable manufacturing
and can be deleted of multiple regions (e.g., E1, E3, and E4) to provide space for multiple transgene cassettes, provide multiple
blocks to eliminate replication competent virus and to ensure safer transgene delivery. They are not recognized by human sera
from either the United States or from Africa and a single intramuscular injection can induce durable immune responses to encoded
antigen. For example, intramuscular injection of GC Ads expressing RSV fusion (F0) glycoprotein induced neutralizing antibody
titers that were durable. > 24weeks, and protected the lung and nose against RSV challenge at even low doses of immunization in
cotton rat. A single intramuscular administration of GC Ads expressing CSP generated CD8+IFNg+ T cell responses to CSP that were
greater than that generated with Ad5 in a malaria mouse model. A single intramuscular administration of GC Ads expressing two
HSV antigens, UL19 and UL47 resulted in high level cellular responses that were protective against intravaginal HSV2 challenge in
mice and the CD8+IFNg+T cell responses were shown to be durable, 26 weeks. Moreover, repeat administration of the same GC Ad
resulted in a boost of antigen specific immune response. Unlike other adenoviral vectors, homologous prime boost regimens with
these GC Ads resulted in a boost of the CD8+IFNg+T cell responses at all dose and intervals tested. These new GC Ads based on
gorilla adenoviruses show generation of strong, durable T cell and neutralizing antibody responses to multiple antigens from a single
administration that can be further enhanced by repeat administration. Based on these data the platform shows great promise for
development of a broad array of immunotherapeutic and molecular vaccine product candidates.
Biography
Dr. Douglas E Brough joined GenVec in 1993 and currently serves as Chief Scientific Officer of the Company having previously
held several senior positions in GenVec Research.
Epstein-Barr virus Vaccines
Jeffrey I. Cohen
Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA
Abstract
Epstein-Barr virus (EBV) is the major cause of infectious mononucleosis and is associated with several malignancies including
EBV lymphoma in transplant recipients and nasopharyngeal carcinoma, gastric carcinoma, Hodgkin lymphoma, non-Hodgkin
lymphoma, and Burkitt lymphoma. The virus is associated with almost 200,000 cases of cancer annually in the world. In
1985 Epstein and colleagues reported that vaccination of cottontop tamarins with EBV glycoprotein 350 (gp350) protected the
animals from development of lymphomas after a high dose challenge with EBV; however, no vaccines for EBV have been licensed.
Most prophylactic vaccines to prevent EBV infection and/or disease have focused on EBV gp350, while therapeutic vaccines to
treat persons with EBV-associated malignancies have been directed against EBV LMP2 and EBNA1. In the only phase 2 trial
of a soluble EBV gp350 vaccine performed to date, the vaccine reduced the rate of infectious mononucleosis by 78%, but did
not prevent infection. We have developed a nanoparticle-based vaccine expressing gp350 and show that the vaccine induces
~100-fold higher levels of B cell neutralizing antibody in mice compared with soluble gp350. We are also studying other EBV
glycoproteins to try to protect other cell types from EBV infection.
Biography
Dr. Jeffrey I. Cohen is chief of the Laboratory of Infectious Diseases at the National Institute of Allergy and Infectious Diseases
at the National Institutes of Health. He studies the molecular genetics, pathogenesis, and clinical aspects of human herpes
viruses, including Epstein-Barr virus (EBV), herpes simplex virus (HSV), varicella-zoster virus (VZV), and cytomegalovirus
(CMV). His laboratory focuses on vaccine development, genes important for virus entry, and identification of cellular mutations
in patients with severe herpes virus infections. Clinical projects include a Phase I study of a replication defective herpes simplex
virus vaccine and evaluation of patients with severe herpes virus infections.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
33
Development of Live-Attenuated Respiratory Syncytial Virus Vaccines
Ursula J. Buchholz1*, Cindy Luongo1, Ruth A. Karron2, and Peter L. Collins1
RNA Viruses Section, LID, NIAID, NIH, USA
Center of Immunization Research, Johns Hopkins University, USA
1
2
Abstract
Respiratory syncytial virus (RSV) is the leading cause of viral lower respiratory tract disease in infants and children. We use
reverse genetics to develop live-attenuated vaccine candidates that are safe and immunogenic for infants and children ages
6-24 months. Vaccine candidates based on three different approaches are currently being evaluated in pediatric phase I studies.
One approach relies on genetically stabilized point mutations, conferring attenuation and temperature sensitivity. A second
approach is based on deletion of the interferon antagonist RSV NS2, which also functions as pathogenicity factor promoting
airway cell shedding. The third and most promising approach is based on deletion of the RNA regulatory protein M2-2 (∆M2-2),
resulting in attenuation, upregulation of viral protein expression, and increased immunogenicity. A single dose of RSV∆M2-2
was safe and well tolerated in adults, RSV-seropositive and seronegative children, and immunogenic in seronegative children.
When replication and immunogenicity was compared to a previous vaccine candidate with temperature-sensitivity mutations,
we found that shedding was significantly reduced while the mean serum antibody titer was significantly higher in children who
received RSV∆M2-2. Over the following RSV season, several vaccines exhibited substantial antibody increases without reported
RSV illness, suggesting that RSV∆M2-2 primed for anamnestic responses to wild-type RSV infection and could protect against
RSV disease. The M2-2 deletion seemed to increase the inherent immunogenicity per infectious unit of virus, and may overcome
a major conundrum in the development of safe and immunogenic pediatric RSV vaccines. Further clinical evaluation of vaccine
candidates with the M2-2 deletion is in progress.
Biography
Dr. Ursula J. Buchholz, DVM, PhD, is an Associate Scientist at the RNA Viruses Section in the Laboratory of Infectious Diseases
at NIAID, NIH, USA (Section Chief: Peter L. Collins). She began working on respiratory syncytial virus (RSV) as a research
fellow and then as a principal investigator at the German Federal Research Center for Virus Diseases of Animals. In 2002, she
joined Dr. Peter Collins’ laboratory, driven by her interest in live-attenuated pediatric RSV vaccines. She currently is a scientific
investigator on several pediatric vaccine studies.
Adjuvant-Guidance of T cell Mediate Immune Responses
Magdalena Tary-Lehmann
Cellular Technology Limited (CTL), and Case Western Reserve University School of Medicine (CASE), USA
Abstract
Adjuvants are important enhancers of the immune response. The choice of adjuvant is especially crucial in the context of
subunit vaccine approaches. In the past, adjuvants have been used with little knowledge of the mechanism by which they exert
their effects. Studies on the role of adjuvants on CD4+ T cell responses have shown that different types of adjuvants can polarize
the cytokine response while inducing the same proliferative capacity, specificity and avidity of CD4+ T cells. Such responses
were shown to be independent of the antigen used and genetic background of the host. Would this also be the case for CD8+ T
cells? What is the contribution or importance of providing CD4+ T cell help or toll-like receptor (TLR) ligation in the generation
of these CD8+ T cell responses? Studies have shown that CD8+ T cells are guided differently than CD4+ T cells. In addition, it
was found that the use of different adjuvants can induce the generation of different CTL populations: cells which kill but do
not produce IFN-gamma, cells which do not kill but produce IFN-gamma, and cells which both kill and produce IFN-gamma.
By understanding the extent to which one can guide the T cell responses through the use of adjuvants and appropriate CD4+ T
cell help or TLR agonists, one can improve both vaccine efficacy and safety. These have broad implications not only for vaccine
development, but also in the fields of autoimmunity, transplantation, and tumor biology.
Biography
Dr. Magdalena Tary-Lehmann is an Adjunct Associate Professor of Case Western Reserve University (CASE) Department of
Pathology, Co-Founding Scientist and Chief Scientific Officer for Cellular Technology Limited (CTL). She has published more
than 75 papers in peer-reviewed journals. She provides guidance and oversight for technical operations in the GLP laboratory,
ensuring the ongoing scientific excellence of CTL. Over the past decade, she has worked with clients and regulatory agencies to
develop and validate reference samples and controls for use in regulated immune monitoring assays.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
34
Vaccine Platforms for Lassa Fever
Juan Carlos Zapata
Institute of Human Virology-School of Medicine University of Maryland, USA
Abstract
Lassa Fever (LF) disease is a serious public health problem in West Africa, causing around 300.000 cases per year with a mortality
rate around 1% in the general population and around 20% in hospitalized cases. Ribavirin is the only recommended treatment
but its efficacy is limited when it is administrated in early stages of the infection. In spite of all efforts to produce a vaccine
against Lassa virus, currently there are not licensed products. There are several obstacles that have delayed the development
of a LF vaccine. One of them is related to the high pathogenicity of the virus that limits its manipulation to BSL-4 facilities
and the availability of animal models for vaccine protection trials. Another limitation is the genetic diversity among LASV
strains that will require a vaccine candidate to induce a broad cross-protective immunity. Finally, the lack of knowledge on the
pathogenesis and immune responses critical to LF disease has been an additional constrain. That is how, forty years after LF
disease was described, we are still lacking rapid diagnostics, an effective vaccine, and a clear description of its pathogenesis
and immune-suppression mechanism. However, there are some promising vaccine candidates that are been testing with good
results and seem to be ready for clinical trials. Among them are reassortant viruses, viral vectors, virus-like-particles, and codon
attenuated viruses. Our hope is that those clinical trials will be supported soon with good outcomes for the communities from
those affected countries.
A Broadly Cross-Reactive Anti-Endotoxin Vaccine for the Prevention and Treatment of
Sepsis
Alan Cross
Center for Vaccine Development, University of Maryland School of Medicine, USA
Abstract
Gram-negative bacteria (GNB) are a leading cause of nosocomial infection and sepsis. The mortality remains unacceptably
high. The dramatic increase in multidrug resistant (MDR) GNB has left clinicians with fewer therapeutic options. Targeting
bacteria with antibodies is not subject to or drives the same mechanisms by which bacteria evade antimicrobials and avoids
the risk of immune compromise by modulators that target the host response. A heat-inactivated bacterial vaccine induced
antibodies against a highly conserved region of GNB lipopolysaccharide (LPS) that was highly effective in protecting patients
from lethal GNB sepsis (NEJM 1982; 307:1225). We made a subunit formulation of this vaccine in which the purified LPS of
E. coli O111 (J5 mutant) was detoxified and complexed with outer membrane protein from group B N. meningitides. This vaccine
was highly immunogenic and protective in multiple animal models of sepsis. When given without adjuvants to humans, the
vaccine was well-tolerated but elicited only a modest antibody response. However in a Phase I study with CpG adjuvant there
was enhanced immunogenicity. This vaccine also markedly increased the anti-endotoxin antibody response in the serum and
colostrum of cows. The hyperimmune bovine colostrum (HBC) reduced circulating endotoxin levels in an animal model of
microbial translocation from the gut. Oral administration of HBC has been proposed as therapy for multiple clinical conditions
characterized by “leaky gut”, such as inflammatory bowel disease, HIV infection and after surgery. This vaccine merits further
investigation for the prevention and treatment of GNB infections, including those caused by MDR GNB.
Biography
Dr. Alan Cross, MD is a Professor of Medicine at the University of Maryland School of Medicine and Associate Director for
Adjuvant Biology Research at the Center for Vaccine Development. A graduate of Harvard College and the University of
Pennsylvania School of Medicine, he was a medical research officer at the Walter Reed Army Institute of Research before joining
the faculty at Maryland. He has had a long-standing interest in vaccine development for Gram-negative bacterial infections.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
35
PCSK9 Vaccines – A Novel Immunotherapy for the Treatment of Hypercholesterolemia
Gergana Galabova*, Claudia Juno, Marie Le Bras, and Guenther Staffler
AFFiRiS AG, Austria
Abstract
Background: Elevated low-density lipoprotein cholesterol (LDLc) is associated with an increased risk of atherosclerotic
cardiovascular disease (CVD). The Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) is expressed in the liver and is a key
regulator of the LDL receptor (LDLR)-mediated cholesterol clearance. It circulates in the blood in mature (~62kDa) and furin
cleaved (~55kDa) forms, and both modulate LDLR cell surface expression, and LDLc.
Aim: Development of mono and bi-valent peptide-based active immunotherapy that inhibits all active plasma PCSK9 forms.
Methods: AFFITOPE®s are antigenic peptides that mimic PCSK9 epitopes. They are coupled to carrier-protein and together with
adjuvant administered subcutaneously in mice. Concentration of AFFITOPE®-induced PCSK9 specific antibodies are monitored
by anti-PCSK9 protein ELISA and affinity by Biacore®. Their ability to block PCSK9 furin cleavage is evaluated in vitro and plasma
PCSK9, total cholesterol, and liver LDLR levels are determined. Moreover, lipoprotein profile (VLDL, LDL and HDL) is analyzed.
Results: Anti-PCSK9 vaccines induce multi-functional antibodies able to block the interaction of circulating PCSK9 with the LDLR.
Furthermore, they clear the PCSK9 protein from the circulation, block PCSK9 furin cleavage, and thus abrogate the generation of
the truncated form of PCSK9 (~55kDa). Altogether, these events result in blockage of PCSK9, up-regulation of LDLR levels and
strong LDLc reduction.
Conclusions: The AFFITOPE®-based PCSK9 vaccines are a powerful strategy for the prevention or treatment of LDLc
hypercholesterolemia.
Biography
Dr. Gergana Galabova is a senior scientist at AFFiRiS AG. As such, she is responsible for the development of immunotherapy for
the prevention or treatment of hypercholesterolemia and atherosclerosis. She studied veterinary medicine, and after completing
her PhD she joined the Department of Microbiology, Immunobiology and Genetics, University of Vienna as a Post Doctoral
Research Fellow. Her work was focused on the in vivo and in vitro deciphering of the Raf/Mek/ERK signal transduction cascade,
which alterations are involved in several human malignancies and developmental disorders. Her research culminated in several
publications in high impact factor journals, review and textbook chapter.
A Novel Live, Attenuated Respiratory Syncytial Virus Vaccine with Rationally Designed
Immunogenicity, Stability, and Enhanced Pre-Fusion F Expression
Christopher C. Stobart1,2, Christina A. Rostad1,2, Raymond J. Pickles3, Zunlong Ke1,2, Jia Meng1,2, Kaori Sakamoto4, Anne L
Hotard1,2, Sujin Lee1,2, Pedro A Piedra5,6, Brian Gilbert5, Elizabeth R Wright1,2, and Martin L Moore 1,2*
Department of Pediatrics, Emory University School of Medicine, USA
Children’s Healthcare of Atlanta, USA
3
Department of Microbiology and Immunology, University of North Carolina Chapel Hill, USA
4
Department of Pathology, College of Veterinary Medicine, University of Georgia, USA
5
Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA
6
Department of Pediatrics, Baylor College of Medicine, USA
1
2
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract illness in infants. Live, attenuated RSV vaccines have a track
record of safety in pediatric populations. However, several attenuated candidates have failed due to low immunogenicity and genetic
instability. We hypothesize that live RSV vaccines attenuated based on virulence rather than replication will be more immunogenic. The
RSV nonstructural proteins, NS1 and NS2, counter the innate immune response. We previously reported that codon-deoptimization, a
genetically stable approach, of NS1 and NS2 genes attenuates while enhancing immunogenicity. Here, we generated and characterized a
recombinant RSV vaccine candidate, designated OE4, that expresses codon-deoptimized NS1, NS2, and G genes, has the secreted form
of G ablated, the (small hydrophobic) SH gene deleted, and expresses a novel fusion (F) F gene, line19F-I557V. The line19F-I557V F
conferred thermal stability, high levels of the pre-fusion conformation of F to virions, and enhanced immunogenicity. RSV OE4 was not
attenuated in Vero cells. In two different primary differentiated lung cell culture systems, OE4 was significantly attenuated. Compared
to the prototypical strain RSV A2, OE4 was attenuated in the lungs of BALB/c mice, yet elicited higher neutralizing antibodies than A2.
In cotton rats, OE4 was more attenuated yet more immunogenic than RSV ΔM2-2, a comparator vaccine. These data establish a novel
approach for the development of a RSV live, attenuated vaccines with enhanced immunogenicity, thermal stability, and genetic stability.
Biography
Dr. Martin L. Moore received a PhD in Genetics from the University of Georgia in 2003, studying virology. He was a postdoctoral fellow at
Vanderbilt from 2004 to 2008, working on mouse models of respiratory syncytial virus (RSV) pathogenesis. In 2008, Dr. Moore joined the
faculty of Pediatric Infectious Diseases at Emory University. The main goals of the Moore lab are to elucidate mechanisms of RSV pathogenesis
and advance vaccines for respiratory viruses. Dr. Moore is a co-founder of Meissa Vaccines, Inc., an early stage biotech located in San Francisco.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
36
Identification of Novel Pre-Erythrocytic Malaria Antigens that Enhance Protection Induced
by Circumsporozoite Protein Vaccines
Alexander Pichugin1, Cate Speake2, Robert Morrison2, Stasya Zarling1, Charles Anderson3, Stefan. H. Kappe2, Patrick E.
Duffy3, and Urszula Krzych1*
Walter Reed Army Institute of Research, USA
Seattle Biomedical Research Institute, USA
3
National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA
1
2
Abstract
Malaria vaccine development has been hampered by the limited availability of antigens identified through conventional discovery
approaches and improvements are needed to enhance the efficacy of the leading vaccine candidate, RTS, S that targets the
circumsporozoite protein (CSP) of the infective sporozoite. We undertook a transcriptome-based approach to identify novel preerythrocytic vaccine antigens that enhance CSP-induced immunity. We hypothesized that stage-specific upregulated genes would
enrich for protective vaccine targets, and used tiling microarray to identify P. falciparum genes transcribed at higher levels during liver
stage versus sporozoite or blood stages of development. We prepared DNA vaccines for 21 genes using the predicted orthologues in
P. yoelii and P. berghei, and tested their efficacy using different delivery methods against pre-erythrocytic malaria in rodent models.
Using P. yoelii in BALB/c mice, we found that 16 antigens significantly reduced liver stage parasite burden. In our confirmatory screen
using P. berghei in C57Bl/6 mice, we confirmed 6 antigens that were protective in both models. When tested in a combination vaccine,
2 antigens significantly enhanced the protective efficacy of CSP in both models. Analyses of immune mechanisms that mediated
reduction of parasite burden revealed involvement of CD8 T cells. Based on these observations, transcriptional patterns of Plasmodium
genes can be useful in identifying novel pre-erythrocytic antigens that induce protective immunity alone or in combination with CSP.
Biography
Dr. Urszula Krzych received her PhD degree from Rutgers University and subsequently she trained with the late Eli Sercarz at UCLA.
For the past 2 decades she has been conducting research in malaria at WRAIR, Washington, DC, where she currently is the Chief of the
Department of Cellular Immunology. Her laboratory has been investigating immune mechanisms and particularly the role of CD8 T
cells in protective immunity induced by attenuated Plasmodium sporozoites and other pre-erythrocytic vaccine candidates. She has also
been involved in the analyses of human immune responses induced by many malaria vaccine candidates, including the RTS,S vaccine.
The Human Vaccines Project: Changing the Paradigm of Vaccine R&D
Theodore Schenkelberg1* and Wayne C. Koff2
Human Vaccines Project/West Monument Consulting, USA
Human Vaccines Project/International AIDS Vaccine Initiative, USA
1
2
Abstract
The Human Vaccines Project is a new, large scale, global public-private partnership with the mission to accelerate the development
of vaccines and immunotherapies against major global infectious diseases and cancers by decoding the human immune system.
The Project has been endorsed by 35 of the world’s leading vaccine researchers as potentially “transformative” for human health,
and brings together some of the world’s leading research universities, nonprofits, companies and governments to execute a
ten-year, milestone-driven scientific plan. Research will be carried out by a world class network of scientific hubs located at top
academic research centers globally. Recent breakthroughs in genomics, bioinformatics, systems biology and other disciplines
now allow an understanding of the human immune response that was not possible even five years ago, and the Project seeks
to harness such technologies to transform our approach to vaccine and immunotherapy development. Seeking to decode the
human immune system, the Project’s strategic plan is organized around two primary areas: 1) The Human Immunome Program
which is an ambitious 7-10 year effort to sequence the adaptive components of the immune system across diverse populations
to provide an indispensable “parts list” allowing for the design of highly targeted and safer vaccines/therapies; 2) The Rules of
Immunogenicity Program which is focused on conducting large numbers of small, iterative human clinical research trials to probe
the immune system to systematically solve the key problems impeding vaccine/immunotherapy development. If successful, the
Project has the potential transform global health for a range of pressing diseases.
Biography
Theodore Schenkelberg is co-Founder of the Human Vaccines Project and Principal of West Monument Consulting, a firm focused on
accelerating the development of global health technologies for leading nonprofits. As an R&D Director at the International AIDS Vaccine
Initiative (IAVI) he played a key role in the building of IAVI’s global vaccine design program. Ted’s private sector work includes serving
as a Technology/Health Analyst at Carnegie Capital Asset Management. Ted has an MPH (Infectious Disease) from Johns Hopkins, an
MBA from The University of Chicago, a BA from Grinnell College, and serves on the Board of the African Services Committee.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
37
A Virus-Like Particle (VLP) Respiratory Syncytial Virus Vaccine Composed of Distinct
Conformation of the F Glycoprotein F Affords Complete Protection against RSV Challenge
in a Murine Model of the Disease
Jose M. Galarza1*, Velasco Cimica1, Helene Boigard1, Alexandra Alimova2, and Paul Gottlieb2
TechnoVax, Inc., USA
Department of Pathobiology, Sophie Davis School of Biomedical Education, City College of New York, USA
1
2
Abstract
Respiratory syncytial virus (RSV) causes a wide range of respiratory tract infections, particularly serious or life threatening for
infants and elders, although also a major cause of morbidity in adult and children. The respiratory illnesses provoked by RSV
are of global economical and public health significance. Despite of over five decades of research, a licensed prophylactic vaccine
to prevent RSV infection is not currently available. Early human clinical trials with a formalin inactivated RSV (FI-RSV) vaccine
showed to induce serious adverse reaction following natural RSV infection. This precedent has hampered the development of
a safe and effective RSV vaccine, which should stimulate a balanced immune response characterized by the induction of high
titer of neutralizing antibodies. In this presentation, we will describe a new approach to create respiratory syncytial virus-like
particles (VLPs) that display distinct structural conformations of the F glycoprotein (prefusion and postfusion). Alternative
vaccine compositions are formulated with VLPs displaying I) prefusion; II) postfusion and III) a combination of both, prefusion
and postfusion. We use a murine model of RSV to test the immunogenicity, efficacy and safety of these vaccines. A FI-RSV
vaccine is included in the study as control reference of vaccine-induced disease. We will present results on the magnitude and
type of the immune response elicited by vaccination as well as on the protective efficacy and safety of the VLP vaccines following
RSV challenge. The success of this vaccine offers a promising system to advance in the development of a safe and highly effective
vaccine to prevent RSV infection of various demographics.
The Anti-tumor Effects of the HMGN1 Alarmin
Joost J. Oppenheim1, Poonam Tewary2, Feng Wei1, Xin Chen1, and De Yang2
Laboratory of Molecular Immunoregulation, National Cancer Institute, National Institutes of Health, USA
Leidos Biomedical Research, Inc., USA
1
2
Abstract
Recent studies have identified a group of structurally diverse multifunctional host proteins that are rapidly released following
pathogen challenge or cell injury and are able to both chemotactically recruit and activate dendritic antigen-presenting cells.
These potent immunostimulants, called alarmins, including defensins, cathelicidin (LL37), eosinophil-derived neurotoxin
(EDN), granulysin, high-mobility group box protein 1 (HMGB1) and high mobility group nucleosome binding domain 1
(HMGN1) serve as early warning signals to activate innate and adaptive immune systems. They all are antimicrobial proteins
that also interact with chemokine-like receptors and activating receptors on host cells. For example, some beta defensins, LL37,
HMGB1 and EDN mimic chemokine and cytokine activities by interacting with CCR6, FPRL-1, RAGE and Toll-like receptors
(such as TLR2, TLR4, TLR9) respectively. Defensins (HBD2 and HBD3), like cathelicidin (LL37), by binding to DNA also promote
DNA uptake and activation of TLR9 in plasmacytoid dendritic cells, resulting in markedly augmented production of IFN alpha.
We have chosen to study the adjuvant effects of the potent alarmin HMGN1, which activates the TLR4 pathway. Mice deficient
in HMGN1 were immunodeficient and supported increased growth of EG7 and EL4 tumor cells. Conversely, injection of such
tumor cells transfected to express more HMGN1 into normal mice resulted in greater resistance to tumor growth. We therefore
injected HMGN1 intratumorally (i.t.) into CT26 colon tumors growing subcutaneously to about 5 mm in diameter in normal
mice. This slowed tumor growth and prolonged mouse survival only by about 30%. We therefore treated such mice with a
combination of a suboptimal dose of Cytoxan (Cy, i.p.) and HMGN1 (i.t.). The Cy by itself reduced tumor growth by 76%, but all
the mice eventually succumbed. However, therapy with the combination of Cy and HMGN1 completely cured 80% of the treated
mice. By adding another TLR ligand and we have had similar therapeutic effects in mice with 1cm. diam. tumors. Thus, use of
the HMGN1 as an antitumor vaccine together with other antitumor therapies has promising antitumor effects.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
38
Development of a Malaria Vaccine using an Alphavirus Virus-Like Particle (i-αVLP) Platform
Momoko Ishikawa1, Akane Urakami1, Atsuko Sakurai1, Suzanne Mayer1, Kei Tolliver1, Yevel Flores-Garcia2, Fidel P.
Zavala2, Sachiko Kuno1, Ryuji Ueno1, and Wataru Akahata1*
VLP Therapeutics LLC, USA
Johns Hopkins University, USA
1
2
Abstract
Malaria is one of the most serious public health problems worldwide. However, there is no effective, FDA approved vaccine
available to prevent it. Here we develop a novel virus-like particle (VLP) platform based on alphavirus VLP (i-αVLP), presenting
the malaria circumsporozoite protein (CSP) repeat domain. The proprietary i-αVLP contains antigen insertions into two
different sites of its envelope and has displayed 480 copies of the inserted antigen on each αVLP. The highly symmetrical,
dense array of antigens elicits very strong immune responses. Mice and monkeys immunized with the Plasmodium falciparum
(Pf) CSP inserted i-αVLP produced a high titer of antibodies against CSP. Passive transfer of serum collected from immunized
rhesus macaque’s conferred inhibition of parasite liver stage development in mice exposed to P. berghei expressing a functional
Pf CSP. In addition, the ability of anti-CSP antibody induced by i-αVLP-based vaccine was further assessed by using a mouse
malaria challenge model. The mice immunized with i-αVLP displaying P. yoelii (Py) CSP also showed protection against a Py
sporozoites challenge. These results demonstrate a successful VLP platform accommodating CSP increases immunogenicity,
which can provide sterile protection against malaria infection. VLP Therapeutic is launching a clinical development program for
the product, VLPM01, highlighted by a planned Phase I/IIa clinical trial in 2016.
Biography
Dr. Wataru Akahata completed his Ph.D. from Graduate School, Kyoto University and postdoctoral studies at the Vaccine
Research Center, National Institutes of Health (NIH). He has over 10 years of experience in vaccine development against
emerging infectious diseases and received NIH Director’s award for his work in the development of alphavirus vaccines. Dr.
Akahata is the CEO of VLP Therapeutics, with the mission to develop innovative medical treatments, transform traditional
vaccines and targeted antibody therapies in order to address global unmet medical needs.
The Right Fit: Matching the Vaccine Platform to the Disease Indication
Bertrand Georges, Vyjayanthi Krishnan, James Francis, Jianfeng Zhang, Katie Anderson, Christine Shoemaker, Ray
Feng, and M. Scot Roberts*
Altimmune, Inc., USA
Abstract
Selection of a particular vaccine platform technology can have an important impact on the ability to meet current vaccine
challenges. We are exploiting two independent and complementary platform technologies to develop vaccines and
immunotherapies for influenza, chronic hepatitis B, anthrax and cancer. RespirVecTM is an intranasally administered replicationdeficient adenoviral vector technology that underlies our influenza and anthrax vaccine indications. For these indications, a
strong humoral response is required in order to neutralize the spread of the pathogen or the activity of the toxin, respectively.
Because RespirVec infects cells of the respiratory tract and expresses the vaccine antigen intracellularly, other components of
the immune system may also be activated including T cell, innate immunity, and mucosal immunity as a result of the intranasal
route of delivery. Humoral immunity is expected to play a less important role in the immunotherapy of chronic infections
and cancer. We are developing immunotherapies for the treatment of chronic hepatitis B and cancer that are based on our
Densigen technology platform. DensigensTM are long synthetic peptides that are dense in helper and cytotoxic T cell epitopes to
overcome the HLA restriction issue that has limited peptide-based strategies in the past. Densigens are also modified through
the inclusion of an inert fluorocarbon tail that preclinically facilitated aggregation of the peptide antigens to create a depot-like
effect at the site of injection and enhancing the immunogenicity of the antigens. By including peptide antigens representing
multiple conserved proteins, a broad immune response resistant to immune escape may be achieved.
Biography
Dr. Scot Roberts has nearly 20 years of senior technical leadership experience. He has significant experience in biologics drug
development with a focus on viral vectors and antiviral therapies. Dr. Roberts completed a post-doctoral fellowship at the
National Cancer Institute, Laboratory of Molecular Virology and has numerous patents and publications in peer-reviewed
journals, and has been an invited speaker and Chair at several international conferences. Dr. Roberts received his Ph.D. from
the Johns Hopkins School of Medicine, Department of Pharmacology and Molecular Sciences.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
39
Novel Lentiviral-Based DNA Vaccine for Long-Term Memory Responses Against Persistent
Infectious Diseases
Yahia Chebloune*, Maha Moussa, Simaa Ahmid, Deepanwita Bose, Jean Gagnon and Geraldine Arrode-Bruses
Pathogenesis and Vaccination Lentivirus Laboratory PAVAL Lab. Joseph Fourier University, Grenoble, France
Abstract
Chronic infectious pathogens have adapted strategies to persist and to overcome the host’s immune responses. Classical vaccine
strategies failed to induce protective immunity so that novel strategies are strongly needed. Using the HIV model we developed
an innovative lentivector DNA vaccine that undergoes a single cycle of replication without integration to amplify the antigens
in the vaccinated host. We demonstrated that this vaccine induces potent polyfunctional long lasting immune responses both
in mice (1) and macaques (2). Both vaccine-specific CD4+ and CD8+ T cells were composed of effector and a variety of memory
types of cells that we characterized. We found that memory cells contained a proportion of precursors that have high proliferation capacity (PHPC). Furthermore we found that the long lasting immunity induced by our vaccine prototype and strategy was
associated with induction of antigen-specific T stem cell memory (TSCM). Altogether these data suggested that our lentivector
prototype DNA vaccine and strategy are inducing particular long lasting T cell immune responses that might control virus infection/replication. All 6 vaccine and 6 control macaques were challenged intrarectally with repeated low doses of SIVmac251
and virus infection/replication, T cell counts, physiopathological changes and immune responses were examined during one
year post-challenge. As expected, while the vaccine did not prevent SIVmac acquisition, all vaccinated macaques have lower viral
loads at the peak of viremia, controlled progressively their virus to barely detectable level. Virus control didn’t correlate with
neutralizing antibodies, correlated with increased activated CD8+ but not CD4+ T cells.
Novel Staphylococcal Cell Wall Component Functions as an Antigen Molecule of T cell
Subset: A Valuable Vaccine Candidate for MRSA Infection
Min Ja Lee1, Min Young Seong1, Jong-Ho Lee1, Dong Ho Ahn2, and Bok Luel Lee1*
Host Defense Laborartory, College of Pharmacy, Pusan National University, Korea
Virus Vaccine Team, R&D Center of Green-Cross Corp., Korea
1
2
Abstract
Staphylococcus aureus is one of the most frequent causes of serious infections in humans. Recently, infections caused by
community-associated methicillin-resistant S. aureus (CA-MRSA) have been reported to be increasing in healthy children and
young adults. Since an effective staphylococcal vaccine has not been developed yet, identification of novel staphylococcal vaccine
candidate is urgently needed. Many recent basic studies demonstrated that T-cell-mediated cellular immunity are required
to establish protective immunity against MRSA infection, suggesting that development of host cellular immune responses is
essential for effective protection against S. aureus infection. Recently, we have demonstrated that S. aureus wall component is
recognized by mannose-binding lectin (MBL), a key component of human complement system and human serum antibody.
These studies suggest that staphylococcal cell wall derivatives may induce protective immunity against S. aureus infection. To
test this hypothesis, we purified several staphylococcal cell wall components from S. aureus mutant strains. Intraperitoneal
injection of purified staphylococcal cell wall components into C57BL/6J mice resulted in significantly increased T-cell subsetmediated IL-17A production, promoting neutrophil recruitment and phagocytosis. But, T-cell subset-deficient knock-out mice
did not produce any IL-17A production. Furthermore, three times antigen-immunized mice were protected from re-infection of
MRSA USA 300 strain. Taken together, our studies support that newly identified staphylococcal cell wall component functions
as an antigen molecule of T-cell subset and this component is a novel vaccine candidate against MRSA infection.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
40
Surface-Display Platform for Mucosal Vaccine against Influenza and HPV Infection
Chul-Joong Kim
Chungnam National University, South Korea
Abstract
Because the mucosal area is the major route of entry of most infectious pathogens, immune system consists of an integrated
network of numerous immune cells, tissues and effector molecules to build up the first line of defense against numerous
pathogens. The majority of current vaccination methods with parenteral route only induce the systemic immune response with
a weak or no mucosal immune response. Therefore, new mucosal vaccines with appropriate delivery vehicles to induce both
protective mucosal and systemic immune response are highly desirable. We have intensively investigated Lactic acid bacteria
(LAB) as a delivery vehicle of antigens for prophylactic and therapeutic purposes to mucosal areas. The antigen location is also
important to the efficacy of vaccination along with the route of administration and immunization schedule etc. We have adopted
a membrane bounding protein, poly-γ-glutamic acid synthetase complex A (PgsA) from Bacillus subtilis as an anchoring motif
to express viral antigens on the surface of LAB. Using the surface –display platform, we have developed a mucosal vaccine to
induce specific immune responses to eradicate human papillomavirus type 16 E7-expressing cancer cells from cervical mucosa.
After surface expression of the HPV16 E7 protein on Lactobacillus casei was confirmed by Western blot, flow cytometry and
immunofluorescence microscopy, mice were orally inoculated with L. casei-PgsA-E7. E7-specific serum IgG and mucosal IgA
productions were enhanced after oral administration and significantly enhanced after boosting. Systemic and local cellular
immunities were significantly increased after boosting, as shown by increased counts of lymphocytes and IFN- γ secreting cells
among splenocytes and increased IFN- γ in supernatants of vaginal lymphocytes. Furthermore, in an E7-based mouse tumor
model, animals receiving orally administered L. casei-PgsA-E7 showed reduced tumor size and increased survival rate versus
mice receiving control (L. casei-PgsA) immunization. This platform mucosal vaccine was applied to human Cervical intraepithelial
neoplasia grade 3 (CIN3) patients. Induction of immunological clearance of CIN3 by targeting HPV antigens is a promising
strategy for CIN3 therapy. Cervical lymphocytes (CxLs) and peripheral blood mononuclear cells (PBMCs) were collected and
E7 specific interferon--producing cells were counted (E7 cell-mediated immune responses: E7-CMI) by ELISPOT assay. Most
patients (70%) taking the optimized dose experienced a pathological down-grade to CIN2 at week 9 of treatment. E7-CMI in
CxLs correlated directly with the pathological down-grade. Oral administration of an E7-expressing Lactobacillus-based vaccine
can elicit E7-specific mucosal immunity in the uterine cervical lesions. This is the first report on a correlation between mucosal
E7-CMI in the cervix and clinical response after immunotherapy in human mucosal neoplasia. The development of a universal
influenza vaccine that provides broad cross protection against existing and unforeseen influenza viruses is a critical challenge.
In this study, we constructed and expressed conserved sM2 and HA2 influenza antigens with cholera toxin subunit A1 (CTA1)
on the surface of Lactobacillus casei (pgsA-CTA1sM2HA2/L. casei). Oral and nasal administrations of recombinant L. casei into
mice resulted in high levels of serum immunoglobulin G (IgG) and their isotypes (IgG1 & IgG2a) as well as mucosal IgA. The
mucosal administration of pgsA-CTA1sM2HA2/L. casei may also significantly increase the levels of sM2- or HA2-specific cellmediated immunity because increased release of both IFN-γ and IL-4 was observed. The recombinant pgsA-CTA1sM2HA2/L.
casei provided better protection of BALB/c mice against 10 times the 50% mouse lethal doses (MLD50) of homologous A/
EM/Korea/W149/06 (H5N1) or A/Aquatic bird/Korea/W81/2005 (H5N2) and heterologous A/Puerto Rico/8/34(H1N1), or
A/ Chicken/Korea/116/2004(H9N2) or A/Philippines/2/08(H3N2) viruses, compared with L. casei harboring sM2HA2 and
also the protection was maintained up to seven months after administration. These results indicate that recombinant L. casei
expressing the highly conserved sM2, HA2 of influenza and CTA1 as a mucosal adjuvant could be a potential mucosal vaccine
candidate or tool to protect against divergent influenza viruses for human and animal. Our unique surface-display platform on
LAB was applied to induce mucosal and systemic immune responses as well as cell mediated immune response in preclinical
and human clinical studies including immunotherapy in human mucosal neoplasia. Furthermore, the mucosal administration
of Lactobacillus-based surface-display vaccine may extend to other infectious diseases that develop at mucosal sites including
bowel, bronchial, and oropharyngeal epitheliae as well as metabolic diseases.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
41
Multivalent Adenoviral Vector based Vaccine Technology: Systematic Assessment and
Selection of the Optimal Bivalent Design
Marija Vujadinovic*, Kerstin Wunderlich, Benoit Callendret, Marina Koning, Mark Vermeulen, Esmeralda van der
Helm, Adile Gecgel, Dirk Spek, Karin de Boer, Masha Stalknecht, Maria Garzia Pau, Roland Zahn, Jerome Custers, and
Jort Vellinga
Janssen Infectious Diseases and Vaccines Crucell Holland B.V. (a Janssen company), Netherlands
Abstract
Replication-incompetent adenoviral vectors are currently in clinical development for vaccine and gene therapy applications.
For both purposes it is often necessary to express more than one transgene. In these instances, usage of a mix of the standard
single-transgene adenoviral vector may not be optimal with respect to manufacturing costs and the final dose per vector in
humans. Despite the large variety of bivalent recombinant Adenoviral (rAd) vector technologies described in literature,
it remains unclear whether all strategies are equally suitable for clinical development, while still retaining all the beneficial
properties of the monovalent adenoviral vectors (e.g. immunogenic potency). Here we report that the human Adenovirus 35
and 26 (rHAd35 and 26) vectors expressing multiple antigens from a bidirectional expression cassette in E1 outperformed a
range of tested bivalent vectors. A thorough assessment was performed of different bivalent vectors containing two antigenic
coding sequences: i) in E1 and E3 region, ii) in tandem expression cassettes in E1, iii) fused via a Furin-2A peptide in E1, and
iv) expressed from a bidirectional mCMV promoter in E1, directly compared to the respective monovalent benchmark control
vectors. The vectors were tested for rescue efficiency, genetic stability, transgene expression, and potency to induce an immune
response in mice. Major differences were found between the bivalent E1-E3, E1-E1, E1-F2A, and E1-bidirectional for the tested
parameters. Cumulatively the findings show that the rHAd35 and rHAd26 containing an E1-bidirectional mCMV expression
cassette performed similarly to the monovalent benchmark control and were superior to the other tested bivalent designs.
Biography
Marija Vujadinovic is currently a PhD candidate at the Janssen Infectious Diseases and Vaccines Crucell in Leiden, The Netherlands.
She has completed two Master’s Degrees in Biomolecular Sciences and Management, Policy Analysis & Entrepreneurship at
the Faculty of Life Sciences, Free University in Amsterdam, The Netherlands. During her Master’s traineeship at Janssen she
was offered a position as a PhD candidate on the AdVac®- based vaccine development, which she is currently in the process of
completing.
Immunogenicity and Therapeutic Effects of Rv1419 DNA Vaccine from Mycobacterium
tuberculosis
Xueqiong Wu*, Yan Liang, Xiaoyan Zhang, Li Xiao, Xuejuan Bai, Xiaomei Wang, Junxian Zhang , Yourong Yang , Jinying
Song, and Ni Li
Army Tuberculosis Prevention and Control Key Laboratory, New Tuberculosis Diagnostic and Treatmental Technique Beijing
Key Laboratory, Institute of Tuberculosis Research, the 309th Hospital of Chinese PLA, China
Abstract
The situation of tuberculosis (TB) is very severe in China. New therapeutic agents and regimens to treat tuberculosis are
urgently needed. In this study, a DNA vaccine expressing Mycobacterium tuberculosis (MTB) Rv1419 antigen was constructed
and evaluated. The mice were immunized intramuscularly three times at two-week intervals with saline, plasmid vector pVAX1,
M. vaccae, Ag85A DNA or Rv1419 DNA. Three weeks after the last immunization, higher levels of IFN-γ were observed in the
mice in the Rv1419 DNA group in stimulated spleen lymphocyte cultured supernatant, and a higher ratio of Th1/Th2 cells in
whole blood was observed, suggesting a predominant Th1 immune response. Compared with saline group, injection of MTBinfected mice with Rv1419 DNA reduced live bacterial loads found in lungs and livers all by 0.41 log10. The pathological changes
in the lungs of DNA vaccine treated mice were less, with more limited lesions. These results suggest that Rv1419 DNA was
effective for the treatment of TB, significantly increasing the Th1 cellular immune response, and inhibiting the growth of MTB.
Rv1419 DNA can be used as a new candidate DNA vaccine against TB.
Funding: Special Foundation of China for Serious Infectious Diseases (No.2012ZX10003008-002)
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
42
Track-C
A New Approach on VLP-Based Vaccine Design: Chagas Disease as Test Case
Ibai Lopez-Marijuan1, Sofía M. Alvarez-Rios2, Aritz Durana1,2, and Diego M.A. Guerin 1,2*
Fundación Biofísica Bizkaia – Biofisika Bizkaia Fundazioa, Leioa, Spain
Departamento de Bioquímica y Biología Molecular, UPV/EHU, Leioa, Spain
1
2
Abstract
Chagas disease is a parasitic infection caused by the protozoan Trypanosoma cruzi and no protective or therapeutic vaccine against
Chagas exists. In this work we describe two chimeric vaccines designs based on a VLP from Triatoma virus (Sanchez-Eugenia et
al., 2015. J. Gen. Virol. 96:64). Previous studies showed that T. cruzi exogenous epitopes generate a protective immune response
against Chagas infection (Maranon et al., 2011. Microbes and Infection 13:1025). One vaccine was done 'in silico' trying to mimic
the structural conformation of the two epitopes as displayed in a protein homologous to antigen. For that, we have mutated the
amino acidic sequence of the TrV-VLP in regions where the TrV secondary structure was similar to each of the mimicking native
epitopes. In order to determine how good were the selected substitution places, we have performed an exhaustive analysis
over the mutated residues by estimating to what extent the native conformation was reproduced. This analysis involved H-H
bonds before and after the amino acid substitution, and solvent exposure of each residue of both epitopes. Molecular Dynamic
(MD) simulations where done with the objective of estimating the stability of the epitopes as part of the chimeric vaccine. This
modelization can orient the design by relating "structural flexibility" with "vaccine immunogenicity" (Joshi et al., 2011. Vaccine
29: 9423). The second vaccine was directly constructed in vitro by conjugation of one or more synthetic peptides to the Lysines
exposed on the external VLP surface.
Biography
Dr. Diego Guérin received his PhD in Physics from the Universidad Nacional de la Plata, Argentina (1985). Dr. Guérin took
several postdoctoral and visiting research stays in crystallography at the Purdue University working with Michael Rossmann
(1986-1987), the Pasteur Instutute in Paris (1986 and 1998), and IBMB in Barcelona (2001). In 1988 Dr. Guérin moved to
Argentina where he was Professor at the Universidad Nacional del Sur (UNS), in Bahía Blanca. In 2003 DMAG established in
Spain, and leads a group on Structural Virology at the University of the Vasque Country.
Full Length Single Chain, a Novel GP120-CD4 Fusion HIV Subunit Vaccine, does not Cause
a Deleterious Autoimmune CD4 response in Cynomolgus macaques
Jennifer A. Schwartz1*, Ilia Prado1, Jacqueline Martin1, Gurjinder Jandu1, Wenlei Zhang1, Deborah Weiss2, Jesse Francis3,
Maria Huaman3, Anthony Cristillo3, Ranajit Pal3, Neil Balgobin4, William Follas4, Dan Wierda4, and Timothy R. Fouts1
1
Profectus Biosciences, Inc., USA
2
Bioqual, Inc., USA
3
Advanced Bioscience Laboratory, Inc., USA
4
Xeno Diagnostics, LLC, USA
Abstract
Full length single chain (FLSC) is a novel HIV vaccine that presents conserved CD4i epitopes on HIV envelope. The constrained
structure is achieved with single chain complexes of gp120 and CD4 fragments. FLSC elicits cross-reactive antibodies and heterologous
protection against SHIV/SIV in three independent low-dose rectal challenge studies in rhesus macaques and is being developed as a
subunit vaccine for clinical evaluation. We performed preclinical immunotoxicology studies to assess potential safety issues specifically
derived from a deleterious autoimmune response to CD4. Two studies were performed in cynomolgus macaques. The presence of
deleterious antibody responses to CD4 were assessed by ELISA, CD4+ cell staining, as well as impact to a mixed lymphocyte reaction
(MLR). CD4+ T cell loss and impact to the immune response to KLH were also assessed. In study 1, we verified that depletion of CD4+
cells impacted the induction of primary and secondary KLH-specific IgG and IgM antibody responses, justifying the use of the antibody
response to KLH as an indicator of an autoimmune response to CD4. In study 2, immunization with multiple high doses of FLSC did
not induce an autoimmune response to CD4 that had any deleterious effects in any assays that were employed. Little to no impact was
seen on CD4 binding/function by flow cytometry and MLRs. Therefore, FLSC did not induce any deleterious autoimmune responses
to CD4 and continues to be a promising vaccine candidate for evaluation in a Phase I clinical trial. Supported by: BMGF OPP1017606.
Biography
Dr. Jennifer Schwartz is a scientist experienced in virology and vaccines for infectious diseases. She received her B.S. with honors in
Biological Sciences from Carnegie Mellon University. She earned her M.A., M.Phil. and Ph.D. from Columbia University at the College of
Physicians and Surgeons where she studied the molecular biology of alpha herpesviruses under Dr. Saul Silverstein. Dr. Schwartz further
pursued her scientific career in the development of virus-based vaccine vectors for infectious diseases and cancer at Yale University School
of Medicine under Dr. John Rose. Since 2012 she’s developed vaccines for HIV and influenza at Profectus Biosciences in Baltimore, MD.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
43
Production of Soluble IL-15 Complexes by Pathogen Infections and Adjuvants
Kimberly S. Schluns*, and Scott M. Anthony
Departments of Immunology, The University of Texas MD Anderson Cancer Center, USA
Immunology Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, USA
Abstract
During the steady state, Interleukin (IL)-15 is primarily located either intracellularly or on the cell surface associated to IL15Ra, where it stimulates lymphocyte development and homeostasis via transpresentation. Conversely, soluble (s) IL-15/IL15Ra complexes are generated after total body irradiation (TBI), nonmyeloabablative chemotherapy, or TLR stimulation. This
led us to investigate if sIL-15 complexes are generated in response to other types of immune stimulation. We report here that
sIL-15 complexes are transiently increased in mice in response to IFN-a treatment, anti-CD40 stimulation, and VSV infection.
Interestingly, although CD40 stimulation or VSV infection enhanced systemic IFN-α protein expression, Type I IFN signaling
was not required for the induction of sIL-15 complexes during either event as examined in IFNAR-/- mice. This is in contrast
to the requirement for IFNR signaling in generating sIL-15 complexes after TBI or poly I:C. Overall, these findings indicate
sIL-15 complexes are induced in vivo by both type I IFN dependent and independent pathways. Since IFN-α induces sIL-15
complexes, we also investigated if sIL-15 complexes are induced upon activation of the Stimulator of Interferon Genes (STING)
pathway. Indeed, sIL-15 complexes were increased in serum and splenic homogenates of mice were given STING agonists (c-diGMP) either i.v. or i.p. In addition, STING agonists directly induced sIL-15 complexes in BM-derived DCs. Overall, our studies
demonstrate that production of sIL-15 complexes is a common event occurring during various types of immune activation
and inflammation suggesting sIL-15 complexes contribute to enhanced immune responses during vaccination, infection, and
autoimmunity.
Biography
Dr. Kimberly Schluns, Ph.D. is an Associate Professor at the MD Anderson Cancer Center. Dr. Schluns holds a B.S. in Physiology
from the University of Illinois and a Ph.D. in Cell Biology from Loyola University Chicago. She obtained her postdoctoral
training in the laboratory of Dr. Leo Lefrancois, at the University of Connecticut Health Science Center, where she investigated
the roles of IL-7 and IL-15 in T cell homeostasis. In 2004, she joined the department of Immunology at MD Anderson Cancer
Center. Currently, her lab is elucidating the mechanisms and functions of soluble IL-15 complexes during immunotherapy, viral
infections, and inflammation.
The Characterized Immune Response and the Evaluation Vaccines of EV71 and CA16
Qihan Li*, Longding Liu, Ying Zhang, Jingjing Wang, Chenghong Dong, Lichun Wang, Li Jiang, Yun Liao, Yanchun Che,
Hongling Zhao, Jing Pu, Erxia Yang, Lei Guo, Min Feng, Yan Liang, and Ruiju Jiang
The Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, China
Abstract
Enterovirus 71 (EV71) and Coxsackievirus A16 (CA16) are major cause of hand, foot, and mouth disease (HFMD) in children
and may be fatal. The vaccines against EV71 and CA16 are needed. Different strains of EV71 or CA16 isolated from different
areas of China and respectively selected one strain for the development of the formalin-inactivated EV71 or CA16 vaccines.
In this study, we found that there are many differences in rhesus macaques by comparing the characters of immune response
induced by the two inactivated whole-virus vaccines, including the levels of neutralizing antibodies and interferon (IFN)-γ, and
many immune-associated genes. Most importantly, the immunized macaques by EV71 vaccine were protected against the EV71,
but not CA16. In contrast, CA16 vaccine strongly aggravated the rashes of the immunized macaques the rather than against the
CA16 when viral challenge.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
44
Towards Affordable Conjugate Vaccines: Efficient Chemistry and Low Cost CRM197 Carrier
Protein
Andrew Lees* and Natalia Oganesyan
Fina Biosolutions LLC., USA
Abstract
Conjugate vaccines to prevent Streptococcus pneumoniae, Haemophilus influenzae type b and meningococcal disease have been
very effective at reducing disease. Many additional conjugate vaccines are under development, including ones in diverse areas
such as malaria transmission blocking, anti-cancer, anthrax and drug addiction. Among pediatric vaccines, conjugates are some
of the most complex and expensive vaccines to produce, resulting in a high financial burden, especially for lesser developed
countries and vaccine funders such as the Global Alliance for Vaccines. Efficient manufacturing methodologies can help to
reduce costs and increase vaccine supply. Another costly element of conjugate vaccines is the widely used carrier protein,
CRM197, which traditionally has been produced in Corynebacteria at low yield. Commercially available CRM197 has been too
expensive for many researchers, inhibiting R&D work. We have achieved high expression levels of soluble, properly-folded
CRM197 in E. coli (EcoCRM), resulting in low cost CRM197. “EcoCRM”, economical CRM from E. coli, can reduce the cost of
conjugate vaccine research as well as the price of the clinical product.
Biography
Dr. Andrew Lees is the founder and scientific director of Fina Biosolutions LLC, in Rockville, MD. He is an expert in the
chemistry of conjugate vaccines with over 20 patents to his name and an author on over 60 peer-reviewed articles. Conjugation
chemistry developed by Dr. Lees is used by GSK, the Serum Institute of India, the Chengdu Institute of Biological Products and
others in their S. pneumoniae and meningococcal conjugate vaccines.
Dr. Lees is also an Associate Professor at the Center for Vaccine Development at the University of Maryland, School of Medicine.
Dr. Lees was formerly Director of Protein Purification and Macromolecular Sciences at Biosynexus, Inc and Director of Vaccine
Development at Virion Systems, Inc. Andrew Lees holds a BS in Chemistry from Harvey Mudd College (Claremont, CA) and a
Ph.D. in Biophysics from The Johns Hopkins University (Baltimore, MD).
Automated and Standardized Interpretation of Hemagglutination Assays
Kathy L. Rowlen
InDevR, Inc., USA
Abstract
Cypher One™ is a new, cost-effective tool for rapid automated analysis of hemagglutination (HA) and hemagglutination
inhibition (HI) assays. Until now interpretation of HA and HI assays required specialized expertise to accurately read and
manually record the data, with experienced users often differing in their interpretation of the titer value. The Cypher One
system will not only standardize analysis but also provide a digital, traceable record to enhance data quality and reliability.
Cypher one provides results in less than a minute and has a small footprint for easy incorporation into laboratory workflow.
This presentation will focus on comparison studies conducted in academic, government, and vaccine industry labs. Results that
compare hundreds of HA/HI titer values to those obtained by human experts reading “by eye” demonstrate excellent correlation
to the manual read on both U-bottom and V-bottom plates for multiple blood types.
Biography
Dr. Kathy L. Rowlen has more than 20 years of management and research experience in business and academics, with more than
10 years in the field of biotechnology. Rowlen was a successful Professor of Chemistry at the University of Colorado in Boulder
from 1991 to 2008. After co-founding InDevR in 2003 and serving as its Chief Science Officer, she transitioned to a full-time
position as InDevR’s CEO in 2008. Dr. Rowlen has a well-earned reputation as a resourceful business entrepreneur and has
raised substantial non-dilutive funding. Under her leadership, InDevR has demonstrated excellence in translating innovative
ideas into high-impact commercial products. For example, InDevR developed the Virus Counter® as an innovative new tool for
virus quantification. The product is now being marketed by ViroCyt LLC and nearly every global vaccine producer has at least
one unit. Dr. Rowlen serves on the Board of Directors for InDevR, 2B Technologies, and ViroCyt.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
45
Moving into IND Enabling Studies and Beyond- Using SBIR Grants as Leverage
Daniel H Zimmerman1*, and Kenneth S Rosenthal2
Senior Vice-President Research, Research and Development, CEL-SCI Corporation, USA
Professor of Biomedical Sciences, Director of Microbiology and Immunology, Roseman University of Health Sciences, USA
1
2
Abstract
Over the last 25 years there have been significant major changes in the SBIR programs including eligibility, award amounts, set
asides, options and submission process. This focus is mainly on NIH SBIR grants which have shown major initiatives such as
the Fast track phase I and II programs and most recently a direct to phase II and inclusion of IND enabling studies. The STTR
program resembles the SBIR programs in many ways but there are some significant differences. Previously, SBIR proposals
were reviewed with the same hypothesis-driven mindset as R-01 proposals and the necessary IND enabling and analytical assay
studies were viewed as developmental and inappropriate. The use of the word “development” in an application, even in a SBIR
grant, was considered the kiss of death for funding. Inclusion of IND enabling studies is key to developing a health care product
that are regulated by FDA, a point at which implementation of many SBIR funded concepts became stalled. When seeking an
SBIR, establish a dialogue with the Scientific Review Administrators (SRA), many of whom will take the time be open and willing
to schedule a telephone call or visit. They do not break confidences and they can be very helpful, especially in interpreting the
expectations and comments of the reviewers. Important changes in grant reviews include more extensive guidance from NIH
and expansion of the scoring range, use of Grants.gov for submission portal, shortening and other changes in the application
and use of the common government wide application. The current SBIR program of NIH is much friendlier to small business
than in the earlier days of the program and thanks has to be given to CSR, initiatives of the NIAID SBIR staff and finally the
many SRAs. Suggestions for improvement of grant reviews are made and include requiring all grantees to serve on a study
section for at least one year. This would increase the pool of reviewers and provide insight to the applicants.
Biography
Dr. Daniel H. Zimmerman has been CEL-SCI’s Senior Vice President of Research, Cellular Immunology since June 1998. Prior
to that he held various positions companies. He has been a guest worker at NIH and NCI and collaborated with NIAID and NCI
scientists. He was a Senior Staff Fellow at NIH after graduation from the University of Florida with a Ph.D in Biochemistry.
He has also lectured and taught as an adjunct professor at several local colleges and universities. Dr. Zimmerman has an
extensive background in the biotechnology industry including research and development, product transfer and clinical
practice with expertise in innate and inductive immunology. He has over 30 years industry experience in researching and
developing successfully commercial diagnostic products for HSV, HBV and HIV. He has been awarded grants and contracts
from NIH and DOD, including several phase I and a phase II SBIR grants and currently is PI on a SBIR phase I award for LEAPS
immunomodulation of PGIA model rheumatoid arthritis. He has served as a study section reviewer on sixteen NIH study
sections for SBIR and other types of grant applications.
An IL-12 Adjuvanted HSV-2 DNA Vaccine is More Protective than a Glycoprotein D Subunit
Vaccine in a High-Dose Murine Challenge Model
Kenneth Bagley1*, Wenlei Zhang1, Timothy Fouts1, Hanne Anderson3, and William Halford2
Profectus Biosciences, USA
Southern Illinois University School of Medicine, USA
3
BioQual Inc. USA
1
2
Abstract
HSV-2 causes a serious disease for which an effective vaccine has not been developed. A recombinant glycoprotein D (gD)
vaccine (Herpevac) recently failed to show efficacy in a phase III trial (1). The hypothesis tested under this study was that
an IL-12 adjuvanted DNA vaccine expressing HSV-2 gD would be significantly more protective than a HerpeVac-like vaccine
consisting of recombinant soluble gD protein formulated in an AS04-like adjuvant (e.g. alum + MPL). Mice were immunized
on days 0 and 21 with gD and IL-12 expressing plasmids by electroporation, with a Herpevac-like vaccine or with a potent liveattenuated HSV-2 0ΔNLS vaccine (2). All mice were vaginally challenged on day 81 with 2,000 times the LD50 of HSV-2 MS.
Virus shedding, survival and weight loss / regain were monitored for 30 days post challenge and compared using Mann-Whitney
Rank Sum Tests. Survival was compared using Log Rank tests. The gD DNA vaccine statistically reduced virus shedding, weight
loss and death compared to the HerpeVac-like vaccine. The survival and weight loss profiles of DNA immunized mice were more
similar to those of HSV-2 0ΔNLS vaccinated mice than to HerpeVac-like vaccinated mice. Binding antibody titers to gD did not
correlate with protection, whereas neutralizing titers and IgG1/IgG2A ratios did correlate. CD4+ but not CD8+ cells were also
found to play a substantial role in protection. Conclusions: The gD immunogen delivered as a DNA vaccine was significantly
more protective than the same immunogen delivered as a recombinant protein formulated in an AS04-like adjuvant.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
46
Balance of Cellular and Humoral Immunity Determines the Level of Protection by HIV
Vaccines in Rhesus Macaque Models of HIV Infection
Timothy R. Fouts1*, Kenneth Bagley1, Ilia J. Prado1, Kathryn L. Bobb1, Jennifer A. Schwartz1, Rong Xu1, Robert J.
Zagursky6, Michael A. Egan1, John H. Eldridge1, Celia C. LaBranche8, David C. Montefiori8, Helene Le Buanec5, Daniel
Zagury4, Ranajit Pal3, George N. Pavlakis7, Barbara K. Felber7, Genoveffa Franchini7, Shari Gordon7, Monica Vaccari7,
George K. Lewis2, Anthony L. DeVico2, and Robert C. Gallo2
Profectus Biosciences, Inc., USA
Institute for Human Virology, USA
3
Advanced BioScience Laboratory, Inc., USA
4
NEOVACS S.A., FRANCE
5
Université Paris Diderot, FRANCE
6
Rochester General Hospital, USA
7
National Cancer Institute, USA
8
Duke Human Vaccine Institute, USA
1
2
Abstract
Obtaining a practical, efficacious vaccine against HIV has been a public health priority for over 30 years. A guiding principle
for vaccine design has been that cellular and humoral immunity will work together to provide the strongest level of efficacy.
This concept has been called into question after the failure of several large clinical trials, giving rise to the ideas that vaccineinduced immune responses may not act in concert and might even increase risk by immune activation, thereby providing more
targets for infection. We have addressed this issue through a series of nonhuman primate studies using a selected HIV/SIV
envelope-based subunit antigen, called the Full-length Single Chain (FLSC). The FSLC was developed in order to induce broader
immunity, including against new conserved epitopes, compared to conventional gp120. Here, we tested the FLSC in a variety
of formulations and vaccination regimens that generated a variety of immune response profiles. We used two repeat low dose
challenge models (heterologous SHIV (162P3) and cross-clade SIVmac251) to test for protection in three separate studies.
Under certain conditions, immunization with the FLSC was able to protect against heterologous neutralization resistant
challenge viruses in all three studies. The data show that antibody dependent cellular cytotoxicity (ADCC) is associated with a
reduced rate of acquisition against repeat challenges with either challenge virus provided that the concurrent anti-vaccine T cell
response is minimal. Protection is lost over time as requisite antibody titers declined consistent with the evanescent quality of
anti-envelope humoral responses. Vaccine efficacy was consistently lost in animals exhibiting higher T cell responses that were
predominately single or dual function. These data indicate that in the case of HIV the balance between the humoral immune
response and cellular immune activation is the critical element for generating protective efficacy. Achieving and sustaining such
a balance, while enhancing antibody durability, is a significant challenge for HIV vaccine development.
This work was supported by the following grants from the NIH: AI47490, CA148473, AI091567, AI074334, AI074334; A160481,
HL59796; Henry Jackson Foundation, HJF 715319; and the Bill and Melinda Gates Foundation: BMGF OPP1017606 and
OPP41351.
Biography
Dr. Timothy R. Fouts is one of the founders and principle scientists at Profectus Biosciences. He directs a team of scientists in the
discovery and preclinical development of vaccines, small molecule and antibody based antiviral therapies and microbicides that
are within the Profectus research portfolio, in particular HIV and certain biothreat viruses. Dr. Fouts has more than 32 scientific
publications that have appeared in peer-reviewed journals and book chapters. Dr. Fouts received his Ph.D. in Immunology
from the University of Maryland, Baltimore and did a postdoctoral fellowship at the Aaron Diamond AIDS Research Center at
Rockerfeller University in NYC.
Designing a Staphylococcus aureus Vaccine for a New Century
Ingrid L. Scully*, David Cooper, Robert Donald, Naglaa Mohamed, Li Hao, Alejandra Gurtman, Kathrin U. Jansen, and
Annaliesa S. Anderson
Pfizer Vaccine Research & Development, USA
Abstract
The Staphylococcus aureus four antigen vaccine (SA4Ag), comprised of capsular polysaccharide types 5 and 8 individually
conjugated to CRM197, clumping factor A and manganese transporter component MntC, is designed to prevent invasive
S. aureus disease in patients undergoing elective surgery. Novel bacteriological and immunological approaches were used to
discover and validate the target antigens in SA4Ag. These approaches included both identifying S. aureus virulence factors
involved in enabling invasive disease as well as confirming that SA4Ag antigens induce functional antibodies that are effective
at killing clinical isolates that represent the diversity of invasive disease causing strains in the US. As a result, SA4Ag is now
undergoing clinical studies for the prevention of invasive S. aureus disease.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
47
Research Progress in Immune Escape and Immune Prevention of MRSA Infection
Hao Zeng
National Immune Products Engineering Technology Research Centre, China
Abstract
The pathogen Staphylococcus aureus, a leading cause of human bacterial infections worldwide, is an opportunistic bacterial
pathogen responsible for a diverse spectrum of human diseases which are from mild and common skin and soft tissue infections
to severe, necrotizing and highly invasive disease. Given the high prevalence of S. aureus infections, it is, perhaps, not surprising
that -30% of non-institutionalized individuals are permanently colonized with the organism. Therefore, S. aureus may be
considered a component of normal human flora. These findings are noteworthy because nasal carriage of S. aureus has been
associated with subsequent infection. S. aureus expresses a wide array of secreted and cell surface- associated virulence factors,
including surface proteins that promote adhesion to damaged tissue and to the surface of host cells, that bind proteins in
blood to help evade immune responses, and that promote iron uptake. Most strains express a polysaccharide capsule. The
organism can secrete an array of extracellular enzymes such as proteases, a hyaluronidase, a lipase and a nuclease that facilitate
tissue destruction and spreading, membrane-damaging toxins that cause cytolytic effects on host cells and tissue damage, and
superantigens that contribute to the symptoms of septic shock. S. aureus -Infectious diseases are a leading cause of morbidity
and mortality worldwide and are a major challenge for the biomedical sciences, the development of vaccines and therapeutics
is important, and this requires an understanding of the host immune system. Recently, much progress has been made towards
discovering the mechanisms of S. aureus pathogenesis and host–microbe symbiosis. And knowledge about the immune system
has also been steadily increasing. Yet many challenges remain, perhaps the most daunting being effective vaccine development.
Indeed, it is not known how to elicit protective immunity against S. aureus in a safe and practical manner. To achieve this
and other goals, further developments in basic research are clearly required. So our pathway for developing the novel vaccine
against MRSA is the technique of High-throughput Immunodominant Antigenome. In brief, based on the whole pathogen
genome and bioinformatics, we pick up a large number of potential vaccine candidate to do the proteome array. After identified
the Antigen-Specific Antibody Responses and T cell Responses in MRSA infected patients, we categorize the candidates into
B cell Antigenome and T cell Antigenome. And then select the multiple Immunodominant antigens as the different vaccine
formulation, hope to both enhance the Innate Immune response and induce the strong adapted Immune response to prevent
the MRSA infection.
Biography
Dr. Hao Zeng, M.D, Postgraduate Tutor, Professor of College of Pharmacy, Third Military Medical University (TMMU) been
engaged in immune prevention and genetic diagnosis of pathogenic microorganisms more than fifteen years. As Director
of vaccine R&D of National Immune Products Engineering Technology Research Centre NERCIP, mainly take responsibility
of the research details of genetically engineering vaccine, antibody engineering projects. As the Project Leader, successfully
applied for three "NSFC" projects, one “863” Program and one "national significant new drugs creation" project. The review
expert of CFDA and NSFC, the member of American Association of Immunologists (AAI). Reviewer of IAI, Vaccine, Microbes
Infection International Journal of Nanomedicine, Colloids and Surfaces etc. Military academies Yucai Award (2014), Chinese
Pharmaceutical Association – SANOFI Young biopharmaceuticals Award (2013) "The 11th Five-Year Plan " Technology advanced
individual of TMMU (2011, National first-class new drug license ( S20090002, 2009 ) and the first prize for technical invention
Government of Chongqing (FM080100106, 2009) of "Oral recombinant Helicobacter pylori vaccine".
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
48
Pre-Clinical Toxicology Considerations in Vaccine
Nabil Al-Humadi
U S Food and Drug Administration, USA
Abstract
Toxicology studies are performed to support the nonclinical safety of vaccines prior to their clinical investigation. To ensure
the effectiveness of these studies and integrity of the data collected, careful consideration should be given in any pre-clinical
toxicology study to the collection of precise information which is dependent on proper design of study. Various endpoints
are useful in the assessment of safety. These include infiltration of inflammatory cells at the site of injection, decreased food
consumption, loss of body weight and changes in body temperature findings. These preclinical toxicology endpoints are the
counter-parts for inflammation and pain at the site of injection, malaise, fatigue, and slight febrile responses which may be
produced by vaccines in a clinical study. Thus, proper design and techniques (e.g., sampling sites, type of anaesthesia, etc.) used
in any pre-clinical toxicology study for vaccines is very important. Multiple factors should be taken into consideration when
analysing the final outcome of any toxicology study. Careful considerations should be given to site of blood collection, food
intake, stress, and age-related changes. Sampling sites (tail, foot, eye, and heart) affects the values for murine white blood cell
counts and other hematological parameters. Moderate to severe food restriction causes decreases in reticulocyte, neutrophil,
lymphocyte, and platelet levels in rats. Acute restrain stress decreases monocyte and lymphocyte levels and increases
neutrophil levels in rats. Anaesthesia is another example of factors that may alter the outcome of toxicology studies. Choosing
the type of anaesthesia (CO2, isoflurane, pentobarbital, and ketamine/xylazine) is an important factor influencing the results
of clinical pathology. When compared to CO2, isoflurane, pentobarbital, and ketamine/xylazine caused increase in aspartate
aminotransferase (AST) levels and decrease in total protein, albumin and triglyceride levels. Blood volumes that are drawn on
a weekly or monthly basis from each animal should not affect the clinical pathology results. For example, on a weekly basis,
blood collected from mouse, rat, dogs, monkeys, and rabbits not to exceed 0.075, 1, 50, 10, and 10 mL respectively. Increases in
blood volume collection above this specified volume for each animal species may significantly alter hematology and/or clinical
chemistry parameters levels. Acute phase reactants [APR] (e.g., C-reactive protein, Haptoglobin, fibrinogen, serum amyloid,
albumin, pro-inflammatory cytokines, alpha1-acid glycoprotein (α1AGP), α2-macroglobulin (α-2M), and thiostatin) are used
in the assessment of inflammation. The suitability of each APR as a marker of inflammation depends upon certain criteria.
Additionally, validating sample collection time points for specific protein should be carefully considered. Other factors like the
quality assurance elements (SOPs, instrument validation, lab certification, etc.) should also be considered.
Lessons Learned from Studying Vaccination Administration and Completion Using a Large
Administrative Claims Database
Scott C. Quinlan*, Crystal N. Holick, and Stephan Lanes
HealthCore, Inc., USA
Abstract
Health insurance claims databases are being used to assess vaccine utilization and safety including risks of rare outcomes such as
intussusception following rotavirus vaccination. Database studies are vulnerable to certain potential sources of error, including
misclassification of exposure and outcome information and substantial losses to follow-up. We conducted analyses to assess
certain sources of potential error in studies of rotavirus vaccination. Intussusception risk following rotavirus vaccination has
been found to be preferentially associated with the first dose. Misclassification of dose number or administration date could bias
risk estimates. We conducted a cohort study using the HealthCore Integrated Research Database (HIRD) with medical record
validation to assess the accuracy of claims data regarding rotavirus vaccine dose and administration date. The HIRD includes
longitudinal medical and pharmacy claims data from health plan members across the United States. Our results indicated
that information on vaccine dose and administration date are largely accurate in the HIRD, however there was some degree
of misclassification that needs to be considered when conducting epidemiologic studies of vaccine risks. Additionally, we used
the HIRD to examine rotavirus vaccine completion rates using different methods to handle losses to follow-up. Administrative
claims databases commonly have large losses to follow-up for reasons such as health plan disenrollment. Our investigation
found that using different approaches for addressing losses to follow-up had little impact on rotavirus vaccine completion
rates. In conclusion, claims data have special limitations when used to conduct medical research, but can still provide valid
assessments of vaccine utilization and safety.
Biography
Dr. Scott Quinlan, PhD, MS serves as Lead Epidemiologist/Research Manager for HealthCore, Inc. and leads pharmacoepidemiology
and active monitoring programs in support of medical product safety research studies in a variety of disease and therapeutic
areas including drug-induced liver injury, epilepsy, respiratory disease, infectious disease, and vaccine safety. Dr. Quinlan
received his Bachelor of Science degree in biochemistry from the University of Delaware (Newark, DE) and his Master of Science
and Doctor of Philosophy in epidemiology from The George Washington University (Washington, DC).
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
49
Developing Tuberculosis Vaccines: Current Challenges and Strategies
Lewis Schrager
Aeras, USA
Abstract
Developing new vaccines to prevent tuberculosis (TB) remains a global public health priority. The outcome of the Phase 2b trial
of the MVA85a TB vaccine trial in infants highlighted many of the challenges confronting efforts to develop TB vaccines. This
presentation will review the results of this trial and will describe some of the most important challenges currently confronting
TB vaccine development efforts that were highlighted by the trial outcome. Examples of such challenges include the difficulty
inherent in selecting Mycobacterium tuberculosis (Mtb) antigens and antigen delivery platforms, the preclinical assessment of
vaccine candidate efficacy in animal challenge models, the absence of immunologic correlates of protection against Mtb infection
and active TB disease, and “de-risking” vaccine candidates in earlier stage clinical development prior to the implementation of
later-stage, more resource-intensive clinical trials. Strategies being undertaken to overcome these challenges, including new
approaches to animal challenge studies, efforts to develop a human challenge model of Mtb infection, and new, “proof-ofconcept” clinical trial strategies for TB vaccines will be discussed. The current status of the clinical TB vaccine pipeline also will
be reviewed.
Biography
Dr. Lewis Schrager is Vice President, Scientific Affairs at Aeras, a non-profit biotech with a mission to develop vaccines for
tuberculosis. Dr. Schrager is a graduate of the Johns Hopkins University and the Vanderbilt School of Medicine. He trained in
medicine at the Bellevue Medical Center, NYC, and received his training in infectious diseases at the Harvard School of Medicine
and the Albert Einstein College of Medicine/Montefiore Medical Center. He has served as chief of the Epidemiology Branch,
NIAID, NIH and as a chief of clinical review in the OVRR, CBER, USFDA. Dr. Schrager joined Aeras in September, 2013.
Development of a Live Attenuated Oral Vaccine against Shigellosis and Typhoid Fever
Yun Wu1*, Sumana Chakravarty2, Minglin Li1, Tint Tint Wai1, Dennis J. Kopecko1, Stephen L. Hoffman1,2, B. Kim1,2, and
Lee Sim1
Protein Potential, LLC, USA
Sanaria, Inc. USA
1
2
Abstract
There are over 125 million cases of shigellosis globally; a significant fraction is caused by Shigella sonnei (Ss). We address several
significant vaccine challenges simultaneously: 1) The lack of a licensed vaccine for prevention of morbidity and mortality due to
shigellosis; 2) The need for a multivalent vaccine that simultaneously protects against multiple disease agents, and 3) The need
for an easy-to-administer, child-friendly, safe, oral vaccine vector platform for administration of multiple foreign immunogens
that generates long term efficacy following a rapid immunization regimen and can be distributed without refrigeration. We
have exploited the extensive safety record of the live, oral, attenuated Salmonella typhi vaccine (Ty21a) by utilizing it as a
vector to develop a safe, stable, easily administered combination oral vaccine that simultaneously protects against shigellosis
and typhoid fever. As a first step, we have recombineered the Ss form 1 O antigen gene cluster into Ty21a chromosome, and
created Ty21a-Ss, which stably expresses S. sonnei O antigen. A fully characterized seedbank was used to immunize mice either
intraperitoneally or through mucosal routes. Mice immunized with Ty21a-Ss produced high levels of serum IgG antibodies
against both S. sonnei (53G) and S. typhi, while mice immunized with Ty21a only produced antibodies against S. typhi but not
S. sonnei. We are currently assessing the protective efficacy of Ty21a-Ss by mucosal immunizations followed by challenge with
lethal S. sonnei infection. Our work provides the foundation for a multivalent anti-shigellosis vaccine that will protect against
> 85% of shigellosis worldwide and against typhoid fever.
Biography
Dr. Yun Wu, PhD is a scientist of Protein Potential LLC working on vaccine discovery and development. She has been involved
in multiple projects using live, attenuated bacterial vector platforms, including Ty21a. She is the lead scientist of the Ty21aShigella vaccines, leading the efforts of genetic engineering and assay development for the construction, characterization, and
evaluation of Ty21a-derived multivalent shigellosis vaccine candidates.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
50
Next Generation Alphavirus-based Expression Systems and its Application
Dal Young Kim1*, Svetlana Atasheva1, Elena Frolova1, David WC Beasley2, and Ilya Frolov1
University of Alabama at Birmingham School of Medicine Department of Microbiology, USA
University of Texas Medical Branch at Galveston Microbiology and Immunology Department, USA
1
2
Abstract
Since the development of infectious cDNA clones of viral RNA genomes and the means of delivery of the in vitro-synthesized
RNA into cells, alphaviruses have become an attractive system for expression of heterologous genetic information. Alphaviruses
replicate exclusively in the cytoplasm, and their genetic material cannot recombine with cellular DNA. Alphavirus genomebased, self-replicating RNAs (replicons) are widely used vectors for expression of heterologous proteins. Their current design
relies on replacement of structural genes, encoded by subgenomic RNAs (SG RNA), with heterologous sequences of interest.
The SG RNA is transcribed from a promoter located in the alphavirus-specific RNA replication intermediate and is not further
amplified. In this study, we have applied the accumulated knowledge of the mechanism of alphavirus replication and promoter
structures, in particular, to increase the expression level of heterologous proteins from Venezuelan equine encephalitis virus
(VEEV)-based replicons. During VEEV infection, replication enzymes are produced in excess to RNA replication intermediates,
and a large fraction of them are not involved in RNA synthesis. The newly designed constructs encode SG RNAs, which are not
only transcribed from the SG promoter, but are additionally amplified by the previously underused VEEV replication enzymes.
These replicons produce SG RNAs and encoded proteins of interest 10- to 50-fold more efficiently than those using a traditional
design. A modified replicon encoding West Nile virus (WNV) premembrane and envelope proteins efficiently produced subviral
particles and, after a single immunization, elicited high titers of neutralizing antibodies, which protected mice from lethal
challenge with WNV.
Design and Development of a Novel Vaccine for Protection against Lyme borreliosis
Par Comstedt*, Markus Hanner, Wolfgang Schüler, Andreas Meinke, Robert Schlegl, and Urban Lundberg
Valneva Austria GmbH, Austria
Abstract
Lyme borreliosis (LB) is the most common vector-borne disease in the northern hemisphere and there is no vaccine available for
disease prevention. However, it has been shown that the disease can be averted by immunization with an OspA based vaccine
(LYMErix™). The majority of LB cases in Europe are caused by four different Borrelia species expressing six different OspA
serotypes, whereas in the US only one of these serotypes is present. Outer surface protein A (OspA) is one of the dominant
antigens expressed by the spirochetes when present in the tick vector. Immunization with the C-terminal part of OspA is
sufficient for protection against infection transmitted by Ixodes ticks. In order to target the Borrelia species expressing the
six different OspA serotypes, we have designed a multivalent OspA-based vaccine. The vaccine includes three proteins, each
containing the C-terminal half of two OspA serotypes linked to form a heterodimer. In order to stabilize the C-terminal fragment
and thus preserve important structural epitopes, disulfide bonds were introduced and the immunogenicity increased by addition
of a lipidation signal. Active immunization with the adjuvanted Lyme borreliosis vaccine protected mice from a challenge with
spirochetes expressing either OspA serotype 1, 2 or 5, using infected ticks or in vitro grown bacteria as a challenge. Further
immunological analyses (ELISA, surface binding and growth inhibition) indicated that the vaccine can provide protection
against the majority of human pathogenic Borrelia species. This rational designed OspA-based vaccine is therefore suitable for
global prophylaxis of Lyme borreliosis.
Biography
Dr. Par Comstedt received his PhD in 2008 from the department of molecular biology, Umea University, Sweden. Subsequent
to his work he started at Intercell AG (now Valneva Austria GmbH) in Vienna, as project leader for the Lyme borreliosis vaccine
project. At present he is managing the infectious disease models group part of the preclinical vaccine development department.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
51
Vaccines against HSV-2 and HSV-1 Genital Herpes: Lessons from the Cotton Rat Model
Marina Boukhvalova
Sigmovir Biosystems, Inc., USA
Abstract
In spite of the high health burden of genital herpes, there is still no effective intervention against the disease. The significant
gap in knowledge on genital herpes pathogenesis has been further highlighted by the recent failure of GSK HSV-2 glycoprotein
D vaccine Simplirix™ (gD/AS04) to protect humans against HSV-2 and the surprising finding that the vaccine protected against
HSV-1 genital herpes instead. Our recent work using the cotton rat model demonstrated that gD/AS04 has higher efficacy
against HSV-1 compared to HSV-2 genital herpes in cotton rats as well. Severity of HSV-1 genital herpes was less compared to
HSV-2 in cotton rats, yet the model allowed for comparative evaluation of gD/AS04 immunogenicity and efficacy. Cotton rats
were intramuscularly vaccinated using a prime boost strategy with gD/AS04 (Simplirix™ vaccine), hepatitis B vaccine FENDrix™
(adjuvant control), or partially (or completely) inactivated HSV-2 virus and subsequently challenged intravaginally with HSV2 or HSV-1. gD/AS04 vaccine was immunogenic in cotton rats, induced serum IgG directed against gD-2 and serum HSV-2
neutralizing antibodies, but failed to efficiently protect against HSV-2 disease or to decrease HSV-2 viral load. However, gD/
AS04 significantly reduced vaginal titers of HSV-1 and better protected animals against HSV-1 compared to HSV-2 genital
disease. The latter finding is generally consistent with the clinical outcome of Herpevac trial of Simplirix™. Passive transfer of
serum from gD/AS04-immunized cotton rats also conferred stronger protection against HSV-1 genital disease. These findings
suggest the need for alternative vaccine strategies and the identification of new correlates of protection.
Biography
For the past thirteen years, Dr. Marina Boukhvalova have been working on the pathogenesis of human infectious diseases
in the cotton rat S. hispidus model. This work was first conducted at Virion Systems, Inc., the pioneer in cotton rat model
development, and then at Sigmovir Biosystems, Inc., a company Dr. Boukhvalova co-founded in 2010 after dissolution of Virion
Systems. The main goal of Sigmovir is to conduct translationally-relevant research, predicting efficacy of human vaccines and
therapeutics, and reproducing findings from clinical trials. Genital herpes has become a subject of their teams work several years
ago and results have quickly confirmed translational applicability of the model.
Rapid Viral Detection and Quantification Using Laser Force Analysis
Colin G. Hebert*, Christopher Field, Shweta Krishnan, Katrina Shah, and Sean J. Hart
LumaCyte, LLC., USA
Abstract
The quantification of viral infectivity is an integral step at multiple stages in the process of virally producing recombinant protein,
studying the mechanism of viral infection, and developing vaccines. Accurate measurements of infectivity allow for consistent
infection and expansion, maximum yield, and assurance that time or environmental conditions have not degraded product
quality. Traditional methods of infectivity, including the tissue culture infective dose assay (TCID50) and viral plaque assay, are
slow, labor intensive, and can vary depending upon the skill and experience of the user. Other methods including quantitative
polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA) can be faster but rely on specific molecules
that are often expensive and time-consuming to produce, especially when adapting tests to novel or altered targets. Here we
present the application of laser force analysis for the rapid detection and quantification of viral infection in a label-free and
non-specific manner. LumaCyte’s Radiance™ instrument is an automated high content cell analyzer and sorter that measures
the optical force, size, shape, and deformability of single cells. By measuring these intrinsic properties, differences can be seen
as a result of cell differentiation, chemical or biological exposure, and viral infection. Infected cells can be detected within 24
hours using this instrument. Quantitative infectivity data that correlates with traditional titer methods will be presented and
discussed for several viral systems in the context of improving the development and production of vaccines.
Biography
Dr. Colin Hebert earned his Ph.D. in Bioengineering from the Fischell Department of Bioengineering at the University of
Maryland before receiving a National Research Council postdoctoral fellowship at the U.S. Naval Research Lab (NRL). As a
research scientist at NRL, he helped develop the patented technology behind RadianceTM, LumaCyte’s label-free cell analysis
and sorting platform. Since joining LumaCyte, he has continued to refine LumaCyte’s core technologies while at the same time
collaborating with internal and external researchers to develop and identify new applications that would benefit from a labelfree approach.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
52
Gas Vesicle Nanoparticles for Antigen Display and Vaccine Development
Shiladitya DasSarma*, and Priya DasSarma
University of Maryland School of Medicine, USA
Abstract
Gas vesicle nanoparticles or GVNPs, are hollow, gas-filled organelles that are bounded by an extremely stable protein membrane
produced by some prokaryotic microorganisms. GVNPs are easily purified by flotation from Halobacterium sp. NRC-1 and
shown to be ideal for antigen display and vaccine development, due to their adjuvanting properties, bioengineerability, and
non-toxicity. A large gvp gene cluster encoding more than a dozen proteins has been implicated in their biogenesis, with two
proteins, GvpA and GvpC, constituting a major component of the nanoparticle. GvpC is bound to the exterior surface of the
nanoparticles and promotes growth and stability, and insertion of sequences near the C-terminal region of its gene results in
bioengineered GVNPs displaying the inserted sequence. This system allows the display of antigenic sequences including diverse
peptide and proteins, which have been studied both in vitro and in vivo. The source of antigens displayed on the surface of
GVNPs have thus far included a virus, simian immunodeficiency virus (SIV), two bacteria, the obligate intracellular pathogen
Chlamydia trachomatis and facultative intracellular pathogen Salmonella enterica, and a eukaryote, the parasitic protozoan
Plasmodium falciparum. GVNP-displayed antigenic proteins have ranged from secreted proteins to coat and envelope proteins,
to transcription factors and protection has been tested by challenge in one case. Recently, genetic tools for displaying multiple
antigens and an active enzyme have been developed, which may lead to improved vaccines and other biomedical applications.
Biography
Dr. Shiladitya DasSarma earned his PhD at the Massachusetts Institute of Technology, and received postdoctoral training at
the Massachusetts General Hospital, Harvard Medical School. His career as a professor of microbiology began at the University
of Massachusetts Amherst in 1986 and since 2001 he has been at the University of Maryland in Baltimore. His research has
been continuously funded throughout his career by federal agencies (National Science Foundation, National Institutes of
Health, National Aeronautics and Space Administration) and private foundations (Bill and Melinda Gates Foundation, Henry
M. Jackson Foundation). He is well-known for pioneering work in microbial genomics and biotechnology.
Targeting Intracellular Toll-like and Interleukin Receptor Signaling Pathways for Immune
Modulation and Protection against Lethal Microbial Infection
Greg A. Snyder1,2*, and Lindsey J. Brown1
Institute of Human Virology, School of Medicine, University of Maryland, USA
Department of Microbiology and Immunology, School of Medicine, University of Maryland, USA
1
2
Abstract
Inflammation is a significant factor leading to mortality for a number of diseases. Recent viral pandemics, as well as increasing
prevalence of antimicrobial resistant bacterial strains, elicit lethal inflammation and death, underscoring a global health need
for novel treatment approaches. Conventional approaches have primarily focused on the pathogen for developing highly specialized vaccines and antiviral therapies that target the infectious agent‘s replication or transmission. In contrast, more recent
studies have shown that modulation of Toll-like receptor (TLR) signaling is a novel therapeutic approach to blunt the lethal
inflammatory response. Mice deficient in TLR4 or wild-type mice treated with agents that target TLR4 signaling are protected
against lethal influenza infection. Thus, it may be possible to limit the pathogenic effects of microbial (bacterial and viral) induced lethal inflammation while still achieving pathogen clearance and immunity. We have structurally characterized critical
adaptor TIR signaling molecules MyD88, TIRAP and their interactions with microbial TIR proteins TcpC from uropathogenic
E. coli and TcpB from Brucella melitensis. Targeting intracellular Toll-like and Interleukin-1 receptor signaling pathways may
serve as a novel treatment platform for immune-modulation and protection against lethal inflammation resulting from microbial infection.
Biography
Dr. Greg A. Snyder is an assistant professor at the Institute of Human Virology, University of Maryland Baltimore.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
53
Dose Sparing and Cost Savings with Intradermal Injection
Doug Hostetler
West Pharmaceutical Services, USA
Abstract
The majority of vaccines today are administered by intramuscular or subcutaneous injection with typical dose volumes of
0.5 to 1.0 ml. Due to the cost and cold chain storage requirements, stakeholders are interested in technologies that reduce
both of these factors for large vaccination campaigns. Intradermal injection was first popularized by Dr. Charles Mantoux in
1907 for TB vaccination. Since that time we have learned that the robust immune response within the skin layers are ideal
for vaccination. Intradermal injection using the Mantoux method is difficult to master and requires extensive training and
continued practice to be effective. Evidence of a successful injection is typically determined from the generation of a wheal
or bleb indicating the vaccine has been delivered to the appropriate layer. In practice it has been difficult for novel or nonfrequent injectors to consistently form a bleb indicating a successful ID injection. To solve this challenge, we have developed
an intradermal needle guidance adapter which controls the depth of needle penetration to provide consistent delivery to the
target skin layer. This adapter is easily snapped on to the standard syringe used for ID injection. Clinical studies have shown
significant improvement and consistency of wheal or bleb formation as well as a preference by clinicians for the intradermal
needle adapter. By implementing this technology using best practices in clinical trial design, vaccine manufacturers can easily
assess the viability of ID injection for dose sparing and reduced cold chain requirements.
Biography
Doug Hostetler is a Senior Business Development Manager at West Pharmaceutical Services in Exton, PA. Since joining West
in 2008, he has had responsibilities for business development efforts in analytical laboratory analysis and most currently
pharmaceutical delivery systems. In his current role, Doug work’s with West’s customer base in providing specific device options
and solutions for the various challenges associated with drug product preparation, reconstitution, transfer, and delivery.
Analytical Control Strategy of Bacterial Subunit Vaccines and or third generation Viral
Vaccines
Niranjan M. Kumar
ABS Novacell Biopharmaceuticals, USA
Abstract
Our work in the area of Vaccine Analytical & Manufacturing Strategy discusses both innovative and current product release
methods and stability indicating. This presentation also discusses on novel vaccine manufacturing 2nd and 3rd generation
technologies for viral and bacterial and conjugate vaccines. We will speak on the "Analytical Control Strategy of Bacterial Subunit
Vaccines" and or third generation Viral Vaccines: Best designed Analytical Methods are critical for each unit operations quality,
quantity and sterility of the upstream and downstream manufacturing and fast track vaccine approvals by FDA.
Biography
Dr. Niranjan Kumar holds Ph.D in Biochemistry (Major) and Microbiology (Minor) from IARI, New Delhi and Executive Masters
in Management degree from Wharton Business School, UPenn, PA. Dr. Kumar Worked as Sr. VP, Wockhardt Biotechnology;
Executive Director, Novavax, USA; Dep. Director, Sanofi Pasteur, PA, USA; Group Leader, Wyeth (Pfizer) and Senior Scientist
Merck, PA, USA.
Prior to biopharmaceutical career Dr. Kumar was Asst. Professor of Medicine, SUNY @ Buffalo, NY. Dr. Kumar’s major
contributions are as follows:
1. Contributed as core team to launch Prevnar, Menactra, Pandemic Flu, and First Biosimilar Glargine.
2. Developed and validated number of analytical methods in viral, bacterial and conjugate vaccines.
3. Led teams for US-FDA compliance, quality and authored number of BLAs.
4. Six Sigma Black belt certified in Vaccines Operations and analytical Controls.
5. USP elect Expert Committee (2005-20120) for BBVV.
6. Wrote number of Protocols & reports in Methods and Process Development and Manufacturing Process.
7. Published > 50 peer reviewed Journals and Invited lectures (~200) and authored Book chapters.
8. Developed Novacell for making 3rd generation viral vaccines.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
54
Cytokine Assay (ILs 4, 10, 12, 17, 23, IFN-γ), CD8+&CD4+ in Pre Challenge Balb/c Mice
Vaccinated by the Leishmania major New Vaccine
Afshineh Latifynia¹,²*, Gharagozlou Mohammad Javad³, Niknam Mohammad Hossein¹, Farashi Bonab Samad¹,
Ansaripour Bita¹, Gheflati Zahra¹, Hajjaran Homa4, and Khansari Nematollah¹
¹Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Iran
²Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Iran
³Department of Pathobiology, Faculty of Veterinary Medicine, University of Tehran, Iran
4
Department of Parasitology, School of Public Health, Tehran University of Medical Sciences, Iran
Abstract
Objective: Leishmaniasis, a zoonotic protozoan disease is common in the south and Central America, Asia and Africa
continents where phlebotomus mosquito species transmit the disease between susceptible species. The companion animals
and wild species can act as reservoir and maintain the parasite in the environment. This parasite is seen as amastigote form
in vertebrate animals and promastigote form in the insects. Leishmaniasis affects about 12 million people from 88 countries
and creates a complex and unpleasant disease that is long lasting, hard to treat and sometimes a life threatening disease that
is known as visceral leishmaniasis. For this reason, development of an effective and a safe vaccine that protects the susceptible
species is necessary. According to our previous finding of the author related to the new formulated provisional vaccine, it seems
that a complex humoral and cellular response followed by vaccination to be involved, which required further investigation.
Methods: In the present project, we had six vaccinated groups received either 100 or 200 microgram/0.1ml L. major cocktail
antigen, each of them also received Leishmania + BCG (LB), Leishmania + Teucrium (LT), or Leishmania + BCG + Teucrium
(LBT) groups, and also one control group was considered. After vaccination and booster dose, CD4+ and CD8+ T cells profile of
the lymphoid tissues and serum levels of immune effector cytokines including ILs-4, 10, 12, 17, 23, IFN-gamma were evaluated
and the findings were analyzed statistically.
Results: Considering the six vaccinated groups compared together and normal group : number and size of pulps, percent of
spleen weight /mouse weight, interferon gamma with mean of white pulp size, CD4+ with CD3+,IL- 23 with IFN-γ have had
significant differences. And also without considering two injection doses, and considering to three injection groups: IL-10, lL23, IL-12, CD25+, CD3+&CD4+ have had significant differences which may indicate a satisfactory immune response to the new
formulated L. major antigen as a provisional vaccine.
Evaluation of Serum Levels of IL‐17 and Il‐23 and Spleen White Pulp Changes of BALB/c Mice
Following Administration of New Formulation of Leishmania major Antigen as Provisional
Vaccine
Gharagozlou Mohammad Javad¹*, Latifynia Afshineh²,³*, Vodjgani Mohammad2, Agha Ebrahimi Samani Reza¹,
Charedar Soror4, Bagheri Hadi ¹, and Mohebali Mehdi 4
¹Department of Pathobiology, School of Veterinary Medicine, University of Tehran, Islamic Republic of Iran
²Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Islamic Republic of Iran
³Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Islamic
Republic of Iran
4
Department of Parasitology, School of Public Health, Tehran University of Medical Sciences, Iran
Abstract
Objective: Leishmaniasis is an infectious disease caused by various species of leishmanial parasites including L.major protozoan
that is transmitted to humans by phlebotomus mosquito bites. The prevalence of this disease in the world including Iran is
increasing in almost double every decade. It seems that the best solution is development of an effective vaccine to prevent the
disease.
Method: The experiment was conducted using six groups of female BALB/c mice received 100 or 200 micrograms of cocktail
antigen of L.major adjuvanted with Teucrium polium plant extract, BCG or both nominated as LB (leishmania Ag+BCG),
LTopt(leishmania Ag+Teucrium) and LBT (leishmania Ag+BCG+Teucrium).The first three group and the second three group
received 100 and 200 micrograms of the fortified cocktail Ag respectively .the group seven was received no Ag injection and kept
as control group. To evaluate the immunological responses following vaccination, clinical parameters, spleen changes and serum
levels of IL-17 and IL-23 were considered.
Results: Preliminary results of statistical analysis of the data related to the spleen parameters and serum cytokines levels
indicated significant differences among vaccinated groups either received 100 micrograms or 200 micrograms of the fortified Ag
or vaccinated groups compared to control group. The findings have been in the favor of the new formulated provisional vaccine
as shown also in our previous experiments.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
55
Track-D
Uncovering a Sex Bias in SIV Vaccine-Induced Protection
M. Robert-Guroff1*, I. Tuero1, V. Mohanram1, T. Musich1, L. Miller1, D.A. Vargas-Inchaustegui1, T. Demberg1, D. Venzon1,
I. Kalisz2, V.S. Kalyanaraman2, R. Pal2, M.G. Ferrari2, C. LaBranche3, D.C. Montefiori3, M. Rao4, M. Vaccari1, G. Franchini1,
and S.W. Barnett5
National Cancer Institute, USA
Advanced BioScience Laboratories, Inc., USA
3
Duke University, USA
4
Walter Reed Army Institute of Research, USA
5
Norvartis Vaccines, USA
1
2
Abstract
Here we report for the first time a sex bias in response to a SIV vaccine. Rhesus macaques were primed twice mucosally with
replication-competent Ad-SIV recombinants and boosted twice intramuscularly with SIVmac239 monomeric gp120 or oligomeric
gp140 in MF59 adjuvant. Controls received empty Ad and MF59. Up to 9 weekly intrarectal challenges with low-dose SIVmac251
were administered until macaques became infected. We assessed vaccine-induced systemic and mucosal antibodies and T and B cell
responses. Post-challenge, only one macaque (gp140-immunized) remained uninfected. However, SIV acquisition was significantly
delayed in vaccinated females but not males, correlated with Env-specific IgA in rectal secretions, rectal Env-specific memory B
cells, and PC in rectal tissue. gp140 vaccination was more immunogenic, stimulating elevated binding and functional antibody
activities, bone marrow Env-specific PB/PC, and rectal gp140-specific IgG. However, immunization with gp120, the form of
envelope immunogen used in the RV144 trial, provided more significant acquisition delay. Further over 40 weeks of follow-up, no
gp120 immunized macaques met euthanasia criteria in contrast to 7 gp140-immunized and 2 control animals. Although males had
higher binding antibodies than females, ADCC and ADCP activities were similar. The complex outcomes may reflect differences in
IgG subtypes, Fc glycosylation, Fc-R polymorphisms, and/or the microbiome, key areas for future studies. This first demonstration
of a sex-difference in SIV vaccine-induced protection emphasizes the need for sex-balancing in vaccine trials. Our results highlight
the importance of mucosal immunity and memory B cells at the SIV exposure site for protection.
Biography
Dr. Marjorie Robert-Guroff, Chief, Immune Biology of Retroviral Infection Section, Vaccine Branch, NCI, received her Ph.D. from
Georgetown University (1971). Following postdoctoral training at NCI and the Friedrich Miescher-Institut, Basel, Switzerland, she
returned to NCI where her seroepidemiologic studies linked HTLV with human adult T cell leukemia. She participated in the earliest
investigations on the etiology of AIDS. Her HIV neutralizing antibody research led to continued studies on HIV vaccine development.
She was elected a AAAS Fellow (2011), and cited for HIV/AIDS vaccine development, replicating and non-replicating Ad vectors,
mucosal/systemic vaccination strategies, and neutralizing and non-neutralizing mechanisms of antibody protection.
Novel DNA Vaccine Strategy to Alter HIV Immune Hierarchy
Barbara K. Felber
Vaccine Branch, National Cancer Institute/NIH, USA
Abstract
HIV sequence diversity and potential “decoy” epitopes are hurdles in the development of an effective AIDS vaccine. The HIV
Gag and Env proteins represent key immunogens for vaccine development. To target immune responses towards invariable viral
regions, we engineered gene-based immunogens encoding conserved elements (CE) of HIV-1 selected on the basis of stringent
conservation, functional importance, HLA-independence, and association with immune control and tested the immunogenicity
in macaque model. All CE DNA-vaccinated macaques developed robust CE–specific memory responses with a significant fraction
of cytotoxic T cells. In contrast, upon repeated vaccination with full-length gag DNA, only half of the animals developed T cell
immunity targeting any of the CE. Vaccination with gag DNA was very inefficient in inducing primary CE responses as judged by
the reduced frequency of responders and the restricted number of CE recognized. Interestingly, gag DNA vaccination significantly
boosted the preexisting CE responses, increasing both magnitude and breadth. CE DNA vaccination induces long-lasting CEspecific cytotoxic memory T cell responses that can be rapidly activated and have robust cytotoxic potential. The concept of
combining p24CE and p55gag DNA as candidate vaccine for the induction of efficient and durable T-cell mediated responses is
currently being developed for a clinical trial supported by NIAID/HVTN. The combination of CE and full-length immunogens
provides a novel strategy to allow for the development of robust T cell responses targeting subdominant conserved viral epitopes.
The expanded breadth of the responses could provide an advantage in restricting viral propagation.
Biography
Dr. Barbara K. Felber received her Ph.D. in molecular biology from the University of Berne, Switzerland and has been associated with the NCI/
NIH, since 1980 conducting postdoctoral research and establishing the Human Retrovirus Pathogenesis Section. Her work focuses on the
post-transcriptional mechanisms of gene regulation, the study of pathogenesis of HIV/SIV and the development of DNA vaccine against HIV
both as prophylactic and therapeutic regimen. She has a long-standing interest in the development of the DNA vaccine platform and use of
cytokines in cancer and AIDS. She has published more than 170 papers and has more than 25 issued US and international patents.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
56
Tools to Assess Immunogenicity in the Human Ebola Vaccine Trials
Wayne R. Hogrefe1*, and Rick Nichols2
Focus Clinical Trials, a Q2Solutions Company, USA
NewLink Genetics, USA
1
2
Abstract
As the Ebola pandemic in West Africa has subsided, the ability to measure vaccine efficacy in the field has also diminished. This
will mean an increased reliance on data measuring vaccine immunogenicity and bridging that data to animal data. Several tools
have been developed to assess immunogenicity including quantitative ELISA and neutralization assays. One quantitative ELISA
developed under the direction of USAMRIID/JVAP by Battelle utilizes the Ebola Zaire envelope glycoprotein (GP). The assay has
been used to generate IgG data supporting early phase vaccine clinical trials. The assay is now being validated to support later
phase vaccine trials. The description of the quantitative ELISA and early phase trial data will be presented. Another tool used
to assess post-vaccine immunogenicity is the functional assay plaque reduction neutralization test (PRNT). Several options to
determine post-vaccine virus neutralization have been developed that do not require the use of wild-type Ebola virus in a BSL4 environment. A brief overview of neutralization assay options will be presented; however, a more detailed description of a
PRNT using the rVSV-ZEVOB-GP replication competent vaccine vector will be presented. The PRNT qualification data will be
presented, and where applicable, preliminary post-vaccine data will also be presented.
Biography
For the past 8 years Dr. Wayne R. Hogrefe has directed Focus Clinical Trials business unit, a Q2 Solutions Quintiles Quest Joint
Venture Company, and for the prior 20 years has held senior management positions in laboratory operations, R&D, licensing,
and scientific affairs in other business units within Focus Diagnostics, a Quest Diagnostics company. Dr. Hogrefe is a graduate
of Case Western Reserve University in Experimental Pathology and the University of Dayton in Chemical Technology.
Methodological Challenges in the Clinical Development of HIV Therapeutic Vaccines
Shahin Gharakhanian
Shahin Gharakhanian MD Consulting LLC, Cambridge Innovation Center, USA
Abstract
Infection with Human Immunodeficiency Virus (HIV) is a chronic clinical state where discrete viral replication and homeostatic
proliferation of infected memory T-cells maintain HIV reservoirs. HIV is regarded as a major R&D achievement and collaborative
model. Antiretroviral therapy has averted~6.6 million AIDS-related deaths. Globally, an estimated 35.3million people (32.2–
38.8) are living with HIV. Key R&D considerations for HIV therapeutic vaccines include:
• Validated, robust methods and biomarkers to quantify HIV viral reservoirs, reproducible in clinical trials.
• Standardized requirements for pre-clinical assessment, including for vaccine combinations.
• Clinical trial design sufficiently adaptive and innovative to enable robust phase II evaluations of vaccines and combinations
to achieve an HIV functional cure.
Two approaches are possible: the more empirical advocating more rapid clinical trials in humans, and the more theoretical
designed first of all to complete our mechanistic understanding of the HIV immune response. These approaches currently
confront but should ultimately complement each other.
Biography
Dr. Shahin Gharakhanian MD, specializes in Pharmaceutical Medicine and Infectious Diseases (ID). He trained (Internal
Medicine/ID) in the AP-HP Paris Public Hospitals network (France) one of the largest worldwide, also obtaining degrees in
Public/International Health. Within the pharmaceutical industry, he managed projects for 12 companies from Asia, EU
and the USA, involving 10 brands and four NDA/MAA/new drug launches. Most recent corporate position: Vice-President,
Vertex Pharmaceuticals Inc., Medicines Development Group, Global R&D, Cambridge MA, USA. Currently USA-based at the
Cambridge Innovation Center, he focuses on Therapeutic Vaccines (HBV, HIV), acting as Chief Medical Officer or Executive
Medical Consultant for several biotechnology companies.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
57
Genetic Adjuvants that Improve the T cell and Antibody Response Induced by VirusVectored Vaccines
Peter Johannes Holst
Center for Medical Parasitology, University of Copenhagen, Denmark
Abstract
Virus vectored vaccines encoding the MHC class II associated invariant chain as a genetic adjuvant has shown enhancement
of CD8+ T cell responses towards otherwise poorly immunogenic antigens. We have used adenovirus vaccines encoding the
weakly immunogenic tat, vif, rev and vpr antigens linked to the MHC class II associated invariant chain. We immunized rhesus
macaques intramuscularly and rectally with an SIV vaccine construct that induces strong T cell responses against the weakly
immunogenic tat, vif, rev and vpr antigens. Following repeated low dose intrarectal challenges with SIV mac251, vaccinated
animals demonstrated significantly reduced risk of acquisition (P=0.04, likelihood ratio test for leaky effect), and marked
suppression of early viral replication in animals that became infected. Infected vaccines also had better protection of the CD4+
T cells population, maintenance of naïve CD4+ T cells, and reduced T cell exhaustion. Induction of potent T cell responses
against weakly immunogenic accessory antigens is capable of preventing viral transmission, or markedly diminishing early viral
replication. These results indicate that a vaccine that does not incorporate envelope proteins may be an effective component of
a protective vaccine against HIV.
HIV Vaccine-Elicited Antibody Responses in Infants
Genevieve Giny Fouda
Duke University, Durham, USA
Abstract
Because of stark differences in the adult and infant immune system, it is unclear if HIV vaccine discoveries in adults are applicable
to infants. In the moderately effective adult RV144 vaccine trial, antibodies against the HIV Envelope variable loop 1 and 2 were
associated with decreased risk of HIV acquisition, whereas Envelope-specific IgA responses were associated with an increased
risk of infection. We recently investigated if infant vaccination can induce potentially-protective antibodies using samples
from the completed phase I/IIa pediatric HIV vaccine trial PACTG 230. In this trial, infants born to HIV-infected women were
immunized with a recombinant gp120 vaccine with either MF-59 (Chiron, n= 48), or aluminum hydroxide (VaxGen, n=49) as
adjuvant. At peak immunogenicity, 98% of infants immunized with the Chiron vaccine and 73% of infants immunized with
the VaxGen vaccine had detectable anti-V1V2 IgG responses. This frequency of responders is comparable to 72% response rate
observed in RV144 adult vaccines at peak immunogenicity. Importantly, at peak immunogenicity the magnitude of V1V2 IgG
was 22 times higher in Chiron vaccinated infants than in adult vaccines from the RV144 trial. Binding to a cross-clade panel of
V1V2 constructs demonstrated that the infant vaccine-elicited antibodies were cross-clade reactive. The vaccine-elicited antiV1V2 antibodies were predominantly from the IgG1 subclass, but 47% of vaccinated infants had detectable V1V2-specific IgG3.
No IgG2 or IgG4 responses were detected. Finally, vaccine-elicited Envelope-specific IgA antibodies were rarely detected in
infants. These results demonstrate that HIV vaccination can elicit high levels of potentially-protective antibodies in infants.
Biography
Dr. Genevieve Fouda, MD, PhD is an Assistant Professor in Pediatrics at Duke University. She obtained her Medical degree
from the Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Cameroon and her PhD degree for Georgetown
University, Washington, DC. Her research focuses on the development of immune-based interventions to prevent pediatric
infectious diseases.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
58
Activation of Protective Innate-Adaptive Immunity Duo for Conferring Rapid-SustainedBroad Protection of Vaccines against Pathogens
De-chu Christopher Tang
VaxDome LLC., USA
Abstract
We report that intranasal administration of an E1/E3-defective (∆ E1E3) adenovirus serotype 5 (Ad5)-vectored influenza vaccine
could induce seroconversion in human volunteers without appreciable adverse effects, even in subjects with pre-existing Ad5
immunity. Mice and ferrets were well protected against challenge by a lethal dose of an H5N1 avian influenza virus following
intranasal instillation of an Ad5 vector encoding hemagglutinin (HA) in a single-dose regimen. Moreover, the ∆ E1E3 Ad5
particle itself without transgene could confer rapid-sustained-broad protection against influenza by inducing an anti-influenza
state in a drug-like manner, conceivably by activating a specific arm of innate immunity. An Ad5 vector encoding HA thus
consolidates drug and vaccine into a single package, which allows the Ad5 backbone to induce protective innate immunity capable
of conferring nearly-immediate and prolonged (e.g., 5 hours to 47 days) protection as the first wave against influenza; followed
by HA-mediated adaptive immunity as the second wave before the innate immunity-associated anti-influenza state declines
away. In addition to ∆ E1E3 Ad5’s capacity to rapidly induce an anti-influenza state, an Ad5 vector encoding a bioengineered
Bacillus anthracis protective antigen (PA) could also confer rapid (e.g., 1-2 days) prophylactic or post-exposure anthrax therapy
with synergy to antibiotic treatment in a murine model. Both rabbits and macaques were well protected by an Ad5-PAvectored nasal anthrax vaccine in a single-dose regimen against inhalational anthrax following challenge with a lethal dose of
Bacillus anthracis Ames spores. Overall, the work conceivably would foster the development of a novel noninvasive drug-vaccine
duo platform technology capable of conferring rapid-sustained-broad protection against pathogens with neither the potential
to induce drug resistance nor that to trigger harmful systemic inflammation.
Biography
Dr. De-chu Christopher Tang is the Founder of VaxDome LLC and Vaxin Inc. (Vaxin’s nameplate was changed to Altimmune, Inc.
in 2015). He obtained his PhD in Microbiology from Indiana University in 1989. He carried out his postdoctoral work at Baylor
College of Medicine, Duke University, and University of Texas Southwestern Medical Center. He joined the faculty at University
of Alabama at Birmingham (UAB) in 1994; subsequently founded Vaxin Inc. on UAB campus in 1997; and was responsible for
Vaxin’s daily operation as the Chief Scientific Officer until 2012. Dr. Tang was one of the pioneers during the development
of DNA vaccines, noninvasive skin-patch vaccines, adenovirus-vectored nasal vaccines, adenovirus-vectored poultry vaccines,
as well as the protective innate-adaptive immunity duo platform technology. He received the Wallace H. Coulter Award for
innovation and entrepreneurship in 2000; and Vaxin Inc. was selected as a Tech Museum Awards Laureate in 2007. Dr. Tang
was selected as a Distinguished Overseas Scientist by the South Korea KOFST Brain Pool Program in 2012; subsequently joined
Chung-Ang University and International Vaccine Institute (IVI) in Seoul; and was appointed as a Scientist at IVI after the
Brain Pool Program Award expired in 2013. He founded VaxDome LLC in Birmingham, Alabama, USA in 2014 and moved the
company to Dallas, Texas, USA in 2015.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
59
Universal Influenza Vaccines: Prevention of Infection against Matched and Mismatched
Strains
Harry Kleanthous1*, Timothy Alefantis1, Josh Dinapoli1, Stephen Anderson1, Thorsten Vogel1, Corey J. Crevar2, Donald
M. Carter2, Ray Oomen2, Mark Parrington1, and Ted M. Ross2
Sanofi-Pasteur, Inc., USA
Vaccine and Gene Therapy Institute of Florida, USA
1
2
Abstract
Annual vaccination against seasonal Influenza A and B virus subtypes with well-matched inactivated virus (INV) vaccines are
highly effective against upper respiratory tract (URT) Influenza infection and induced disease. Protection against infection is
thought to be mediated principally by neutralizing antibodies targeting the receptor binding site (RBS) of the hemagglutinin
globular head (HA1). Immune pressure on HA1 results in antigenic drift, necessitating worldwide surveillance with subsequent
WHO recommendations on strain selection for manufacture of forthcoming seasonal influenza vaccines. The development of
Universal Influenza Vaccines (UIV) that could protect against matched as well as drifted or mismatched strains would provide
significant improvement over standard of care (SOC). Additionally, a target product profile that also offers long-lasting immunity
would be a substantial advantage of current annual vaccination practices, potentially enabling year-round manufacture. UIV
that induce both breadth and durability across multiple influenza seasons would be paradigm shifting for the Influenza field
and offer significant health care benefits. Using the H1 subtype as proof of concept (POC), we have built consensus-based,
computationally optimized broadly reactive antigens (COBRAs). These prototypes fold properly and bind conformation-specific
mAbs (HA1 & HA2). Prototype H1N1 HA proteins presented on virus-like particles (VLPs), elicit broadly cross-neutralizing
functional antibodies in multiple species and protect against homologous and heterologous virus challenge. This is the first
report describing the induction of universal, broadly-reactive, protective immunity against H1N1 isolates using a consensusbased HA strategy focusing on the globular head.
Biography
Dr. Harry Kleanthous has over 20 years industry experience in the research & development of recombinant live attenuated and
subunit-based vaccines against viral and bacterial pathogens. He joined Sanofi Pasteur as US Head of Discovery Research in 2008
with responsibility for evaluating and developing novel viral vaccine platforms and delivering novel targets to the Development
pipeline. Previously, Dr. Kleanthous was Vice President of Research at Acambis Inc. (formerly OraVax) with responsibility for
developing a new exploratory portfolio. His research interests are in the field of replication-defective viral vaccine platforms,
targeting Influenza, Flaviviruses and Herpes viruses, as well as their use for foreign antigen delivery.
VaxArray: A New Analytical Tool for Influenza Vaccine Potency Determination
Kathy L. Rowlen
InDevR, Inc., USA
Abstract
The VaxArray Influenza potency assay (formerly Titer on Chip) is a new technique for quantification of influenza hemagglutinin
concentration in both upstream and downstream processes in vaccine development. In order to evaluate the potential of
this new technique, several comparative studies of VaxArray to conventional methods have been conducted. Results from
comparison to single radial immunodiffusion, an enzyme-linked immunosorbent assay, and the purity adjusted bicinchoninic
acid assay will be presented. In general, good correlation is observed between these methods and VaxArray. These results,
combined with the speed of the assay (2 hours), demonstrated multiplexed analysis of all components within a quadrivalent
formulation, and robust response to HA strains over a wide time period, indicate that VaxArray can be used as a reliable and
time-saving alternative potency assay for influenza vaccines.
Biography
Dr. Kathy L. Rowlen has more than 20 years of management and research experience in business and academics, with more than
10 years in the field of biotechnology. Rowlen was a successful Professor of Chemistry at the University of Colorado in Boulder
from 1991 to 2008. After co-founding InDevR in 2003 and serving as its Chief Science Officer, she transitioned to a full-time
position as InDevR’s CEO in 2008. Dr. Rowlen has a well-earned reputation as a resourceful business entrepreneur and has
raised substantial non-dilutive funding. Under her leadership, InDevR has demonstrated excellence in translating innovative
ideas into high-impact commercial products. For example, InDevR developed the Virus Counter® as an innovative new tool for
virus quantification. The product is now being marketed by ViroCyt LLC and nearly every global vaccine producer has at least
one unit. Dr. Rowlen serves on the Board of Directors for InDevR, 2B Technologies, and ViroCyt.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
60
Vaccine Escape of H3N2: Targeting the Best Strain
J. C. Phillips1*, and M. W. Deem2
Rutgers University, USA
Rice University, USA
1
2
Abstract
After peaking ~ 50% in 2007-2009, the vaccine efficiency for H3N2 viruses has dropped steadily, reaching in 2013-2014 ~
15% in the USA. This ongoing failure has not been solved by traditional strain selection methods based on two-dimensional
clustering of antigenicity and numbers of HA1 head sequence mutations in the receptor region 130-230. We have developed
an HA1 epitopic clustering refinement (I) that predicts H3N2 vaccine effectiveness with R2 = 0.76, which is significantly higher
than R2 ~ 0.5 for H3N2 ferret-derived distances in humans. It predicted the failure of vaccines in 2014 based on Texas 2012, and
it predicts a similar failure for H3N2 vaccines based on the 2015 target strain Switzerland 2013. An independent hydropathic
method (II) for calculating glycoprotein shape, more specifically the globular roughness, involves N2 sequences. It describes
H1N1 evolution (1932 – present), and identifies H1N1 swine flu 2007 as dangerous as early as 2003. (I) and (II) combined
indicate that the Switzerland 2013 target strain should be at best only a small improvement on Texas 2012, and could be worse.
Better target strains would be Nebraska or Santa Fe 2014. Both (I) and (II) agree that the efficiency for H3N2 vaccines based
on these improved target strains could recover to be ~ 50%. Both (I) and (II) are predictive and can reduce time delays by years.
It is possible that even higher efficiencies could be achieved by identifying adjuvants for optimizing the hydropathic shape of
sequenced vaccines.
Biography
Dr. J. C. Phillips is a retired theoretical physicist, who spent most of his career at Bell Labs, and was elected to NAS in 1978.
He has solved many “impossible” problems, and currently is using bioinformatic scaling methods to connect changes in amino
acid sequences to changes in protein function. Dr. M. W. Deem works in the area of evolution, immunology, physiology, and
materials. Dr. Deem has developed sequence/structure methods to estimate vaccine effectiveness for influenza and methods to
sculpt the immune system to mitigate immunodominance for dengue fever. Dr. Deem has developed a number of widely used
methods in the nanoporous materials field.
Prospects and Challenges for Delivery of Influenza Vaccines
Sushma Kommareddy
Novartis (NVS) Influenza Vaccines, USA
Abstract
Vaccine delivery using microneedles has emerged as a pain-free delivery system to improve the ease of vaccine administration
while exploiting the active immune cells in the skin to elicit a strong immune response. Lately, a wide range of microneedle
technologies are being researched extensively for delivery of flu vaccine. After overcoming the initial challenges of HA
concentration, we have fabricated flu antigens into micro needles and showed preclinical proof of concept of skin delivery
with immune response comparable to vaccine administered intramuscularly. The microneedle patch technologies evaluated
have shown the influenza antigen to be stable in dried form upon storage at refrigerated conditions with a potential for room
temperature stability. In addition to the advantages of microneedle patch delivery, achieving a room temperature stable product
with improved cross-reactivity and breadth of coverage could be game changing for administration of influenza vaccines.
Biography
Dr. Sushma Kommareddy is a Lab Head in vaccine formulations at Novartis (NVS) Influenza Vaccines, Cambridge, MA.
She has formal training in pharmaceutical sciences and earned her doctoral degree in the area of drug & gene delivery from
Northeastern University, Boston. Dr. Kommareddy has 10 years of experience in product development and characterization of
biopharmaceuticals. She served as Principal Investigator for multiple external collaborations on exploring new technologies for
delivery of vaccines and related biologics. She has authored a number of articles in peer-reviewed journals and book chapters in
research and early development of protein and nucleic acid products.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
61
An Ultra-low Dose Live Influenza Vaccine Enabling Rapid Production
John Robert Coleman
Codagenix, Inc., USA
Abstract
Current seasonal influenza vaccines have suboptimal efficacy (<59%) due to in-season drift of the circulating viruses, and
mutations in the HA antigen from the antiquated egg-based production. Also, the 2009 H1N1 pandemic further highlighted
shortcomings of how influenza vaccines are manufactured (e.g. long production time due to a high dose requirement; sourcing
900 million eggs). These shortcomings have led many in the field, such as the Center for Infectious Disease Research and Policy,
to state that there is a “Compelling Need for Game- Changing Influenza Vaccines” (Osterholm, et al. 2012).We have developed
a new, next generation approach to influenza vaccines. Our core technology, termed Synthetic Attenuated Virus Engineering
(SAVE), utilizes computer-aided gene design and de novo gene synthesis to produce live-attenuated vaccines through a process
called gene “de-optimization”. SAVE Influenza vaccine strains are i) a 100% perfect match to the target strains in each and
every viral antigen, ii) have high efficacy in ferrets at doses that is 2,000 lower than current influenza vaccines, and iii) can be
manufactured efficiently in cells; thereby achieving the goal of a highly effective, very safe Live Attenuated Influenza Vaccine
(LAIV) that could be quickly scaled to generate sufficient doses for an entire nation. Given the perfect antigenic match of all
viral proteins, together with the avoidance of egg induced antigenic mismatches we propose that SAVE-designed LAIVs have a
better capacity to protect against in-season genetic drift than the current standard of care vaccines. Since our vaccine strains
are a perfect antigenic match in all segments, our strains could provide more relevant CD8 T-cell epitopes of the seasonal target
strains than the current inactivated vaccines (“flu shot”) or LAIV (FluMist®). First developed in poliovirus (Coleman, et al. 2008
Science), SAVE has been validated in seven distinct viruses. SAVE was used to yield influenza A strains capable of protecting mice
(Mueller et al., 2010; Yang et al., 2013) and now ferrets, at ultralow doses.
Heterotypic T cell Responses to Inactivated and Live Attenuated Seasonal Influenza
Vaccines
Amritha Ramakrishnan1, Margaret Inokuma2, Shaun Stewart1, Andrew Pekosz1, Lisa P. Jacobson3, Vernon C. Maino2,
and Jay H. Bream1*
Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA
BD Biosciences, USA
3
Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, USA
1
2
Abstract
The effectiveness of both the trivalent influenza vaccine (TIV) and the live attenuated influenza vaccine (LAIV) continues to
spark debate. To facilitate new vaccine strategies, a more complete understanding of the immune response to influenza is
needed. Recent studies suggest that T cells may play an important role in mitigating disease severity during infection and
contribute to enhanced antibody responses to vaccination. We examined the capacity of TIV and LAIV to induce influenzaspecific T cell responses to circulating and novel influenza strains in adults vaccinated in 2006. We used a platform technology
designed to streamline and standardize flow cytometry-based T cell analyses. At baseline we observed broad CD4+ and CD8+ T
cell reactivity against 3 circulating and 2 novel viruses (including 2009 H1N1). Interestingly, the strongest T cell responses were
against the 2009 H1N1 pandemic virus. Meanwhile, serum HI titers showed limited cross-reactivity against non-circulating
strains with the lowest activity against the 2009 H1N1 influenza virus. Regarding vaccine responses, somewhat surprisingly,
we found that both TIV and LAIV induced significant increases in influenza-specific T cell frequency against some but not all
vaccine viruses. This was accompanied by changes in the cytokine secretion profiles of influenza-specific T cells in recipients of
TIV but not LAIV. We also observed increased influenza-specific CD4+ T cells against a novel H1N1 and the 2009 H1N1 virus,
though the latter was not statistically significant. These results provide insight into the cellular response to current vaccine
strategies which should be informative for vaccine development and pandemic preparedness.
Biography
Dr. Jay H. Bream is an Associate Professor in the Department of Molecular Microbiology and Immunology at the Johns Hopkins
Bloomberg School of Public Health. He has broad experience in human and mouse cellular/molecular immunology with a
particular emphasis on cytokine gene regulation and T cell biology. He has a particular interest in infectious diseases, biomarker
discovery, immunogenetics and new technologies to assess human immunity. Dr. Bream is a member of the Johns Hopkins
Centers of Excellence for Influenza Research and Surveillance (JHCEIRS) and the PI of a sub-project focused on influenzaspecific T cell responses to vaccination and infection in the elderly.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
62
Ultra-Sensitive Assays in HIV Vaccine and Cure-Related Research - Assessment of HIV
Nucleotide levels in CD4 T cell subsets from Virologically Suppressed Donors
Catherine N. Kibirige*, Hao Zhang, Hanhvy Bui, and Joseph B. Margolick
Johns Hopkins Bloomberg School of Public Health, USA.
Abstract
Standardized ultra-sensitive HIV assays are needed for HIV vaccine and cure research. We have developed qPCR- and ddPCRbased assays that can detect 3 input copies of HIV-1 nucleic acid with 90 - 95% reliability. We used the assay sequences described
in the Journal of Virology by Brussel et al in 2003, but utilized ZENTM/Iowa doubly quenched probes and Taqman PCR. In our
hands, the best results were achieved by using ddPCR to quantify assay standards and RTqPCR to quantify HIV-1 RNA. With
this approach, we analyzed purified CD4 T cell subsets (CD4+CD27+CD45RA+ naïve, CD4+CD27+CD45RA- central memory,
and CD4+CD27-CD45RA+/- peripheral memory/effector, sorted by flow cytometry) from 18 virologically suppressed (<20
copies HIV-1 RNA/ml plasma by Taqman assay) HIV-infected men in the Baltimore-Washington DC site of the Multicenter
AIDS Cohort Study (MACS). These men were selected as having high or low inflammation (serum IL-6 and C-reactive protein
levels in the highest or lowest quartiles, respectively, in a previous study) at the time point studied. HIV-1 RNA was detected
significantly more frequently in cells from the high inflammation group (21/27; 78%) than in samples from the low inflammation
group (7/27; 26%; p=.0003). Thus, a) the new laboratory-developed assays using qPCR and ddPCR allowed the measurement of
residual HIV-1 RNA in virologically suppressed donors; and b) residual inflammation in men with virologically suppressed HIV
infection correlated with HIV RNA in T cells. Further studies using these assays may help determine if higher cellular HIV levels
are driving or resulting from this residual inflammation.
Biography
Dr. Catherine N. Kibirige, PhD, recently worked as a Research Associate for the Johns Hopkins Bloomberg School of Public
Health. Baltimore, MD, USA. She developed ultrasensitive HIV-1 Total DNA, Integrated DNA, 2-LTR DNA and Total RNA assays.
Previously, she designed ultra-sensitive Total DNA, Integrated DNA and Total RNA qPCR HIV-1 and -2 assays as a consultant
for the U.S. Military’s HIV Research Program, and also worked with BioRad Laboratories on the design and validation of droplet
digital PCR (ddPCR) Total RNA and DNA HIV-1 assays.
Development of High Yield Reassortants for Influenza Type B Viruses and Analysis of their
Gene Compositions
Jianhua Le*, Edward J. Orff, Andrew A. Fulvini, Liling Huang, Shiroh Onodera, Barbara A. Pokorny, Andrew Malewicz,
Patricia Primakov, and Doris J. Bucher
Department of Microbiology and Immunology, New York Medical College, USA
Abstract
A critical step in producing the annual inactivated influenza vaccine is the development of high yield (hy) seed viruses by
reassortment for improved growth in ovo. Although hy reassortants for type A influenza viruses have been developed for many
years, hy B influenza reassortant virus development for vaccine production has proven difficult. In this study, we have developed
fourteen hy influenza type B reassortants as vaccine candidate strains with B/Lee/40 as the donor virus. Upon characterization
by the Influenza Division at the Centers for Disease Control and Prevention (CDC) and the verification of HA by sequencing,
all B reassortants were found to be antigenically indistinguishable from the wild type (wt) parents and suitable for vaccine
production. However, only one hy reassortant seed virus from this group was used by a manufacturer for vaccine production.
In general, hy reassortants showed an increase in hemagglutination (HA) titers over their wt parents by approximately 8 fold
(range 1–32 fold). Gene compositions of the hy B reassortants were analyzed by restriction fragment length polymorphism
(RFLP) and the wt origin of the HA and neuraminidase (NA) were confirmed. However, in contrast to hy A reassortants which
require the M gene (hy donor A/PR/8/34) for high yield, all fourteen hy B reassortants obtained the NP gene from the hy donor
strain (B/Lee/40). The parental source for the remaining genes varied among the hy B reassortants. The results indicate that
the B/Lee/40 NP and PB1 gene segments are important contributors to high yield growth in influenza B reassortant viruses for
both Yamagata and Victoria lineages. The B/Lee/40 PB2 gene along with wt NS gene also contributed to the improved growth
for hy reassortants of Yamagata lineage.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
63
Progress in the Clinical Development of the Plant-made Influenza VLP Vaccines
E. Aubin1*, B.J. Ward2, S. Pillet1, 2, S. Trepanier1, N. Charland1, and N. Landry1
Medicago Inc., Canada
Research Institute of the McGill University Health Centre University, Canada
1
2
Abstract
Poor effectiveness reported for seasonal influenza vaccines reinforces the need for newer technologies such as Medicago’s plantmade virus-like particles (VLPs) to provide better vaccines that match circulating strains. Over the past few years, Medicago
has conducted and initiated nine clinical trials with influenza VLP vaccines against pandemic and seasonal strains. Medicago’s
VLP clinical trials results showed that immunogenicity responses met licensure criteria and cell-mediated immunity studies
demonstrated a strong polyfunctional CD4 T cells response. Memory T cells persisted at least for 6 months after vaccination.
Lastly, analysis indicated that T cell response induced by VLP vaccination is cross-reactive towards heterosubtypic strains, an
important aspect in protection against antigenic drift. Plant-made influenza VLPs have the potential to provide better and
broader immune response that could translate into better efficacy, especially in the elderly population.
Biography
Dr. Eric Aubin has more than 15 years of experience in immunology research and development. His expertise in humoral- and
cell-mediated mechanisms involved in vaccine and inflammatory-solicited immune response conducted him to join Medicago
team in 2012. As Scientist in the Clinical Immunology group, he works at elucidating the mechanisms of action induced by the
vaccines developed by Medicago. He also supports other groups in company to develop new or improved analytical platforms
and actively participates to the development efforts to evaluate and improve the effectiveness of new vaccine products.
Mucosal Vaccination and Checkpoint Modulation for Curative Therapy of HPV Cancers
Todd Bartkowiak1,2, Shailbala Singh1, Guojun Yang1, Gloria Galvan2, Dhwani Haria1, Midan Ai1, James P. Allison1,2,
K. Jagannadha Sastry1,2*, and Michael A. Curran1,2
Department of Immunology, University of Texas MD Anderson Cancer Center, USA
University of Texas Health Science Center at Houston Graduate School of Biomedical Science, USA
1
2
Abstract
High-risk type HPV-16 and HPV-18 are responsible for majority of cervical and oropharyngeal cancers as well as a significant
percentage of other genital cancers. Currently available HPV L1-gene based vaccines are effective in preventing but not treating
existing infections/lesions. The HPV E6 and E7 oncogenes are necessary and sufficient for cellular transformation as well
as tumor formation and thus serve as ideal tumor-antigen targets for specific immunotherapy. We discovered HPV-16 E6/
E7 peptides for which induction of T cell immunity correlated with disease-free survival in patients treated for high-grade
cervical pre-neoplastic lesions. We developed a novel intranasal vaccination strategy delivering the HPV E6/E7 peptides and
alpha-galactocyl ceramide (a-GalCer), a potent agonist ligand to natural killer T cells (NKT) that are effective innate immune
modulators to activate dendritic cells and induce potent adaptive immunity. In a preclinical mouse model this vaccine induced
significant reduction in tumor growth affording prolonged survival. We now report complete and sustained regression of genital
HPV tumors by combining the intranasal E6/E7 peptide and a-GalCer adjuvant vaccination with systemic delivery of an agonistic
antibody to 4-1BB, a T cell co-stimulatory receptor of the tumor necrosis factor receptor superfamily. Significant increase in
CD8 to regulatory T cells ratios along with enhanced tumor-killing potential of CD8 T cells are among the mechanisms for the
efficacy. These data support direct clinical utility of this nasal E6/E7 peptide vaccine with a-GalCer adjuvant and immunotherapy
for directed immune cells trafficking to the tumors and overcoming immunosuppressive tumor microenvironment towards
improved/sustained anti-tumor efficacy.
Biography
Dr. Jagannadha Sastry is a Professor in the Department of immunology at The University of Texas MD Anderson Cancer Center.
His research over the past 25+ years concentrated on designing candidate vaccines against HIV and HPV with more recent
focus on screening adjuvants for mucosal vaccination strategies in mice as well as Indian-origin rhesus macaques. Dr. Sastry’s
group identified and patented the HPV peptides proposed for use in the therapeutic vaccine tested in the mouse models. His
group published on the discovery of these peptides from studies employing patient samples as well as subsequent mouse model
studies demonstrating their immunogenicity.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
64
Novel IL-4R Antagonist and IL-13RA2 Adjuvanted HIV Vaccines Can Induce Excellent High
Quality T and B cell Immunity
Charani Ranasinghe*, Zheyi Li, Alice Ng, Matthew Worly, Shubanshi Trivedi, and Ronald Jackson
John Curtin School of Medical Research, The Australian National University, Australia
Abstract
Our studies demonstrate that efficacy of a viral vector-based prime-boost immunization is strongly influenced by the cytokine
milieu at the priming vaccination site, where endogenous IL-4/IL-13 is detrimental to the avidity of HIV specific CD8 T cells.
We have designed two novel HIV vaccines that co-express i) IL-13Rα2 which can transiently sequester IL-13, and ii) an IL4R antagonist that can bind to IL-4 type I and II receptors with high affinity, and transiently prevent the signalling of both
IL-4 and IL-13 activity at the vaccination site, by modulating cells at the innate immune compartment 24h post vaccination.
Using intranasal recombinant fowl pox prime, intramuscular Modified Vaccinia Ankara virus booster followed by gp140 Env
protein booster strategy, the novel vaccines were shown to induce not only high avidity poly-functional mucosal/systemic gagspecific CD8 T cell immunity but also elevated gag and env-specific B cell immunity compared to the unadjuvanted vaccine.
Unlike IL-13Rα2 adjuvanted strategy, the IL-4R antagonist vaccine strategy was able to induce not only excellent sustained
p55gag-specific IgG1 immunity but also IgG2a antibody differentiation. Collectively, the immune responses observed with IL4R antagonist adjuvanted vaccine strategy was similar to what has been reported in HIV elite controllers (i.e. high avidity CD8
T cells and gag-specific B cell class switching) and also the responders of the RV144 trail (i.e. elevated Env-specific antibodies).
Thus, this IL-4R antagonist “triple action” vaccine platform sets us apart from other HIV vaccines and offers good hope a future
HIV vaccine.
Biography
Dr. Charani Ranasinghe is the Leader of the Molecular Mucosal Vaccine Immunology laboratory at the JCSMR, ANU. For the
last decade she has tried to understand, why systemic HIV vaccines are failing, “how and why” mucosal vaccination can induce
better protective efficacy. She was the first to discover that IL-13 plays an important role in dampening CD8 T cell avidity in a
vaccine route dependent manner. Recently, her team has developed two novel IL-4R antagonist and IL-13Ra2 adjuvanted HIV
vaccine platforms that can induce high quality systemic/mucosal HIV-specific CD8 T & B cell immunity.
Advanced Wildlife Biosurveillance or Vaccinate the Camels Already!: Epidemic Preparedness
at the Human/Animal Interface
Amanda Arnold
Arizona State University, USA
Abstract
From MERS to Chikungunya, zoonotic disease outbreaks are ravaging food and wildlife animal populations and posing
significant epidemic threat to humans.
We will start with a brief evaluation of what we learned from the Ebola Outbreak, which originated from a bat bite of a 2-year
old boy in a small village in Guinea, and will review two areas of priority action distilled from numerous expert reports and
published over the last decade.
The two areas of priority action to be explored include:
1) Advancing biosurveillance towards modeling capability for epidemic prediction; and
2) Developing animal vaccinations as a critical barrier to zoonotic disease transfer and outbreak in humans.
This discussion will conclude with relevant and actionable federal policy recommendations that can be taken immediately to
affect epidemic preparedness at the human/animal interface.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
65
Track-E
Accelerated Development of an Ebola Vaccine Based on Replication Incompetent Ad26- and
Modified Vaccinia Ankara-Based Vectors
Benoit Callendret
Crucell Holland BV, a Janssen Pharmaceutical Company of Johnson & Johnson, Leiden, The Netherlands
Abstract
The current Ebola outbreak in West Africa has already caused more than 28,000 cases of Ebola Viral disease (EVD) and claimed
more than 11,000 deaths. As such it is by far the largest and deadliest outbreak of the disease since its discovery in 1976.
In response to the epidemic, Johnson & Johnson accelerated the development of an Ebola vaccine regimen using a primeboost combination of two different components, namely Ad26.ZEBOV and MVA-BN-Filo, that are based on Janssen’s AdVac®
technology and the MVA-BN® technology from Bavarian Nordic, respectively. Both vaccine components have key characteristics
that make them suitable for deployment in Africa, including high capacity manufacturing and stability at temperatures compatible
with existing vaccine distribution channels. Furthermore, heterologous prime/boost vaccine approaches are promising, as they
may induce a robust and sustained immune response, providing strong and long term protection against EVD, which might be
of critical importance to prevent chronic resurgence in the affected region, or to prevent future outbreaks. Proof-of-concept
was achieved in a highly stringent animal model using the combination of the Ad26 and MVA vectors expressing an Ebola Zaire
glycoprotein and this vaccine regimen has been rapidly advanced into clinical Phase 1 evaluation. Preliminary results from
ongoing Phase 1 studies showed that both vaccines are safe and well-tolerated, and induced a robust and balanced humoral
and cellular immune response. This Ad26/MVA prime-boost combination has advanced into clinical Phase 2 evaluation, and
preparations to test this vaccine regimen in affected countries and for licensure are ongoing.
Progress Towards the Development of a Chlamydial Vaccine for Koalas
Peter Timms
University of the Sunshine Coast, Australia and Institute of Health and Biomedical Innovation, Queensland University of
Technology, Australia
Abstract
Wild koala populations continue to experience serious declines as a result of factors including, (i) loss of habitat, (ii) motor
vehicle trauma; (iii) dog attacks; (iv) chlamydial disease. Chlamydial infections have been associated with diseases ranging
from ocular disease leading to blindness, as well as urinary and genital tract disease. Modeling shows that targeting chlamydial
disease would have the greatest potential impact on stabilizing population decline. In the first trial we vaccinated groups of
captive healthy koalas via the sc route, using the chlamydial MOMP antigen. We observed good serum and vaginal secretion
antibodies as well as specific lymphocyte proliferation responses. In the next trial we utilised a recombinant MOMP protein,
cloned from a C. pecorum koala isolate. We vaccinated two groups of koalas, (i) wild caught animals that were clinically healthy
and Chlamydia PCR negative, (ii) wild caught animals with no signs of disease. Following vaccination, there was no increase
in inflammatory pathological changes in any animals. Strong antibody (including neutralizing antibodies) and lymphocyte
proliferation responses occurred in all vaccinated koalas. So far, we have shown that a multi-subunit chlamydial vaccine can be
safely administered to both healthy koalas as well as koalas that have a previous or current chlamydial infection. Specific antiMOMP antibodies are produced at high levels and, importantly, these antibodies are neutralizing in vitro.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
66
Identification of a Basis for the Impaired Responses to Bacterial Polysaccharide Vaccines
in Young Children
Gregory S. Dickinson1, John F. Kearney2, and Kishore R. Alugupalli1*
Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, USA
Department of Microbiology, University of Alabama at Birmingham, USA
1
2
Abstract
Unlike human adults or adult mice, young children or young mice respond poorly to bacterial polysaccharide vaccines. In mice,
B1b lymphocytes are the major responders to a variety of bacterial polysaccharides. Despite having B1b cells, young mice are
impaired in responding to these polysaccharides, suggesting that B cells in the young are qualitatively distinct from those in
adults. B lymphopoeisis early in life is largely Interleukin-7 (IL-7)-independent, while in adults it is IL-7-dependent. B cells
generated early in life exhibit a biased expression of D proximal VH genes therefore comprised of a limited pre-immune B cell
antigen receptor repertoire. Since IL-7- mediated signals are necessary for the efficient expression of VH gene families that are
distal to D such as VHJ558, which are associated with extensive BCR diversity, we hypothesized that B cells generated in the
absence of IL-7 do not recognize a variety of antigens including bacterial polysaccharides. In support of this we have shown
that similar to young mice, adult mice deficient in IL-7 failed to generate a protective antibody response to all the 23 serotypespecific pneumococcal polysaccharides in the PNEUMOVAX®23 vaccine as well as TyphimVi®, a polysaccharide vaccine against
Salmonella enterica serovar Typhi. We found that an enforced expression of IL-7 in young mice promotes efficient antibody
responses to both PNEUMOVAX®23 as well as TyphimVi® that is comparable to adult wildtype mice. Numerous microorganisms
including Enterobacter cloacae express α1-3 dextran and in mice B1b cells generate rapid and long-lasting antibody responses
to α1-3 dextran. Immunization of heat-killed E. cloacae induced a robust anti-α1-3 dextran response in adult but not in the
young mice. To test whether the diminished response to bacterial polysaccharides in the young mice is as result of a limitation
in the availability of antigen-specific B cell precursors, we have immunized VHJ558 transgenic mice, which recognizes α1-3
dextran. We found that the VH J558 transgenic mice generate a robust anti-α1,3 dextran response even in infant mice and
bypassed the IL-7 dependent B lymphopoiesis. These results indicate the inability to generate a response to wide range of
bacterial polysaccharides early in life is due to a limitation in the B cell antigen receptor repertoire.
Viral Vector-Based Prime/Boost Vaccination to Maximize Tissue-Resident Memory T Cell
Responses in the Cervicovaginal Mucosa
Nicolas Cuburu 1*, Geoffrey Guittard2, Cynthia D Thompson1, Christopher B Buck1, Douglas R Lowy1, Barney S Graham3, and
John T Schiller 1
Laboratory of Cellular Oncology, National Cancer Institute / CCR / NIH, USA
Laboratory of Cellular and Molecular Biology, National Cancer Institute, NIH, USA
3
National Institute of Allergy and Infectious Diseases, VRC, NIH, USA
1
2
Abstract
It was recently discovered that a population of tissue resident memory CD8+ T cells can confer immediate and enhanced protection
compared to their circulating counterpart. In this study, we have evaluated in mice whether prime/boost vaccination regimen
via heterologous routes could maximize the number of tissue resident memory CD8+ T cells in the female genital tract and confer
enhanced protection against a genital infection. We used non-replicating adenovirus (Ad) and human papillomavirus (HPV)
vectors given intramuscular and intravaginal, respectively. The order of immunization was critical to maximize the induction
of both circulating and genital CD8+ T cells. Intramuscular priming with Ad vectors followed by a single intravaginal boost with
HPV vectors was the best regimen to induce high numbers of antigen-specific intraepithelial CD8+ T cells with a tissue resident
memory phenotype. Importantly, a massive expansion of genital CD8+ T cells resulted from a combination of recruitment, in
situ proliferation, and differentiation of CD8+ T cells in the cervicovaginal mucosa, as demonstrated by confocal microscopy and
adoptive transfer assays. Expansion required expression of the cognate antigen by cervicovaginal keratinocytes and occurred in
the absence of CD4+ T cells. Finally, sequential intramuscular prime/intravaginal boost immunization conferred CD8-mediated
protection against vaginal challenge with vaccinia virus that was superior to other immunization regimen. In conclusion,
vaccination against sexually transmitted infections might be improved with viral vectors targeting antigen expression and thereby
T cells to the cervicovaginal epithelium.
Biography
Dr. Nicolas Çuburu received a Bachelor degree in Biochemistry and Molecular Biology from the University of Aix-Marseille
(France). He completed a PhD in Immunology and Microbiology at the University of Nice-Sophia Antipolis (France) under the
supervision of Dr. Cecil Czerkinsky and Paul Hofman at the Inserm Laboratory of Mucosal Immunity and Vaccination. He joined
the International Vaccine Institute in Seoul (South Korea) as a postdoctoral fellow to work on vaccine immunomonitoring.
Dr. Çuburu is currently a Research Fellow in John Schiller’s and Douglas Lowy ‘s laboratory of the National Cancer Institute in
Bethesda, where he is developing new methods for genetic immunization with viral vector against sexually transmitted infections.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
67
Role of Toll-Like Receptor Signaling in Regulating Adaptive Immunity against a Candidate
Live Attenuated West Nile virus Vaccine Strain Infection
Tian Wang
Department of Microbiology & Immunology, Department of Pathology, Sealy Center for Vaccine Development, University of
Texas Medical Branch, USA
Abstract
West Nile virus (WNV), a mosquito-borne flavivirus, has induced acute viral encephalitis and neurological sequelae in North
America for more than a decade. One major risk factor for fatality due to WNV infection in humans is aging. No human
vaccines are currently available. Development of safe and effective vaccines, in particular for the potentially susceptible elderly
population, remains as a high priority. The nonstructural (NS) 4B protein of flaviviruses has been associated with evasion of
host immune responses and viral replication. We have previously identified an attenuated mutant with a P38G substitution
in NS4B protein by utilizing site-directed mutagenesis of a wild-type WNV NY99 infectious clone. Upon infection in immune
cells, the mutant produced more viral RNA than wild-type WNV but no detectable infectious virus and subsequently boosted
more potent cell intrinsic innate cytokine responses. The mutant also induced stronger T cell responses than wild-type WNV,
and protected mice from lethal wild-type WNV infection, which together suggest that the NS4B-P38G mutant has suitable
features for an ideal vaccine candidate. Toll like receptor (TLR) s play an important role in the regulation of innate and adaptive
immunity to viral infection. To understand the underlying immune mechanisms, by using young and aged mouse models of
WNV infection, we have further investigated the role of TLR 7-mediated signaling pathways in development of protective
adaptive immune responses to WNV NS4B-P38G mutant infection.
Biography
Dr. Tian Wang is a Professor of Departments of Microbiology/Immunology and Pathology at the University of Texas Medical
Branch (UTMB). She received her Ph.D. degree in Immunology from UTMB in 2000 and later completed a Postdoctoral training
in infectious disease at Yale University. From 2005-2008, she was an Assistant Professor in the Department of Microbiology,
Immunology and Pathology at Colorado State University. In 2008, she joined the faculty at UTMB. Dr. Wang’s primary field
of interest is viral immunology with an emphasis on understanding immune response to West Nile virus infection in host
protection and viral pathogenesis.
Live Attenuated Shigella Vaccine Development at WRAIR
Malabi M. Venkatesan
Bacterial Diseases Branch, Walter Reed Army Institute of Research, USA
Abstract
Recent epidemiological studies have underscored the importance of Shigella causing moderate to severe diarrhea in endemic
countries of Asia and Africa where adequate resources for clean water and food is lacking. The hardest hit are children less
than 5 years of age who suffer the consequences of chronic exposure to diarrheal pathogens by acquiring poor physical growth
and lower cognitive abilities as adults (GEMS study, 2013). While antibiotics are available they are not desirable due to the
widespread multi-drug resistance phenomena among most clinical Shigella isolates. Several strategies are being used to develop
Shigella vaccine candidates that include live attenuated and whole cell-killed products as well as novel protein and carbohydratebased (O-antigen) vaccine candidates. At WRAIR, the approach for live vaccine development is currently based on loss of
VirG(IcsA) that plays a critical role in pathogenesis by enabling Shigella to spread intercellularly. Loss of VirG(IcsA) results in loss
of spreading ability and consequently, significant attenuation of the ensuing disease. Studies with first and second-generation
virG(IcsA)-based Shigella vaccine candidates will be discussed against a backdrop of other promising approaches to vaccine
development. The views expressed are those of the author and do not necessarily reflect the official policy or position of the
Department of the Army, Department of Defense nor the U.S. Government.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
68
Deciphering Maternal Transfer of Immunity for Optimization of RSV Vaccination in
Future Mothers
Jorge C. G. Blanco1*, Lioubov M. Pletneva1, Raymonde O. Oue1, Mira C. Patel1,2, and Marina S. Boukhvalova1
Sigmovir Biosystems Inc., USA
University of Maryland School of Medicine, USA
1
2
Abstract
Respiratory Syncytial Virus (RSV) is the leading cause of pneumonia and bronchiolitis in infants, resulting in significant morbidity and mortality worldwide. There is currently no RSV vaccine. Although maternal serum antibodies against RSV are efficiently transferred through placenta protecting human infants from RSV-induced disease, this protection is short-lived and the
methods for extending and augmenting protection are not known. The objective of this study was to develop an animal model
of maternal RSV vaccination using the Sigmodon hispidus cotton rat. Naïve or RSV-primed female cotton rats were inoculated
with live RSV and set in breeding pairs. Antibody transfer to the litters was quantified and the offspring were challenged with
RSV at different ages for analysis of protection against viral replication and lung inflammation. There was a strong correlation
between RSV-neutralizing antibody (NA) titers in cotton rat mothers and their pups, which also correlated with protection of
litters against virus challenge. Passive protection was short-lived and strongly reduced in animals at 4 weeks after birth. Protection of litters was significantly enhanced by inoculating mothers parenterally with live RSV and inversely correlated with the
expression of lung cytokines and pathology. Importantly, vaccination and boosting of naïve mothers with the live RSV produced
the highest levels of NAs. We conclude that maternal vaccination against RSV in the cotton rat can be used to define vaccine
preparations that could improve preexistent immunity and induce subsequent transfer of efficient immunity to infants.
Immunological Analyses of Antisera with Specificity for a Model Recombinant Protein
Antigen Reveal a Different Linear Epitope Antibody Repertoire Resulting from
Administration of Alhydrogel Formulations Compared to Squalene Formulations
Michael J. Lacy*, and Thomas Hickey
Emergent BioSolutions, USA
Abstract
Polyclonal antibody responses to an Alhydrogel formulation of a recombinant protein antigen, detected more linear peptides
than a squalene formulation. Hyperimmune antisera were generated in mice by IP administration of recombinant Protective
Antigen (rPA) of Bacillus anthracis formulated with Alhydrogel, and recognized 7 linear peptides of the PA molecule. In contrast
to multiple linear peptides, the mouse antibody response to rPA formulated in a squalene emulsion recognized only one linear
peptide. Addition of a TLR3 agonist but not a TLR7 agonist increased slightly the number of peptides recognized. Both agonists
increased the IgG anti-PA response. All antisera were tested after adjustment to 2mcg/mL IgG anti-PA concentration. Antibody
responses to the non-recombinant anthrax vaccine BioThrax™, were confined to the same 7 linear peptides recognized by antirPA mouse antisera. An ELISA was utilized to obtain these data, consisting of biotinylated 20 amino acid peptides tethered
to ELISA plates using hypoglycosylated avidin. The single peptide recognized by antisera obtained after delivery of squalene
formulations of recombinant protein antigen PA was recognized in common by mouse, guinea pig, human and non-human
primate antisera, suggesting a “pan-species” epitope. Therefore, Alhydrogel may increase accessibility of linear portions of a
protein molecule compared to squalene, thereby enlarging the epitope repertoire without penalty to the total antibody response.
Biography
Dr. Michael J. Lacy received a Ph.D. degree in Immunology from the Microbiology Department of the University of Illinois
at Urbana-Champaign, performed postdoctoral research in the BioChemistry Department there, publishing on DNA
autoimmunity, antibody characterization within a fluorescein hapten model, and mouse T cell receptor genetic diversity. During
employment by the former Ribi ImmunoChem later Corixa Corporation, research focused upon melanoma immunotherapy and
characterization of biological activities of TLR-4 agonists including natural LPS, natural monophosphoryl lipid A, and synthetic
lipid A derivatives. More recently at Emergent BioSolutions, investigations have included modified vaccinia Ankara, anthrax
vaccines and TLR9 agonist CPG 7909.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
69
Evaluation of Ebolavirus Glycoprotein Fc Fusion Protein as a Subunit Vaccine
Krishnamurthy Konduru* and Gerardo Kaplan
Center for Biologics Evaluation and Research, Food and Drug Administration, USA
Abstract
Ebola virus (EBOV), a member of the Filoviridae that can cause severe hemorrhagic fever in humans and nonhuman primates,
poses a significant threat to the public health. Currently, there are no licensed vaccines or therapeutics to prevent and treat
EBOV infection. Several vaccines based on the EBOV glycoprotein (GP) are under development, including vectored, virus-like
particles, and protein-based vaccines. Here we describe the development of an EBOV subunit vaccine based on GP fused to the
Fc fragment of human IgG. We expressed the extracellular domain of the Zaire ebolavirus (EBOV) GP fused to the Fc fragment
of human IgG1 (EBOVgp-Fc) in mammalian cells and showed that GP undergoes the complex furin cleavage and processing
as observed in the membrane-bound GP. In vaccinated mice, EBOVgp-Fc induced high levels of total and neutralizing antiGP antibodies against replication-competent vesicular stomatitis virus (VSV)-G deleted recombinant VSV containing EBOV
GP. Vaccinated guinea pigs developed a strong humoral response as measured by total and neutralizing antibodies to GP, and
completely protected against lethal challenge with EBOV. We are currently extending our studies to the monkey challenge
model. Our preliminary data indicates that EBOVgp-Fc induces neutralizing antibodies and recall CD8+ T-cell immunity, as
assessed by the production of IFNγ and TNFα in PBMC activated with EBOVgp-Fc. Further experiments, including protection
against lethal challenge in monkeys, will be required to determine whether Filovirus GP-Fc fusion proteins could be developed
as a candidate vaccine.
Working Together to Advance Vaccinomics in Efficacy and Safety Studies
Linda McKibben
Food and Drug Administration/CBER/OVRR, USA
Abstract
The purpose of this interactive session is to explore with session participants what could be a successful model for incorporating
genomic data within studies of safety and efficacy for vaccines. The goal is to protect more patients and healthy populations
against serious morbidity or mortality related to vaccine-preventable infectious diseases or vaccine toxicity. Approximately 120
genomic biomarkers are included within FDA-approved drug (non-vaccine) labels, but no genetic biomarkers are identified to
predict rare, serious adverse effects after vaccination. (A sub-category of pharmacogenomics is vaccinomics). A preliminary
literature review implicates interwoven contextual factors as barriers to vaccinomic innovation. These are: (1) the complexity
of translating molecular and cellular immunogenomics responses to vaccinomic products for humans; and (2) the perceived
ambiguous investment value of translational research within an empirical, one-size-fits-all paradigm of vaccine development.
Other authors suggest alternative factors for consideration by regulatory authorities: the regulatory framework may be
inadequately prepared, or regulatory science may be insufficiently innovative. FDA’s existing framework for pharmacogenomics
is delineated in published guidances for industry with two main strategies: (1) incorporate existing pharmacogenomic research
into pre- and post-market FDA regulatory activities (e.g., in approved product labels); or (2) set up a mechanism for industry
to voluntarily submit pharmacogenomic data for FDA to review in the pre-market setting. What innovative regulatory or other
strategies could be promising for vaccinomics.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
70
Developing Human and Human-Like Antibodies against Pathogens and Toxins for
Diagnostics and Therapy
Michael Hust
Technische Universität Braunschweig, Germany
Abstract
Antibody phage display is an important technology to generate antibodies against pathogens and toxins. In this presentation,
our pipeline for the development of human and human-like antibodies for diagnostics and therapy will be outlined. Examples
of our pipeline will be given for the development of antibodies for diagnostic and neutralization of Alphaviruses (Venezuelan
equine encephalitis virus and Western equine encephalitis virus) as well as for the development of protective therapeutic
antibodies against botulinum toxin A, B and E, as well as against toxins of Clostridium difficil.
Biography
Dr. Michael Hust studied biology at the Carl von Ossietzky Universität in Oldenburg, Germany, from 1993-1999. He received
his PhD from the Leibniz Universität in Hannover, Germany, in 2002. Since end of 2002 he is working as group leader at
the Technische Universität Braunschweig, Germany. In 2011, he finished his professorial dissertation (Habilitation, venia
legendi for Biotechnology) and was appointed as Privatdozent (PD). In 2014 he was appointed as extraordinary professor
(außerplanmäßige Professur) for biotechnology. He published more than 85 articles (including book chapters and reviews)
on antibody engineering and phage display. He is the scientific coordinator of the EC FP7 funded project AntibotABE (www.
antibotabe.com). He is working on the development of human and human-like antibodies for proteome research, diagnostics
and therapy. Another field of work is the identification of immunogenic proteins of pathogens using phage display. He cofounded the mAb-Factory GmbH in 2007 and the Yumab GmbH in 2012.
Neutralizing Recombinant Human Antibodies against the Vaccinia Virus D8 Protein
Derived From an Immunized Immunoglobulin Library
Ulrike S. Diesterbeck1,2*, Andre Frenzel3, Henrike Ahsendorf1, Thomas Schirrmann3, Stefan Dubel3, and Claus-Peter
Czerny1
Department of Animal Sciences, Georg-August-University Goettingen, Germany
Laboratory of Viral Diseases, NIAID, NIH, USA
3
Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Germany
1
2
Abstract
Despite the eradication of smallpox by active immunization using vaccinia virus (VACV), various reported side effects and its
use as biological weapon as well as zoonotic poxvirus infections raises the need for new treatment options. Beside the antiviral
ST-246, anti-VACV immunoglobulins prepared from sera of vaccinated humans are still the only treatment of orthopox virus
(OPXV) infections. For the development of OPXV specific IgG based human monoclonal recombinant antibodies, 4 donors
were vaccinated via scarification with “Dryvax” (Wyeth). The constructed single chain fragment variable (scFv) library resulted
in a diversity of >4x108. Immunoscreening against VACV Elstree revealed a predominant selection of scFv-clones specifically
binding to the 32 kDa VACV-D8 envelope protein. The scFv α-VACV1.2.2.H9 was characterized in detail. For comparison, it was
converted into the larger scFv-Fc as well as IgG1 format. Using C-terminally truncated D8 proteins, the epitope was mapped to
the N-terminal ectodomain. The scFv-H9 and scFv-Fc-H9 showed similar affinities per antigen binding site of 1.61 nM and 7.68
nM, respectively, whereas the full size antibody was much more affine (43.82 pM). None of the recombinant H9 antibodies was
able to neutralize 100 pfu of VACV Munich 1 in vitro. However, neutralization by scFv-Fc-H9 and scFv-IgG1-H9 was achieved
with the addition of 1% human complement. The calculated EC50 was 0.0776 µM and 0.0077 µM, respectively. In an in vivo
NMRI-mouse-model the purified scFv α-VACV1.2.2.H9 neutralized 4LD50 VACV Munich 1. Four out of six mice passively
immunized intra-peritoneally with 100 µg scFv-H9 survived until day 21 p. inf.
Biography
Dr. Ulrike Diesterbeck studied Veterinary Medicine at the University of Veterinary Medicine Hanover in Germany from 19982004. After receiving PhD in 2006, she worked with Prof. Dr. Czerny at the University of Göttingen and successfully constructed
and selected a human anti-vaccinia virus (VACV) library. In addition, she studied immunoglobulin genetics of horses and cattle
aiming at the construction of animal specific recombinant antibody libraries. Since 2013, Ulrike is a postdoctoral fellow in the
group of Dr. Moss at the NIH in Bethesda. Her current project focuses on the structural analysis of the VACV entry-fusion complex.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
71
Novel Nucleic Acid-Based Immunoprophylaxis Technologies
Daniel Wattendorf
DARPA Biological Technologies Office, USA
Abstract
DARPA is investing in rapid immunoprophylaxis approaches through (1) DNA- and RNA-based vaccine antigen gene delivery
platforms, (2) DNA- and RNA-based passive antibody gene transfer platforms, and (3) rapid discovery of human-derived
potent antibodies. Nucleic acid-based vaccines demonstrate increased effectiveness by encoding the antigen and enhancing
the robustness of the immune response. Nucleic acid-encoded antibody gene transfer bypasses the adaptive immune response
by instructing human cells in vivo to begin producing potent antibodies immediately following injected delivery. Antibodies
to be encoded are selected from available monoclonal antibody clinical candidates. Alternatively, in the case of an emerging
disease, antibodies are selected from human convalescent sera using an array of novel discovery and engineering technologies
to identify the most efficacious, potent, and safe antibodies in a matter of days to weeks. These platforms will decrease the
outbreak response timeline, position new platforms for pharmaceutical investment, and, given their synthetic design, can be
manufactured rapidly at low cost for population distribution.
With successful development and commercialization, these nucleic acid-based platforms can be used to produce
immunoprophylaxis drug candidates for emerging pathogens, natural or engineered, more rapidly than current methods. The
expected outcomes for this DARPA effort include:
- Novel platform(s) for in vivo gene transfer and rapid antibody discovery
- Direct pathogen sequence-to-vaccine production
- Rapid and scalable manufacture
- Increased efficacy
- Streamlined path to clinic
Biography
Col Daniel J. Wattendorf, MC, USAF has been a DARPA Program Manager since 2010. His interests focus on applying
methodological advances in genomics and biotechnology to optimize health and prevent disease, including rapid diagnostics,
new RNA vaccines, and novel immunoprophylaxis strategies to better outpace the spread of infectious disease. In addition to
his DARPA programs, Dr. Wattendorf is a clinical geneticist at the National Naval Medical Center, Walter Reed National Military
Medical Center and the Cancer Genetics Branch, National Cancer Institute, NIH.
Site-specific Immune Requirements for Vaccine-mediated Protection and Immunopathology
against Staphylococcus aureus
Hatice Karauzum*, Christian Haudenschild, Mahta Mahmoudieh, and Sandip K. Datta
National Institutes of Health, NIAID, USA
Abstract
Despite the identification of numerous candidate antigens and vaccination strategies in promising pre-clinical studies, the
failure of a number of these strategies in clinical trials has revealed the need to better understand the requirements for protective
immunity against Staphylococcus aureus. We have previously published the use of a gamma-irradiated, whole cell, killed S. aureus
vaccine to elicit antibody- and Th17-mediated immunity against a model of cutaneous S. aureus infection in mice. We have now
used this vaccination strategy to further elucidate the requirements for a protective adaptive immune response at different
sites of infection. In our current studies, we find that our irradiated vaccine fails to protect against a mouse model of S. aureus
bacteremia but instead elicits a detrimental CD4 T cell and IFNg response that promotes mortality. Furthermore, we have begun
to identify the components of this whole cell vaccine that contribute to these context-dependent protective and detrimental
responses. These findings may be relevant to understanding the increased mortality seen in subjects vaccinated against S. aureus
in a recent clinical trial, and emphasize the need to better understand site-specific immune requirements to design a safe and
protective vaccine against S. aureus.
Biography
Dr. Hatice Karauzum earned her PhD in Microbiology from the University of Basel in Switzerland, where she evaluated the
impact of drug resistance on the human pathogen S. aureus and its virulence in localized and systemic infections. During her
postdoctoral research as a Scientist I and later as a Principal Scientist at the biotechnology company Integrated BioTherapeutics
Inc. in Gaithersburg, MD she managed the design and development of novel vaccine candidates and therapeutics and evaluated
their immunogenicity and efficacy in various experimental models of infection. Currently, as a Research Fellow at the NIH, her
research focuses on the immune responses towards S. aureus that develop during infection and during vaccination.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
72
Effective Global Pandemic Indications, Warning and Response Systems
Ivan Ruzic
BehaviorMatrix, LLC., USA
Abstract
The unprecedented global growth in the adoption of sophisticated and inexpensive mobile technology provides a global, “always
on” capability that can serve as an invaluable component in Global Pandemic Indications, Warning and Response Systems.
Today’s fast, wireless networks cover well over 60 percent of the world population, and cell phone users read over 90 percent of
their text messages compared to less than 25 percent of their emails. Therefore, mobile phones and wireless communications
present significant opportunities for healthcare, economic and other forms of development, and SMS text messaging (including
tweets) has become the most effective ways to communicate important news. As we found during the recent Ebola outbreak
in West Africa, the ability to process text and social media messages in real-time is an invaluable diagnostic aid in identifying
the “what, where, how and when” of potential symptoms and vectors. However, when combined with new technological and
methodological breakthroughs that allow us to “get inside the mind” of the senders of these messages, we are now also able
to predict and shape individual and group behaviors with unprecedented accuracy. These exciting new capabilities will have
profound implications for all stakeholders, and will guide the new age of innovation in execution of the “ground game” that will
determine the effectiveness of a global response.
Biography
Dr. Ivan Ruzic has over 25 years of business leadership and technology industry experience. A long-time resident of the United
States, he is a global citizen who has worked in 38 countries during the course of his career – many in the developing world. He
has been the CEO of four different companies and is now the President at BehaviorMatrix, a signal processing company that has
developed unique technology for identifying and scoring emotion in digital content. An academic early in his career, Ivan holds
a Chemistry and Biochemistry B.Sc. as well as a Ph.D. from Monash University, Melbourne, Australia.
Peptide-Based and Virus-Like Particle Therapeutic Vaccines for Allergic and Autoimmune
Diseases
Zhikang Peng
University of Manitoba, Canada
Abstract
Inflammatory diseases, such as asthma and inflammatory bowel disease, are mediated by excessive immune responses.
Currently used monoclonal antibody (mAb) therapy can reduce symptoms but it has disadvantages of short half-lives and
adverse reactions of developing antibodies to the infused mAb. To overcome them, therapeutic vaccines are being developed.
We have developed IgE- or cytokine-peptide based and virus-like particle vaccines that induce long-lasting antibodies to block
IgE or the target cytokine without use of an adjuvant. To break self-tolerance, truncated hepatitis B core antigen (HBcAg) is
used as a carrier protein. The vaccine is made by inserting a small peptide derived from receptor binding sites of the target
molecule into the immunodominant epitope of HBcAg via gene recombination methods. Recombinant vaccines are expressed,
purified and in vitro identified. in vivo evaluation of the vaccine is performed in mice with ovalbumin-induced acute and/or
chronic airway inflammation or in mice with acute and/or chronic experimental colitis. Immunization with an IgE vaccine
prevents subsequent increase of IgE and down-regulates elevated IgE in sensitized rodents. Vaccines against IL-13 or IL-4
effectively meliorate and/or reverse airway inflammation and remodeling in asthmatic mice, while vaccines against IL-12/
IL-23p40, IL-17 or TGFβ1 significantly meliorate and/or reverse intestinal inflammation and fibrosis in colitis mice. Safety
examination indicates that immunizations with the vaccine to IL-12/IL-23p40 do not increase the susceptibility to the lung
infection of chlamydia muridarum in mice.We conclude that IgE/cytokine vaccines may provide a potential therapeutic strategy
for the long-term treatment of allergic and autoimmune diseases.
Biography
Dr. Zhikang Peng is a professor in the Department of Pediatrics and Child Health and the Department of Immunology, University of
Manitoba. She received an MD in 1968 and an MSc in 1982 from Fudan University Medical School (formerly named Shanghai First
Medical University). From 1986 to 1991 she was trained as a post-doctoral fellow at Johns Hopkins University and the University of
Manitoba. In 1992, Dr. Peng became a faculty member. Her research interests include therapeutic vaccines for treatment of allergic
and auto immune diseases and mosquito allergy. Dr. Peng has published more than 80 papers in peer-reviewed journals.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
73
A Systems Biology Approach to Vaccine Design: We Have Found the Epitopes and They Are
Us
Jane Homan*, and Robert Bremel
EigenBio LLC, USA
Abstract
EigenBio has developed a suite of applied mathematics tools to analyze protein sequences and identify upregulatory epitopes
comprising B cell epitopes, and closely associated MHC binding enabling T-helper responses and cross presentation. Analysis
of primary amino acid sequences also identifies amino acid motifs most likely to down-regulate T-cell responses by engaging
a large cognate T cell population. Motifs which elicit Tregs may enhance pathogenesis and tolerance. Many so-called virulence
factors in bacteria contain a high content of potentially suppressive motifs. Understanding the distribution and balance of T-cell
exposed motifs which may up or down regulate the T-cell response is critical to vaccine design, whether for infectious disease or
cancer. The very extensive polyspecificty of T-cell exposed motifs shared between proteins of microbial and mammalian origin
contributes to off-target and autoimmune responses. We will discuss applications of systems biology tools in epitope analysis
and provide examples of the distribution patterns of T cell exposed amino acid motifs in several microbial and mammalian
settings.
Biography
Dr. Jane Homan is a Founder of EigenBio, LLC and ioGenetics, LLC. Formerly CEO and Co-Founder of Gala Biotech Inc. (now
part of Catalent Inc) where she helped to establish the recombinant protein expression systems. Homan received a PhD in
virology and veterinary science from University of Wisconsin-Madison, a Master’s degree in microbiology from the University
of Saskatchewan. She is a Member of the Royal College of Veterinary Surgeons, having graduated from Glasgow University
Veterinary School. With Dr. Bremel she has built the intellectual property portfolio underpinning EigenBio’s immunoinformatics
work, and has published on its applications.
Computational Prediction of a Continuous Peptide Epitope Reacting with Pandemic
Influenza A(H1N1)
Absalom Zamorano-Carrillo1* Jonathan P. Carrillo-Vazquez1, Jose Correa-Basurto2, Jazmin Garcia2, Rafael CamposRodríguez3, Violaine Moreau4, Jorge L. Rosas-Trigueros5, Cesar A. Reyes-Lopez1, and Marlon Rojas-Lopez6
Lab de Bioquimica y Biofisica Computacional, ENMH-IPN, France
Lab de Modelado Molecular y Diseio de Farmacos ESM-IPN, France
3
Lab de Inmunología Molecular ESM-IPN, France
4
CNRS FRE-Montpellier France
5
Lab Transdisciplinario de Investigacion en Sistemas Evolutivos, France
6
CIBA-IPN, France
1
2
Abstract
Identification of immunological epitopes by bioinformatics tools is part of the challenges in immunological studies. Several
methods show considerable success and concurrently with experimental studies, the capability of the algorithms to classify
potential peptides with biological activity has increased. In this work, an epitope was designed by conventional bioinformatics
tools, docking, and molecular dynamics simulations. As a template, the hemagglutinin protein of the H1N1 influenza pandemic
strain was used due to the interest of designing a scheme of immunization. Next, we used this engineering-epitope to analyze
if any antibodies were in human sera; thus, we assayed an ELISA in samples before and after the influenza outbreak in 2009. A
plaque reduction neutralization test induced by virus-neutralizing antibodies and the IgG measurement confirmed the biological
activity of this computationally conceived peptide. The results of the ELISAs confirmed that the serum of both pre-pandemic
and pandemic recognized the epitope. Further, the plaque reduction neutralization test evidenced the ability of the designed
peptide to neutralize influenza virus in Madin-Darby canine cells.
J Mol Recognit. 2015 Sep; 28(9):553-64. doi: 10.1002/jmr.2470. Epub 2015 Mar 18
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
74
Cross-Reactive Multifunctional T Cell Responses to Salmonella enterica Serovars in Humans
Rezwanul Wahid*, Stephanie Fresnay, Myron M. Levine, and Marcelo B. Sztein
Center for Vaccine Development, University of Maryland School of Medicine, USA
Abstract
Salmonella paratyph A infection has emerged as a major health concern in endemic areas due to several factors, including its
rising incidence, that there is no vaccine available against this organism and the available live oral typhoid vaccine Ty21a offers
minimal, or no cross-protection against S. Paratyphi A. Development of new vaccines against human-restricted Salmonella,
species, including S. Paratyphi A, hinges on a better understanding of the determinants of protective immunity. However, this
endeavor is hindered by the lack of suitable animal models. To improve our knowledge on human immunity against S. Typhi,
over the past 2 decades we conducted immunological studies using PBMC obtained from Ty21a or candidate typhoid vaccine
immunized volunteers. In recent years we extended these studies by evaluating the induction of key cross-reactive cell mediated
immune responses (CMI) against S. Typhi, S. Paratyphi A and S. Paratyphi B in Ty21a vaccines. Our results showed, for the
first time, that Ty21a elicited Salmonella specific cross-reactive multifunctional (MF) CD8+ and CD4+ cells that concomitantly
produced IFN-γ, TNF-α, IL-2, MIP-1β and/or express the cytotoxicity indicator molecule CD107a. A significant portion of these
cells also have the potential to home to the gut. However, MF CD8+ T cell responses were predominantly elicited against S. Typhi
but not against S. Paratyphi A. These data provide insights into the immunological basis underlying the field observations that
Ty21a does not cross-protect against S. Paratyphi A, suggesting that a bivalent S. Typhi/S. Paratyphi A vaccine might be required
to protect against enteric fevers.
Biography
Dr. Rezwanul Wahid, MBBS, PhD is an Assistant Professor at Center for Vaccine Development, University of Maryland, USA.
He has been involved with translational research focusing on the human host-immune responses elicited by natural infection or
immunization with various vaccines. The ultimate goals of his work are to enhance the development of novel vaccines against
various diseases, especially against enteric diseases, by understanding the immune mechanisms that can be correlated with
protection in humans. He has authored or co-authored more than 20 research articles in various peer reviewed journals related
to this subject.
Studies on Mumps Virus Infection, Genotype Identification of Circulating Strains among
MMR Vaccine Recipients
Jeevan Malaiyan1*, Gokul G Radhakrishnan2, and Abinaya Vijayan3
Sri Muthukumaran Medical College Hospital and Research Institute, Chennai, India
Meenakshi Medical College Hospital and Research Institute, Kanchipuram, India
3
Sree Balaji Medical College and Hospital, Chromepet, Chennai, India
1
2
Abstract
Mumps, long considered a vaccine preventable childhood infection has now caused worldwide re-emergence in vaccinated
populations. Thus, a study was done to investigate the cases of vaccine failure among mumps suspect cases in Chennai, India.
Results revealed an alarming 90.5 % of vaccinated cases were positive for anti-mumps IgM antibody, indicating that MMR
vaccine had failed to offer protection. This is the first report to portray the high prevalence of mumps in vaccinated populations
in India. Genotypic characterization of the virus revealed that the circulating strain was genotype C which is distinct from
the vaccine strain of genotype N (L-Zagreb). This is also the first report in India to suggest that genotype C was responsible
for the present mumps infection. Poor efficacy is a contributing factor to the failure of MMR vaccine and hence, its efficacy
was analyzed by determining the seroprotective antibody level. Highest seropositivity (100%) was noticed for rubella, an
intermediate number (76% & 92% who received two doses and first doses of MMR) for measles and the lowest (49% & 83%) for
mumps virus. This warrants a revisit of vaccine preparation using circulating strains and optimization to improve its efficacy.
Biographies
Dr. Jeevan Malaiyan, Ph.D. presently working as an assistant Professor in the Department of Microbiology, Sri Muthukumaran
Medical College Hospital and Research Institute, Chikkarayapuram, India.
Dr. Gokul G Radhakrishnan, M.D, working as Senior Resident in Department of Medicine, Meenakshi Medical College Hospital
and Research Institute, Kanchipuram, India.
Dr. Abinaya Vijayan, M.D, working as Junior Resident in Department of Obstetrics & Gynaecology, Sree Balaji Medical College
and Hospital, Chromepet, India.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
75
Vaccination in TB Burden Countries: Is there a Role of BCG
D. Behera
Postgraduate Institute of Medical Education and Research, India
Abstract
BCG was first used to immunize humans in 1921. It was introduced into the WHO Expanded Program on Immunization in
1974. The global BCG vaccination coverage rates exceed 80% in many countries endemic for TB. Extensive clinical trials have
been conducted to assess the protective efficacy of BCG against pulmonary TB, but a wide range of vaccine efficacy values
have been observed. This is possibly due to differences in study design, geographical location and presence and absence of
NTM. The greatest disadvantage of BCG vaccination is that it does not prevent reactivation of latent TB, which is the main
source of bacillary dissemination in the community. Despite these limitations, and particularly in light of the growing HIV/
AIDS pandemic and the appearance of multidrug-resistant M. tuberculosis strains, BCG vaccines will continue to represent an
important tool in the global fight against TB until new vaccines are available for clinical use. In India, BCG vaccination is covered
under the universal EPI where in it is administered at birth till the age of I month. The only benefit is that it prevents the
development of dissemination and tubercular meningitis. However a large number of pediatric tuberculosis continues to occur.
In fact it constitutes about 6-10% of the total tuberculosis burden of about 2 million cases per annum in India.
Then is it true that BCG vaccination is efficacious? Details will be discussed.
Development and Characterization of Protective Antibodies Against Marburg virus Derived
from a Macaque Immune library and Perspectives for Prophylactic Use
Froude JW1,4*, Pelat T2, Miethe S3, Zak S1, Bakken R1, Hust M.3, P. Thullier2, and Dye J.M1
US Army Medical Research Institute for Infectious Disease (USAMRIID), Fort Detrick , Maryland, USA
Unite de Biotechnologie des Anticorps, Institute de Recherche Biomedicale des Armees [IRBA-CRSSA], La Tronche, France
3
Institut fur Biochemie und Biotechnologie, Technische Universitat Braunschweig, Braunschweig, Germany
4
Currently working at: Walter Reed Army Institute of Research (WRAIR), Silver Spring MD, USA
1
2
Abstract
A growing number of monoclonal antibodies (mAbs) are known to effectively neutralize and have demonstrated protection against
Ebola viruses (EBOV) in non-human primates (NHP) and been utilized under compassionate use provisions in several human
cases. To date, all filovirus mAbs have targeted EBOV, with no product for Sudan virus (SUDV) and just two demonstrating murine
protection to the Ravn strain of Marburg virus (MARV). Utilizing non-human primates (NHP), cynomolgus macaques, immunized
with viral-like particles expressing the glycoproteins (GP) of MARV Ci67 strain at their surface, we generated a library of single chain
variable fragment (scFv) antibodies capable of partial neutralization Marburg in vitro and providing in vivo protection in mouse models
against wild-type virus. The NHP derived immune library directed against MARV was built after four immunogen injections to a size
of 1.2x1014 clones, and was screened against the GP of Marburg virus Ci67 isolate. A screening of 192 sequenced clones identified
18 clones with distinct VH and VL sequences. Of these, 14 scFvs have been identified with a capacity to partially neutralize in plaque
reduction neutralization testing (PRNT). Four of these antibodies (Fr1, Fr2, Fr3, and Fr13) were produced in the scFv-Fc fusion
format with a murine Fc domain for in vivo protection studies. The antibodies have protective efficacy in mice ranging from 70 to
100 percent. The amino-acid sequences of these macaque-derived antibody fragments are similar to those of their human germline
counterparts, sharing an identity ranging between 68 and 100% to human germ-line immunoglobulin variable sequences. These
results further demonstrate the ability to isolate human-like antibodies from NHP immune libraries in the perspective of their
potential for therapeutic use. Additionally, modifications to the Fc structure of the IgG has been shown to greatly enhance the half-life
of these molecules and the potential for prophylactic administration of these antibodies will be discussed.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
76
Poster Session
Recombination Analysis Based on HAstV-2 and HAstV-8 Complete Genome in South Korea
Jae-Woong Lee1*, Hyun-Ju Ha1, Sung-Geun Lee2, Han-Gil-Cho3, Ji-Young Jin1, and Soon-Young Paik1
Department of Microbiology, College of Medicine, The Catholic University of Korea, Republic Korea
Korea Zoonosis Research Institute, Chonbuk National University, Republic of Korea
3
Division of Public Health Research, Gyeonggi Province Institute of Health and Environment, Republic of Korea
1
2
Abstract
Human Astro Virus (HAstV) occurs worldwide and is mainly found as a cause of infant diarrhea, the diarrhea in patients with
seniors and military, and immune dysfunction. The purpose of this study was to identify HAstV strains research and characterize
rare genotypes. Stool samples were collected from patients with symptoms of acute gastroenteritis in Korea. Samples were
analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR). The full-length genome of HAstV was amplified using
10 pairs of newly designed primers. The new primers were designed based on the published sequences of HAstV type8 variants
of HAstV. The HAstV consists of 6776bp and contains three sequential ORF, designated ORF1a (2781bp), ORF1b (1549bp), and
ORF2 (2349bp). This study is the first report of the complete nucleotide sequence of the recombination HAstV-2 and HAstV-8.
Pursuant to SimPlot, the Kor85 strain was a recombinant strain with breakpoint at the ORF1a, ORF1b and ORF2 junction
between HAstV-2 and HAstV-8 strains. The result indicated that there was a recombination event between ORF1 and ORF2
region of the rare strain and the recombination breakpoints belonged to polymerase region. This may represent the standard
sequence of the HAstV variant in South Korea and could therefore be useful for the development of HAstV vaccines.
Full-Length Genomic Sequence of Sapovirus Isolate in South Korea
Yu-jeong Won1*, Hye Lim Choi1, Chang-Il Suh2, Seung-Won Park3, Ji-Young Jin1, Han-Gil Cho4, and Soon-Young Paik1
Department of Biomedical Sciences, College of Medicine, The Catholic University of Korea, Republic of Korea
Department of Medical Consilience, 152, Dankook University, Republic of Korea
3
Department of Biotechnology, Catholic University of Daegu, Republic of Korea
4
Division of Public Health Research, Gyeonggi Province Institute of Health and Environment, Republic of Korea
1
2
Abstract
Sapovirus (SaV) belongs to the group of the viruses that cause gastroenteritis in human beings, to which norovirus, rotavirus,
and astrovirus also belong. SaV is divided into 5 genogroups (GI-GV). GI, GII, GIV, and GV mostly infects human beings, while
GIII cause the infection to pigs in most cases. GII and GIII have 2 open reading frames (ORF) and other 3 genogroups have three
ORF. This research was the first to carry out the complete genome analysis of SaV in Korea, which we called ROK62. ROK62
used in the research has the length of 7431 nucleotides and was found to belong to GI-1. Among other SaVs, Manchester,
Nc114, and N2 have 94% of similarity with ROK62. Recently, the infection of SaV is rapidly increasing in the countries nearby
South Korea. This research provides a good model to monitor the whole genome sequence of SaV. The circulation of a particular
genotype and rapid change in SaV justifies national surveillance and international cooperation to prevent outbreaks. The
monitoring on predominant strains should be based on the detection of particular parts of the SaV genome. The whole SaV
genome sequencing and phylogenetic analysis results will help both to successfully carry out epidemiological surveillance and
to bring the improvement of detection systems.
Biography
Yu-Jeong Won has received bachelor of biology degree from Pai Chai University in Korea (2012). Presently, she is pursuing a
doctorate in immunity of microbiology at the Catholic University Medical School, Seoul, Republic of Korea.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
77
Branched Amphiphilic Peptide Capsules for Delivery of a Therapeutic DNA Vaccine against
HPV-Induced Tumors
Luana R. M. M. Aps2*, L. Adriana Avila1, Pinakin Sukthankar1, Nicoleta Ploscariu3, Patricia Games4, Robert Szoszkiewicz3,
Rubens P. S. Alves2, Mariana O. Diniz2, Luís C. S. Ferreira2, and John M. Tomich1
1
Department of Biochemistry & Molecular Biophysics, Kansas State University, USA
2
Institute of Biomedical Sciences, University of São Paulo, Brazil
3
Department of Physics, Kansas State University, USA
4
Department of General Biology, Federal University of Vicosa, Brazil
Abstract
Branched Amphiphilic Peptide Capsules (BAPCs) display features compatible with clinical applications. BAPC supramolecular
assembly requires two branched peptides: bis(Ac-FLIVIGSII)-K-K4 and bis(Ac-FLIVI)-K-K4. In aqueous solution, they are able
to assemble in hollow capsules that present an uniform size of ~20-30 nm and are capable to encapsulate solutes during this
process. Here, we present a more stable BAPC form, referred to as “locked”, generated when BAPCs are formed at 25°C, cooled to
4°C and then returned to 25°C. Under these conditions, the negatively charged DNA interacted with the cationic surface of the
capsules through numerous electrostatic interactions generating peptide-DNA complexes with a size ranging from 50-100nm.
Biophysical characterization of these complexes showed that the amphiphilic peptide core act as cationic nucleation center
around which the dsDNA wraps. Transfection of HeLa cells with BAPC nanoparticles loaded with a GFP-encoding plasmid
yield better results than those achieved with commercial reagents with minimal cytotoxic effects. In addition, coupling of an
anticancer DNA vaccine encoding an oncoprotein of the type 16 human papillomavirus (HPV-16) to BAPC nanoparticles led to
enhanced antitumor therapeutic effects. DNA-loaded BAPCs delayed tumor growth in mice transplanted with HPV-16 tumor
cells (TC-1 model) without detectable acute toxicity but with a different peptide:DNA ratio required for optimal HeLa cell
transfection. Furthermore, the combination of plasmid DNA and BAPCs resulted in activation of dendritic cells, demonstrating
that these complexes have also an inherent immunomodulatory role and, thus, represent a safe and efficient non-viral gene
delivery approach for in vitro and in vivo applications.
Biography
Luana R. M. M. Aps completed bachelor’s at Industrial Pharmacy from Universidade Federal Fluminense (2009) and master’s
in Biological Sciences working on adjuvants and microcarriers for DNA vaccines, resulting in the latest work in Vaccine (Aps
et al.,2015) and a patent application (INPI BR 10 2014 009679 5). Luana has experience in vaccines R&D as a researcher on
veterinary and human health industries and throughout her career she received four awards including one in Oncology category
as co-author in the work published in Human Gene Therapy journal (Diniz et al., 2013). Currently her PhD is in progress working
on chemotherapy in association with therapeutic DNA vaccines against HPV-induced tumors and new delivery systems.
Screening of HSV 1 Vaccine Candidate Strains via CRISPR-Cas9 System
Xingli Xu*, Jienan Zhou, and Qihan Li
The Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, China
Abstract
Herpes Simplex Virus type 1 (HSV-1) infection is widely recognized to be one of the major threats to human health, which
can cause herpes labialis, genital herpes, encephalitis, and establish lifelong latency in the nervous system. And there are no
effective vaccines or drugs for the prevention and treatment of latent infection of HSV-1 yet. In our study, the multiple growing
non-essential genes of HSV-1 were simply mutated via CRISPR-cas9 system for construction of mutant viruses. Based upon the
brief description of the immunological, molecular biological and pathologic features of the mutant strains, we had identified an
attenuated virus strain called UL7-MU, consisting of 30bp deletion in UL7 gene. The results showed that UL7-MU virus was able
to replicate in Vero cells, with plaque-forming and proliferation strikingly eliminated. There were no substantial pathological
changes in mice infected with the UL7-MU virus, leading to no death of infected mice.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
78
Early Impact in Reducing Cervical Abnormalities in Campos dos Goytacazes-RJ, State of
Rio de Janeiro, Brazil, After Introdution of the Quadrivalent HPV Vaccine for Girls 11-15
Years Old in 2010
Charbell Miguel Haddad Kury
Universidade Federal Fluminense, Brazil
Abstract
Introduction: Human papillomavirus (HPV) is a real challenge in terms of public health. Tracing through Pap test associated
with the use of condoms and HPV vaccines are strategies for preventing this condition. The municipality of Campos dos
Goytacazes, located in the state of Rio de Janeiro, introduced in September 2010 the quadrivalent HPV vaccine to girls 11-15
years old in a hybrid strategy of vaccination (schools and health centers).
Methods: Historical cohort which evaluated the impact of HPV vaccination as a protective factor against high and low risk of
HPV abnormalities. Results of the pap smear test obtained from the Brazilian Ministry of Health database (SISCOLO system)
were categorized in Low grade abnormalities (LGA) and High grade abnormalities (HGA). This preliminary study focused in LGA
rates, which were estimated for 1-month period and stratified by four age groups (< 20; 20-24; 25-30; >30 years) from 2007 to
2014. A quantitative comparison of LGA temporal trends before and after vaccination was done with Quasi Poisson regression
analysis. The protective effect of the vaccine on the timeline was assessed by calculating relative risk (RR) in each age group.
Results: There was a significant decrease of more than 60% in LGA in women < 20 years old, and almost 50% for the other
groups. HPV vaccine was a protection factor, because of the RR result was <0.0001 in all age groups.
Conclusion: although the studies show that the pre-neoplastic lesion caused by HPV may be reversible spontaneously, it’s
undeniable that the vaccine contributed greatly to the high reduction rates, associated with high vaccination coverage. These
results are the first ones in Brazil.
Characterization of Sam® Vaccine Mechanism of Action by Two-Photon Microscopy
Lucia Lapazio1,3*, D. Piccioli1, C. Giovani1, S. Buccato1, C. Pergola1, M. Soriani1, A. Pezzicoli1 , T. Carsillo2, and A. Geall2
GSK Vaccines, Italy
GSK Vaccines, USA
3
University of Siena, Siena, Italy
1
2
Abstract
The SAM® vaccine is constituted by a synthetic self-amplifying mRNA derived from the alphavirus genome and delivered by
a vesicle-based delivery system. Recent data indicate that SAM® vaccination is effective at eliciting both humoral and cellular
antigen-specific immune responses. However, the understanding of the mechanism of action of SAM® vaccine-induced
immunity is still incomplete. Indeed, even if there exist evidences that muscle cells support the production of the antigen
whereas dendritic cells (DCs) seem to have a major role in antigen presentation to CD8 T cells, the dynamic of the events leading
to a productive antigen-specific immunity has not been elucidated yet. To this aim we have applied an intra-vital imaging
approach to track in real time the formulated replicons, the expressed antigens (GFP and mCherry) and the immune cells in live
mice. This real-time setting revealed that up to two days post SAM® injection, antigen expression was detected only in cells with
a dendritic morphology infiltrated within the muscle bundles, while no expression could be detected inside the muscle fibers.
Only after 3 days post-injection the antigen started to be heavily produced by muscle fibers and its presence could be observed
up to several weeks. We are currently setting up experiments to visualize the dynamic of antigen-expressing muscle cells and
dendritic cells interaction. In conclusion, we believe that intra-vital imaging is a powerful tool to dissect vaccine mechanism of
action and to predict the efficacy of intervention based on an accurate understanding of the immune response.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
79
The Dynamics of Antigen-Specific and Regulatory of Lymphocyte Populations in
People Immunized by Live Plague Vaccine
Deryabin P. N1, Ponomaryova T. S1*, Karalnik B. V2, Kustova E. A1, Denisova T. G2, Urazalieva N. T1, Tugambayev T. I1,
Zakaryan S. B1, and Alymkulova Z.T1
Aikimbaev Kazakh Scientific Center for Quarantine and Zoonotic Infections, Kazakhstan
Scientific Center of Hygiene and Epidemiology, Kazakhstan
1
2
Abstract
A total of 13 healthy adults immunized by live plague vaccine EV. 6 people were vaccinated is primary, 7 people - again (not less
than 1 year after previous vaccination). Vaccination performed by the method of scarification, one dose of vaccine contains 3 ×
108 living microbial cells. Blood for research were collected from ulnar vein before vaccination and at 2, 6, 9, 13, 20, 27, 34, 48
and 62 after vaccination.
Determination of the main populations of lymphocytes (CD3, CD19, CD4, CD8, CD16 + 56, HLA-DR), markers of regulatory
T-lymphocyte CD4 / CD25 / FoxP3) conducted using laser flow cytometry with monoclonal antibodies using a "double mark":
two types of monoclonal antibodies labeled with different colorants (FITC - flyuoristsein-5-izotiotsonat and PE - phycoerythrin).
Analysis of samples was carried on a flow cytofluorimeter FacsCalibur (Becton Dickenson, USA) in the program CellQuest. The
results obtained were compared with data from in the control group (50 conditionally healthy people aged 20 to 50 years) and
reference limit of normal by the manufacturer monoclonal antibodies. The content of lymphocyte with receptors (LfR) to Y.
pestis antigens F1 and LPS was determined in response to lymphocyte adhesion of immunoreagents and which we obtained
conjugating F1 and LPS c erythrocytes bovine, fixed acetaldehyde. Simultaneously carried out of such tests with the control
reagent erythrocytes not loaded antigens Y. pestis. It was shown that content of the main of lymphocyte populations (CD3,
CD19, CD4, CD8, CD16 + 56, HLA-DR) in all examined people vaccinated live plague vaccine does not go beyond the normal
values and remained practically unchanged over time. At the same time it should noted that the content of T - regulatory
lymphocytes in the group immunized is primary significantly greater than in the group of people vaccinated repeatedly. The
lymphocytes with receptors to antigens of Y. pestis not found before vaccination, and detected at the 2nd, 6th, 9th and 13th day after
vaccination. After the 20th day of lymphocytes with receptors for Y. pestis antigens were not determined. It should be noted that
the content of lymphocyte with receptors to the Y. pestis antigens on the 2nd and 13th day after the is primary vaccination in the
vaccinated group were significantly less than in the group vaccinated repeatedly. On the 6th and 9th day after vaccination content
of lymphocytes with receptors for Y. pestis antigen in both groups of vaccinated of people were not significantly different.
Thus, it can be noted that the multiplicity immunization live plague vaccine effect on content of T-regulatory lymphocytes, the
content and dynamics of lymphocyte with receptors to antigens of Y. pestis.
Strategically Modulating the Skin Microenvironment to Potentiate Vaccine Efficacy
Jack R. You*, Guo Chen, Cara D. Carey, Jiying Zhang, Rhonda M. Brand, Zhaoyang You, and Louis D. Falo, Jr.
Department of Dermatology, University of Pittsburgh School of Medicine, USA
Abstract
Skin, the most accessible organ, harbors a variety of immunologically responsive cells that contribute to both innate and
adaptive immunity, rendering it an ideal anatomical target for vaccination. Keratinocytes initiate cutaneous inflammation by
producing pro-inflammatory mediators while immunocytes respond to these mediators, leading to fortified cutaneous immune
responses. Targeting keratinocytes to simultaneously produce antigens and pro-inflammatory mediators for a greater immune
response remains a challenge, but is salient for vaccination. Transcription factor x-box binding protein 1 (XBP1) is known to
promote production and secretion of proteins as well as strengthen pro-inflammatory responses; it is even a key molecule in
the body’s response to human influenza virus. We found that transient overexpression of XBP1 in a single cutaneous genetic
immunization not only enhanced expression of pro-inflammatory mediators and co-delivered antigen, but also increased skininfiltrating immunocytes, skin-resident memory CD8+ T cells, durable antigen-specific CD8+ T cells, and protective antitumor
immunity. Keratinocytes genetically engineered with XBP1 and secreted antigen in vitro increased productions of not only
said exogenous antigen, but also pro-inflammatory mediators. Transient overexpression of XBP1 in keratinocytes in situ
was sufficient for increasing expression of pro-inflammatory mediators, skin-resident memory CD8+ T cells, vaccine-induced
durable antigen-specific CD8+ T cell and protective antitumor responses. In human skin explants, XBP1 triggered expression
of an extensive array of genes associated with a pro-immunogenic skin microenvironment and increased expression of the codelivered antigen after cutaneous genetic immunization. These findings provide novel insights into the rational design of potent
vaccines via strategic modulation of the skin microenvironment.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
80
An Extra-Ordinary Formulation for Adenoviral Vaccines
Lara Babich*, and Pol Adriaansen
Janssen Infectious Diseases & Vaccines (Crucell B.V.), The Netherlands
Abstract
The road to developing a stable liquid formulation for a live virus is burdened with challenges requiring a tailored methodology.
The approach described here yielded a stable Adenovirus-based vaccine. In short, formulation components were initially screened
and selected according to their stabilizing potential in accelerated stability and physical stress studies, first in high-throughput
systems later fine-tuned in conventional models. In order to allow operational flexibility, Quality by Design (QbD) principles
were subsequently used to explore the optimal design space of each individual factor of the formulation (excipients, pH, API).
A Design of Experiments (DOE) screening defined the specifications for each component, using potency and quantity assays to
evaluate the long term stability and resistance to physical stress. Data were analyzed by predictive statistical models, optimal
specifications were identified, and as a result this robust formulation was implemented in CTM manufacturing. Importantly,
clinical trials in developing countries may impose unusual cold-chain facilities and vaccination campaigns (e.g. mid-term
storage above the intended temperature and vaccination at uncontrolled ambient conditions). The formulation chosen for our
vaccines guarantees stability under the extra-ordinary situations described above, allowing great flexibility outside the normal
temperature range of a standard clinical trial. This allows the storage at different (increasing) temperatures to adapt to the coldchains of different countries, multiple freeze-thaw cycles, and temperature excursions to extreme temperatures. The vaccine has
been proven stable also in in-use stability studies at extreme temperature.
Biography
Dr. Lara Babich currently works as a scientist Formulation Development at Janssen and is responsible for the development of
stable vaccines against infectious diseases such as Ebola and HIV. Lara holds a MSc degree in Industrial Biotechnology from
Padua University (Italy) and a PhD in (Bio) Chemistry from the University of Amsterdam (the Netherlands).
Immunogenicity Comparison of Two Chimeric Recombinant Proteins Based on
Circunsporozoite Protein for Development of a Vaccine against Plasmodium vivax
Tarsila M Camargo1*, Luciana C Lima2, Mariana V Rocha1, Karina A Caramico1, Kátia S Francoso1, Maurício M Rodrigues2,
and Irene S Soares1
University of São Paulo, São Paulo, SP/Brazil
Federal University of São Paulo, SP/Brazil
1
2
Abstract
Recent studies have contributed to important advances toward the development of a vaccine against human malaria caused
by Plasmodium falciparum based on a liver stage protein, circumsporozoite protein (CSP). Clinical trials performed in African
endemic areas demonstrated that the formulation has a significant, albeit partial, effect in retarding clinical malaria acquisition
in vaccinated children (1). These optimistic results boosted efforts for using the CSP to develop a vaccine formulation against
P. vivax, the most widely distributed malaria worldwide. Toward that goal, we generated 2 chimeric recombinant proteins based
on CSP: CSAllFL and CSAllC-term. Both proteins are based on the primary sequences representing the different allelic forms
of P. vivax CSP (VK210, VK247 and vivax-like). CSAllFL contains both N- and C-terminal regions. The latter differ only in the
lack of an N-terminal region. The proteins were expressed in Pichia pastoris yeast cultures. Supernatants were purified using
Ni-affinity and ion exchange chromatographies and purities were analyzed using RP-HPLC. We vaccinated C57BL/6 mice with
the recombinant proteins in the presence of Poly (I: C) or Montanide ISA-720 adjuvants. After two boosts, disregarding the
adjuvant, mice vaccinated with either protein presented high IgG titers (>105; by ELISA) specific to all PvCSP alleles, showing
that both proteins were significantly immunogenic and promising the generation of high IgG titers. We concluded that the two
chimeric constructs are interesting candidates for developing a global vaccine against malaria caused by P. vivax.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
81
A Review of Preclinical Studies Characterizing the Immunogenicity, Efficacy and Safety of
the Replication-Defective Herpes Virus Type 2 Vaccine HSV529
John Hamberger1, Hector Hernandez1, Ana P. Goncalvez1, Harry Kleanthous1, Catherine Caillet2, Fabrine Pradezynski2,
Sanjay Phogat3, Marie-Clotilde Bernard2, and Simon Delagrave1
Sanofi Pasteur, Research NA, Cambridge, MA, USA; 2Sanofi Pasteur, Research and Development, Marcy-l’Étoile, France
Sanofi Pasteur, Research and Development, Swiftwater, PA, USA
1
3
Abstract
Herpes simplex virus type 2 (HSV-2) is the principal cause of genital herpes and is extremely prevalent, with estimates of more than 530
million people infected throughout the world. The replication-defective virus dl5-29, now called HSV529, was constructed by deleting the
UL5 and UL29 genes of HSV-2 and propagated on a complementing cell line expressing the missing genes. A series of preclinical studies
were performed across multiple sites to assess the immunogenicity and efficacy of HSV529 following immunization in mice and guinea
pigs (Delagrave, S. et al., 2012, Plos One, doi: 10.1371/journal.pone.0046714; Bernard, M.C. et al., 2015, Plos One, DOI: 10.1371/journal.
pone.0121518). Monitored parameters included neutralizing antibody production, characterization of cellular immune responses, viral
shedding and ganglionic viral latency morbidity and mortality. HSV529 replication and pathogenicity were also investigated in sensitive
models of virus replication. Results indicate that HSV529 immunization effectively prevented HSV-2 infection in tested animal models as
evidenced by reducing mortality, disease severity as well as frequency and magnitude of viral shedding and latent infection. Furthermore,
HSV529 vaccination reduces rate of recurrent HSV-2 disease in the pre-infected guinea pig model, thus, suggesting a role of HSV529 in
the treatment of genital herpes disease (unpublished results). The studies also showed that HSV529 does not replicate or propagate in
vitro and in vivo. These results were used to support the ongoing clinical investigation of HSV529 in humans.
Biography
Dr. John Hamberger is currently Manager of Virology at Sanofi Pasteur, Research NA in Cambridge, MA, USA. His career in viral vaccines began
in 2000 at OraVax , and later, Acambis, before their acquisition by Sanofi Pasteur in 2008. Prior to 2000, he was a Senior Scientist for thirteen
years at Bausch & Lomb Research & Development Personal Products Division, Rochester, NY USA in Microbiology and In vitro Toxicology.
The Studies on Latent TB Infection
Yan Liang, Xuejuan Bai, Yourong Yang, Junxian Zhang, Lan Wang, Jie Wang, and Xueqiong Wu*
Army Tuberculosis Prevention and Control Key Laboratory, New Tuberculosis Diagnostic and Treatmental Technique Beijing Key
Laboratory, Institute of Tuberculosis Research, the 309th Hospital of Chinese PLA, China
Abstract
Latent tuberculosis infection (LTBI) is one of the hot and difficult points on tuberculosis research. At present, the definition,
pathogenesis, clinical diagnosis, clinical outcome and other issues for LTBI have not been fully elucidated, there is some controversy.
The studies on LTBI were performed in the following aspects: (1) The epidemiology of LTBI in 907 cases of recruits was studied
using PPD skin test and ELISPOT assay. The rate of LTBI was 30.7%. The rate of LTBI in 897 recruits from 9 provinces was related
to the severity of local tuberculosis epidemic. The results suggest that the ELISPOT assay could reflect the status of LTBI. There
was no significant difference among the LTBI rates of the recruits from the city, urban and rural area, which is probably because the
72.6% recruits were high school graduates. (2) The mechanism of LTBI was studied. We found that resuscitation promoting factor
could promote the resurrection and growth of dormant M. tuberculosis, and improve the positive rate of M. tuberculosis culture.
The biological and immunological characteristics of some hypoxia-related proteins and nutrient deficiency-related proteins were
studied, and found that there were some proteins with better immunogenicity. Some Immune molecular markers were screened
and verified. Under the stimulation of these antigens, the number of effector T cells secreting IFN-γ in LTBI group was significantly
higher than those in active TB group and uninfected normal control group, which will provide new targets for the diagnosis of LTBI
and the development of LTBI vaccine. (3) The new method of LTBI detection was studied. The consistent rate between PPD skin test
and ELISPOT assay had only 51.4%, and between two kinds of IGRA (ELISPOT and ELISA) was not also entirely consistent. ESAT6
protein was the dominant antigen in Chinese population, but the reaction of effector T cells to CFP10 protein was stronger than
ESAT6 protein. The positive rate of ELISPOT in the population with BCG vaccination was significantly reduced in comparation with
the population without BCG vaccination, suggesting that BCG vaccination can partially protect the people against the tuberculosis
infection, and reduce tuberculosis high-risk population. BCG vaccination is benefit to the PPD-negative and ELISPOT-negative
children and adults. Therefore, we implemented the strategy of BCG vaccination in the PPD-negative recruits of Beijing army. The
development of new antigen for skin test will improve significantly the specificity of LTBI detection. (4) 132 PPD-strong positive
or 278 ELISPOT-positive recruits were followed up for 18 months. All did not induce the TB, suggesting that a few PPD-positive or
IGRA-positive persons will develop active tuberculosis in the future. (5) The immunogenicities of 3 hypoxia-related DNA vaccines
(Rv2029c, Rv2628 and Rv1813c) and 1 nutrient deficiency-related DNA vaccine (Rv2659c) were studied, found that these 4 DNA
vaccines could induce low or medium level of IgG antibody, specific IgG1 and IgG2a antibody subtypes in mice, and IgG2a level
was higher than IgG1. IFN-γ and IL-4 levels in the supernatant of murine spleen lymphocyte culture were not increased. The
percentage of Th1 cells secreting IFN-γ was less than that of Th2 cells secreting IL-4 in the blood PBMCs by the flow cytometry,
but CD4+CD25+FoxP3+ regulatory T cells in whole blood were significantly increased, suggesting the DNA vaccines encoding LTBI
proteins induce Th2-type of cellular immune responses. We also try to clarify the existing problems and the prospect of LTBI.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
82
CRISPR/Cas9-Based Humanized Plants for the Production of Influenza Vaccines
Soonil Kwon1, Je Wook Woo1, Jungeun Kim2,3, Jin-Soo Kim2,3, Bum-Soo Hahn4, Hyun-Hwan Kim5, and Sunghwa Choe1,6*
Convergence Research Center for Functional Plant Products, Advanced Institutes of Convergence Technology, Suwon,
Gyeonggi-do, South Korea
2
Center for Genome Engineering, Institute for Basic Science, Gwanak-gu, Seoul, South Korea
3
Department of Chemistry, Seoul National University, Gwanak-gu, Seoul, South Korea
4
Department of Agricultural Biotechnology, Seoul National University, Gwanak-gu, Seoul, South Korea
5
Department of Agricultural Engineering, National Institute of Agricultural Sciences, Rural Development Administration,
Wanju-gun, South Korea
6
School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
1
Abstract
Production of vaccines using plant systems promises multiple benefits including cost, speed, scalability, and safety. To better
exploit the plant system as a vaccine factory, we sought to humanize plants in their post translational modification steps.
Firstly, we genetically engineer diverse plant species in their glycosyltransferase systems. The transferase gens of plants exist
as multiple copies and expressed under the different spatiotemporal regulatory programs, thus it is challenging to control
this system. In addition, conventional modification of these genes are required to go through regulations imposed on living
modified organisms (LMO) before being utilized as commercial purposes. To alleviate these concerns, we developed methods
entitled DNA-free genome editing in plants. We first in vitro assembled the two subunits of Cas9 including small guide RNA
and Cas9 protein, and this complex was introduced into cell wall-less plant cells. The RNA-protein complex successfully edited
the target genes without leaving footprints off-target. CRISPR/Cas9-mediated small deletion or insertion at the target gene
was virtually indistinguishable from naturally spontaneous mutations. Taking advantages of this technology, we are editing
plant specific glycosyl transferases in Arabidopsis, tobacco, lettuce, and Camelina. Under the various plants with different
genetic backgrounds, we express the HA gene of human influenza virus to produce a library of structurally varying proteins.
Combined with the technology of plant production in controlled environmental systems, the proteins to be produced should
meet impending needs of proteins having diverse structural variations.
Biography
Dr. Sunghwa Choe has earned his PhD from University of Arizona and worked for Ceres, Inc., a California-based plant biotech
company as a scientist. He is a professor at Seoul National University, and a director of the convergence research center for
functional plant products at Advanced Institutes of Convergence Technology in South Korea. He has published 50 papers in
reputed journals and has been serving as an editorial board member of journals including Molecules and Cells. He is developing
platform plants for production of designer proteins, engineering plant glycosyltransferases through DNA-free genome editing
technology of CRISPR/Cas9.
Engineering Novel Antibodies for Treatment of Emerging Infectious Diseases
Tianlei Ying*
Fudan University, China
Abstract
Therapeutic modalities based on monoclonal antibodies (mAbs) have shown clinical success in the treatment of many diseases.
By using an extraordinarily large human antibody library and in vitro display technologies we have identified potent neutralizing
mAbs against emerging infectious diseases and cancer. Some of these mAbs have been licensed to top biopharmaceutical
companies and are expected to move into the clinic shortly. We have also been working on the development of small-sized and
long-acting novel antibody constructs. The purpose of this study is to overcome the fundamental limitations for the use of fulllength mAbs as cancer therapeutics-- their high production cost, poor penetration into solid tumors, and poor or absent binding
to some sterically restricted epitopes.
Biography
Dr. Tianlei Ying graduated from Fudan University with a Ph.D. in 2010. He conducted postdoctoral research with Dr. Dimiter
S. Dimitrov at the National Cancer Institute, NIH from 2010 to 2014. In 2014, he joined the Key Lab of Medical Molecular
Virology of MOH/MOE, Shanghai Medical College, Fudan University, as Chief of the Antibody Engineering and Drug Discovery
Group. Dr. Ying's research interests include antibody-based therapies for cancer, AIDS and other infectious diseases.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
83
Affordable CRM197 Carrier Protein
Andrew Lees1, Sanjay Jain2, and Natalia Oganesyan2
Fina Biosolutions LLC, Rockville, MD, USA
Novo Biosolutions Inc., Shrewsbury, MA, USA
1
2
Abstract
CRM197, a genetically detoxified diphtheria toxin, is widely used as a carrier protein in conjugate vaccines. Highly effective
conjugate vaccines against Streptococcus pneumonaie, Haemophilus influenzae b and Neisseria meningiditis have been made using
this carrier protein. CRM197 has been expressed as a secreted protein in Corynebacterium diphtheriae but effective CRM197
production requires precise control of growth parameters and yields are typically low (<100 mg/L). Recently, CRM197 expressed
in the periplasm of Pseudomonas fluorescens has become available, but it is not generally priced to be an affordable alternative.
Fina BioSolutions has developed a highly efficient Escherichia coli expression system for CRM197, along with a simple purification
scheme. Previous methods for CRM197 expression in E.coli have involved refolding from insoluble inclusion bodies or secretion
into the periplasm. We have developed a method to produce soluble CRM197 in the cytoplasm of E.coli and achieved expression
yield of grams per liter in a fermenter. The production process comprises of a simple highly efficient purification method with
minimal number of steps. EcoCRMTM, economical CRM from E. coli, offers the promise of significantly reducing the cost of
conjugate vaccines.
Biography
Dr. Andrew Lees, Ph.D., is the founder and scientific director of Fina Biosolutions LLC, in Rockville, MD. He is an expert in the
chemistry of conjugate vaccines with over 20 patents to his name and an author on over 60 peer-reviewed articles. Conjugation
chemistry developed by Dr. Lees is used by GSK, the Serum Institute of India, the Chengdu Institute of Biological Products and
others in their S. pneumonia and meningococcal conjugate vaccines.
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
84
Notes
Vaccines Research & Development (Vaccines R&D-2015) | Nov 02-04, 2015 | Baltimore, USA
85
We wish to meet you again
at
Vaccines R&D-2016
Group
UNITED Scientific
# 2088 B2 Walsh Avenue, Santa Clara, CA 95050, USA
Ph: +1-408-426-4832, +1-408-426-4833; Toll Free: +1-844-395-4102; Fax: +1-408-426-4869
Email: vaccines@uniscigroup.org; contact@uniscigroup.net
www.unitedscientificgroup.com
Download