Vaccines For Humans
Vaccines For Humans
Vaccines For Humans
FOR HUMANS
PROJECT SYNOPSES
Research funded by
the European Union
EUROPEAN COMMISSION
Directorate Health
Unit F3 - Infectious Diseases
http://ec.europa.eu/research/health/infectious-diseases/index_en.html
Contact: Ole Olesen
European Commission
Office CDMA 02/158
B-1049 Brussels
Tel. (32-2) 29-53999
Fax (32-2) 29-94561
E-mail: [email protected]
EUROPEAN COMMISSION
VACCINES
FOR HUMANS
Research funded by
the European Union
2008
Directorate Health
00 800 6 7 8 9 10 11
(*) Certain mobile telephone operators do not allow access to 00 800 numbers
or these calls may be billed
LEGAL NOTICE
Neither the European Commission nor any person acting on behalf of the Commission is responsible for the use which
might be made of the following information.
The views expressed in this publication are the sole responsibility of the author and do not necessarily reflect the views
of the European Commission.
A great deal of additional information on the European Union is available on the Internet.
It can be accessed through the Europa server (http://europa.eu).
Cataloguing data can be found at the end of this publication.
Luxembourg: Office for Official Publications of the European Communities, 2008
ISBN
DOI
978-92-79-09455-2
10.2777/84341
Table of contents
Commissioners preface Introduction
14
15
Basic Vaccinology
18
Introduction
BASIC VACCINOLOGY
20
MUVAPRED
24
EPIVAC
26
MuNanoVac
28 CompuVac
Rational Design and Standardised Evaluation of Genetic Vaccines
30
BacAbs
32
MICROBEARRAY
Genome Scale Analysis of the Immune Response against Pathogenic Micro-Organisms Leading
to Diagnostic and Vaccine Candidates and Development of an Integrated Micro Array Platform for Clinical Use
34 DEC-VAC
Development of a Dendritic Cell-Targeted Vaccine against AIDS
36 DC-VACC
Dendritic Cells as Natural Adjustments for Novel Vaccine Technologies
38 THERAVAC
Optimised delivery system for vaccines targeted to dendritic cells
40
AIDS-CoVAC
Generation of a Coronavirus-Based Multigene AIDS Vaccine and Evaluation in a Preclinical SIV Model
42
HIVAB
44
Highly Innovative Strategies for Vaccination to Poverty-Related Diseases
INNOVAC
46
ImmunoGrid
48
VaccTIP
Vaccine Strategies for Combined Targeting of Innate and Adaptive Immune Pathways
50
MVACTOR
Introduction
56
AVIP
58 RMVHIV
Recombinant Measles Virus as a Vector for HIV Vaccines
60
AUTO/ALLO CELL-HIV
62
Pox-gene
64
Allomicrovac
66
VIAV
68
HIV VIROSOMES
70 TIP-VAC
Explaining and Improving Efficacy of Targeted Immunodeficiency Virus-like Particle Vaccines against AIDS
72
EPI-VAC
74
EMVDA
76
Preclinical Studies towards an Affordable, Safe and Efficacious Two-Component Paediatric Malaria Vaccine
PRIBOMAL
78 SME-Malaria
SME Led Malaria Vaccine Initiative
80
MALINV
Differential Expression of Malaria Invasion-Associated Proteins in the Porozoite: Novel Vaccination Strategy
82 TB-VAC
An Integrated Project for New Vaccines against Tuberculosis
86 Neotim
Innate and adaptive immunity in clinical and experimental mycobacterial infection in neonates and infants
88
Vaccines4TB
Genome - and HLA- Wide Scanning and Validation of Cytotoxic CD8 T Cell Responses
against Mycobacterium Tuberculosis
90
ImmunoVacTB
92
FLUVACC
94
FluVac
96
PANFLUVAC
98
Intranasal H5vaccine
102
Universal Vaccine
104
HEPACIVAC
106 DISSECT
Development of Intervention Strategies against SARS in a European-Chinese Taskforce
108 SARSVAC
Immunoprevention and Immunotherapy of SARS Infection
112 NOVADUCK
Novel AI DIVA Recombinant Vaccines for Duck
114
116 SCOOTt
Sustainable Control of Onchocerciasis Today and Tomorrow
118 TRANCHI
T Cell Regulation and the Control of Helminth Infections
120
BOVAC
122 SAVINMUCOPATH
Novel Therapeutic and Prophylactic Strategies to Control Mucosal Infections
by South American Bacterial Strains
124 OMVac
Novel Prevention and Treatment Possibilities for Otitis Media
through the Comprehensive Identification of Antigenic Proteins
126
HEVAR
128 SUPASALVAC
Salmonella-Free Broilers by Live Vaccine-Induced Innate Resistance to Colonisation
and Invasion and Novel Methods to Eliminate Vaccine and Field Strains
130 TRYPADVAC2
Development of an Anti-Disease Vaccine and Diagnostic Tests for African Trypanosomosis
132 CANCERIMMUNOTHERAPY
Cancer Immunology and Immunotherapy
134 DENDRITOPHAGES
Therapeutic Cancer Vaccines
136
LCVAC
138
VITAL
140
Alzheimers Disease-Treatment Targeting Truncated A40/42 by Active Immunisation
MimoVax
Introduction
146
EDCTP
148
EUROPRISE
152 DC-THERA
Dendritic Cells for Novel Immunotherapies
154
ENACT
European Network for the Identification and Validation of Antigens and Biomarkers in Cancer and their
Application in Clinical Cancer Immunotherapy
156
ADVAC-EC
Advanced Vaccinology Training for Scientists from ACC and Developing Countries: From Genomics
to Vaccination Strategies for Communicable Diseases Linked to Poverty
158 NeutNet
Standardisation of HIV Neutralisation Assays to be Used in Vaccine Research and Clinical Trials
160 COINFECT
Malaria and Coinfection: New Insights for Malaria Control
162
EURHAVAC
164
PAHPV-1
166 REBAVAC
Novel Opportunities to Develop Vaccines to Control Antibiotic Resistant Bacteria:
from the Trials Back to the Laboratory
Commissioners preface
10
Janez Potonik
European Commissioner
in charge of Science and Research
introduction
EUROPEAN VACCINE
RESEARCH
Basic Vaccinology
Disease specific
vaccine research
Number of projects
15
42
10
67
Total EC contribution
(million euros)
47
134
229
410
TOTAL
The vast number of projects were STREPs, which constitute more than two thirds of all projects (Fig. 2). However,
it is the IPs that form the core of the EC funding pipeline.
Due to their large size and critical mass of multidisciplinary
participants, the IPs may act as uptake of new discoveries
from smaller, innovative projects, while delivering matured
project results in the other end for clinical applications or
further downstream development by industry organisations, the EDCTP, or public-private partnerships.
The EC believes that tighter collaboration between
multiple stakeholders is central to the advancement of
health science. A key component of the EC Framework
programme is therefore to promote cooperation between
researchers in different countries, and across sectors and
disciplines. While undertaking major research activities,
these partnerships are also contributing to a long-term restructuring of the European research community.
During FP6, a total of 575 research groups participated
in human vaccine research projects (Fig. 3). The vast
majority was located in EU Member States (84%), but
increasing globalisation in health science means that more
and more participants come from other countries, either
European non Member States (7%) or non European
countries (9%).
EDCTP
IP
STREP
NoE
SSA
10
46
Number of participants
16
163
319
79
27
Total EC contribution
(million euros)
200
103
80
24.5
2.8
Number of projects
12
COUNTRY NAME
1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 2 2 2 2 2 2 2 3 3 3 3 4 4 4 4 4 4 4 5 5 6 7 10 12 19 28 30 33 34 47 62 74 78 80
NUMBER OF PARTNERS
Fig. 3: Participants in EC funded human vaccine research in FP6 come from a large number of different countries.
13
14
Acknowledgements: This publication was compiled by Ole F. Olesen and Rachida Ghalouci with the assistance of colleagues in DG RTD of the
European Commission, and with the invaluable help of friends and collaborators in the European vaccine research community. Special thanks
go to Alain Vanvossel (Head of Infectious Diseases), Anna Lonnroth (Deputy Head of Infectious Diseases) and Bernard Mulligan (Deputy Head
of Genomics and Systems Biology) for sustained support to compile this catalogue. Similarly, our thanks go to Virginie Braconnier and Evelyne
Bastin for secretarial assistance, and to Yiannos Contrafouris for technical and editorial support.
15
Basic Vaccinology
Introduction
BASIC VACCINOLOGY
Project Acronym
MUVAPRED
Topical vaccinology
Genomic Vaccinology
Dendritic cells
Explorative vaccinology
EC Contribution
Number of Partners
15.250.000
29
EPIVAC
2.400.000
MuNanoVac
1.505.702
CompuVac
7.969.442
15
BacAbs
2.269.999
MICROBEARRAY
1.401.002
DEC VAC
3.400.000
10
DC-VACC
2.000.000
THERAVAC
2.267.000
AIDS-CoVAC
958.000
HIVAB
950.000
INNOVAC
2.000.000
ImmunoGrid
1.950.000
VaccTIP
1.000.000
MVACTOR
1.000.000
19
MUVAPRED
Acronym: MUVAPRED
Project number: LSHP-CT-2003-503240
EC contribution: 15 250 000
Mucosal Vaccines
for Poverty-Related
Diseases
Duration: 60 months
Type: IP
Starting date: 1 December 2003
Project website: www.mucosalimmunity.org/
muvapred
BACKGROUND
AIMS
20
21
Project Coordinator:
Rino Rappuoli
Rino Rappuoli
Chiron S.r.l.
Via Fiorentina 1
Siena, Italy
Tel: +39-0 57 72 43 414
Email: [email protected]
Partners:
Gordon Dougan
Microbial Pathogenesis
(Team 15: Wellcome Trust Sanger Institute)
Cambridge, England
UK
Jan Holmgren
Gteborg University
Gteborg, Sweden
Nils Lycke
Gteborg University
Gteborg, Sweden
Brigitte Gicquel
Institut Pasteur
Unit de Gntique Mycobacterienne
Paris, France
Nathalie Winter
Institut Pasteur
Unit de Gntique Mycobacterienne
Paris, France
Gianni Pozzi and Donata Medaglini
Universit di Siena
Siena, Italy
Stefan H.E. Kaufmann
Max Planck Institute for Infection Biology
Berlin, Germany
Barbara Ensoli
Istituto Superiore di Sanit
Rome, Italy
Antionio Cassone
Istituto Superiore di Sanit
Rome, Italy
Antonio Lanzavecchia
Institute for Research in Biomedicine (IRB)
Bellinzona, Switzerland
22
Paul Racz
Bernhard-Nocht-Institute for Tropical Medicine
Hamburg, Germany
Thomas Lehner
Kings College
London, England
UK
Kingston Mills
Trinity College
Immune Regulation Research Laboratory
Dublin, Ireland
Peter Andersen
Statens Serum Institute
Copenhagen, Denmark
Francesco Dieli
Consiglio Nazionale delle Ricerche
Istituto di Biomedicina e Immunologia Molecolare
Palermo, Italy
Alberto Mantovani and Mario Negri
Istituto di Ricerche Farmacologiche
Milan, Italy
Andreas Radbruch
German Arthritis Research Centre
Berlin, Germany
David Lewis
St Georges Hospital Medical School
London, England
UK
Peter Sebo
Academy of Sciences of the Czech Republic
Prague, Czech Republic
Philip Marsh
Health Protection Agency
Salisbury, England
UK
23
EPIVAC
Acronym: EPIVAC
BACKGROUND
AIMS
EPIVAC aims to generate an effective and affordable DNAbased preventative and therapeutic vaccine against HIV.
The first goal is the development of a reliable, reproducible
and robust delivery system. The plasmids and genes used
in these studies will allow for the quantitative evaluation of
the efficiency and kinetics of delivery of plasmid DNA into
the epidermis and epidermal cells.
The second goal of the project is to analyse the effect on
the nature and extent of the induced immune response
by different HIV1 antigen expression in different cells of
the epidermis.
24
Project Coordinator:
Ioana Stanescu
E-mail: [email protected]
www.fitbiotech.com
Partners:
Henrik Hellqvist
Silex Microsystems AB
Jrflla 595, Sweden
Evgeny Berik
Estla Ltd
Tartu, Estonia
Charlotte Dalba
EPIXIS SA
Paris, France
David Klatzmann
University of Pierre and Marie Curie
Paris, France
Mart Ustav
Tartu University Institute of Technology
Tartu, Estonia
Roger Le Grand
French Atomic Energy Commission
Paris, France
25
MuNanoVac
Acronym: MuNanoVac
Project number: LSHP-CT-2006-037200
Mucosal Nano-Vaccine
Candidate for HIV
BACKGROUND
AIMS
The MuNanoVac project will assess a new vaccine strategy
to prevent HIV-1 infection based on a biodegradable
synthetic colloidal carrier made of polylactic acid (PLA)
nanoparticles covered with absorbed antigens. Such
nanoparticle-based vaccine carriers allow targeting of
dentritic cells or the transportation of the vaccine through
skin or mucosal epithelial barriers. To amplify the mucosal
immune response, the project will investigate the potential
use of immunomodulator molecules co-adsorbed with
HIV antigens onto PLA particles and will compare two
different immunisation routes, mucosal or sub cutaneous,
in different animal models.After selection of the best
route of immunization associated with the more potent
immunomodulatory molecules, two HIV-l antigens,
trimeric gp140 (clade C) and p24 will be formulated onto
PLA particles for assessing its efficacy as an HIV vaccine
in non human primates.
MuNanoVac wants to demonstrate evidence that a
strong and persistent mucosal immune response for a
given antigen can be reached, using biocompatible PLA
nanoparticles as a versatile vaccine vehicle.
26
www.munanovac.eu
Project Coordinators:
Jacqueline Huet
PHUSIS
Saint Ismier, France
Tel: +33-4 76 52 51 60
E-mail: [email protected]
Bernard Verrier
Partners:
Robin Shattock
St Georges Hospital Medical School
London, England
UK
Teresa Gallart
Hospital Clnic de Barcelona
Barcelona, Spain
Roger Le Grand
Commissariat lEnergie Atomique
Paris, France
Milan Raska
Palack University
Olomouc, Czech Republic
Behazine Combardiere
Universit Pierre et Marie Curie
Paris, France
Ulrike Blume-Peytavi
Charit Universittsmedizin Berlin
Berlin, Germany
27
CompuVac
Acronym: CompuVac
BACKGROUND
Recombinant viral vectors and virus-like particles are considered the most promising vehicles to deliver antigens in
prophylactic and therapeutic vaccines against infectious
diseases and cancer. Several potential vaccine designs exist but their cost-effective development lacks a standardised
evaluation system.
We have now assembled a unique set of vaccines of different class, from viral vector derived vaccines to inert VLPs,
analysed their efficacy with standardised methodologies,
and compared them with an intelligent database. This has
already allowed us to make significant comparisons between different vaccine types and to initiate novel vaccine
design and vaccination regimen. We are now evaluating
prime-boost immunisation regimen with these vectors. We
believe that this should have a significant impact on vaccine development, and notably for those vaccines requiring
prime/boost immunizations.
AIMS
CompuVacs main objective is to set up a standardised approach for the rational development of genetic vaccines.
The process comprises the development of:
a large panel of vaccine vectors representing
various vector platforms and all expressing the
same model antigens;
standardized methodologies for the evaluation of Tand B-cell responses and of molecular signatures
relevant to safety and efficacy;
a database for data storage and analysis of large
data sets;
intelligent algorithms for the rational development
of prime boost vaccination. One of our final goals
is to generate and make available to the scientific
community a tool box and an interactive
database allowing the comparative assessment of
future vaccines. We also aim to validate these tools
by the rationale development of preventive and/or
therapeutic vaccines against HCV.
A secondary objective is to apply these vectors, tools
and methods to the development of a preventive and/
or therapeutic vaccine against the hepatitis C virus
(HCV) incorporating one or more of the platform vectors
expressing the HCV envelope protein.
28
Project Coordinator:
Prof. David Klatzmann
Partners:
Dr Anatoly Sharipo
ASLA Biotech Ltd
Riga, Latvia
Prof. Jacek Blazewicz
Poznan University of Technology
Poznan, Poland
Prof. Thomas Brocker
Institute of Immunology - Ludwig-MaximiliansUniversitt Mnchen
Munich, Germany
Dr Franois-Loc Cosset
Institut National de la Sant et
de la Recherche Mdicale
Lyon, France
Dr Stefan Kochanek
Universittsklinikum Ulm
Ulm, Germany
Dr Penelope Mavromara
Dr Frdric Tangy
Institut Pasteur
Paris, France
Dr Albert Osterhaus
Erasmus MC Rotterdam
Rotterdam, The Netherlands.
Dr Charlotte Dalba
Epixis SA
Paris, France
Dr Alberto Epstein
Universit Claude Bernard Lyon 1
Villeurbanne, France
29
BacAbs
Assessment of Structural
Requirements in
Complement-Mediated
Bactericidal Events:
Towards a Global
Approach to the Selection
of New Vaccine Candidates
BACKGROUND
High throughput cloning and expression of large sets
of genomic ORFs (open reading frames) has become a
preferred industrial strategy for genome-wide searches
of new vaccine candidates. For invasive infections in
particular, the aim is to find proteins eliciting antibodies
capable of binding to the bacterial cell surface and, through
interaction with the complement system, effectively kill the
bacteria. However, current data accumulating from reverse
vaccinology studies indicate that only a small fraction of
surface-exposed proteins appears to elicit antibodies with
bactericidal activity.
Using information generated by reverse vaccinology
projects the project will apply a novel multidisciplinary
approach in identifying the structural requirements
for viable bactericidal vaccine candidates, developing
bioinformatics tools to predict compliance with such
structural requirements. Therefore, a systematic analysis
of sequence, structure, dynamics and interactions of
selected protein targets will be undertaken; serogroup-B
Neisseria meningitidis, a pathogen causing septicemia
and meningitis, for which no effective vaccine exists, will
be used as main model system.
AIMS
The aim of the project is to develop a strategy and
tools to identify early in the vaccine development
process those antigens that could induce production of
bactericidal antibodies. To achieve this goal, the project is
investigating the requirements for productive Ag-Ab-C1q
complex formation, and is taking a multidisciplinary and
comparative approach in studying the structural properties
of a number of these complexes. The MenB vaccine
development project of Novartis Vaccines & Diagnostics
is being taken as a model and a source for useful data and
reagent molecules.
30
Acronym: BacAbs
Project number: LSHB-CT-2006-037325
EC contribution: 2 269 999
Duration: 36 months
Type: STREP
Starting date: 1 January 2007
Project website: www.bacabs.org
Project Coordinator:
Xavier Daura
Partners:
Guido Grandi
Novartis Vaccines and Diagnostics
Siena, Italy
Anatoly Sharipo
ASLA BIOTECH
Riga, Latvia
Etienne Lhermite
Bio-Xtal
Mundolsheim, France
Giorgio Colombo
Consiglio Nazionale delle Ricerche
Milan, Italy
Martin Zacharias
International University Bremen
Bremen, Germany
Martino Bolognesi
Universit degli Studi di Milano
Milan, Italy
Alexandre M J J Bonvin
Universiteit Utrecht
Utrecht, Netherlands
Jos Manuel Mas Benavente
INFOCIENCIA SL
Barcelona, Spain
31
MICROBEARRAY
Genome Scale Analysis of
the Immune Response against
Pathogenic Micro-Organisms
Leading to Diagnostic
and Vaccine Candidates
and Development of an
Integrated Micro Array
Platform for Clinical Use
BACKGROUND
AIMS
The project aimed to identify in micro-organisms of great
medical relevance (M. pneumoniae, C. pneumoniae, L.
pneumophila, coronavirus spp and P. falciparum) a large
collection of surface and secreted proteins, as well as
putative endotoxins. This protein repertoire was produced
as recombinant molecules or as sets of overlapping
synthetic peptides, and printed on array slides. The serum
reactivity of groups of individuals with a proven history of
exposure to the selected micro-organisms was analysed
against the arrayed proteins, to identify diagnostic markers
and correlates of protection.
32
Acronym: MICROBEARRAY
Project Coordinator:
Andrea Crisanti
Partners:
Leszek Rychlewski
Bioinformatics Laboratory
Poznan, Poland
Antonio Cassone
Instituto Superiore di Sanita
Rome, Italy
John Attridge
Chelsea Instruments Ltd
London, England
UK
Franck Breitling
Deutsches Krebsforschungszentrum
Stiftung Des Oeffentlichen Rechts
Heidelberg, Germany
Diagnostic Matrices Ltd
London, England
UK
Kevin Bryne
Mikura Ltd
Sussex, England
UK
Vincent Monchois
ProteineXpert
Grenoble, France
Microtest Matrices Ltd
London, England
UK
33
DEC-VAC
Acronym: DEC-VAC
Development of a
Dendritic Cell-Targeted
Vaccine against AIDS
BACKGROUND
AIMS
The project aims to develop an innovative anti-HIV vaccine
able to enhance uptake and presentation of antigens by
dendritic cells (DCs). The projects strategy to enhance
antigen uptake and presentation by DCs can be applied
to protein vaccines, peptide vaccines, DNA vaccines and
viral vector vaccines, thus providing a broad vaccination
platform. Providing a novel HIV vaccine strategy for
safety and efficacy studies in humans by the end of the
project is an important aim. Therefore, the scientific and
technological objectives are to determine the following for
DC-targeted protein, DNA, and viral vector vaccines:
34
Project Coordinator:
Klaus berla
Ruhr-Universat
Bochum, Germany
E-Mail: [email protected]
Partners:
Ralph Steinman
Rockefeller University
New York, USA
Britta Wahren
Swedish Institute for Infectious Disease Control
Stockholm, Sweden
Christiane Stahl-Hennig
Deutsches Primatenzentrum
Gttingen, Germany
Ralf Ignatius
Abt. Med. Mikrobiologie u. Infektionsimmunologie
Berlin, Germany
Paul Racz
Bernhard-Nocht-Institute for Tropical Medicine
Hamburg, Germany
Mariagrazia Uggucioni
Institute for Research in Biomedicine
Bellinzona, Switzerland
Ralf Wagner
Geneart GmbH
Regensburg, Germany
Heribert Stoiber
Innsbruck Medical University
Innsbruck, Austria
Lazare Kaptue
University of Yaounde I
Cameroon, Africa
35
DC-VACC
Acronym: DC-VACC
Dendritic Cells
as Natural Adjustments
for Novel Vaccine
Technologies
BACKGROUND
The immune system of vertebrate animals has evolved
to respond to different types of perturbations, such as
pathogens, whilst limiting self-tissue damage. Initiation of
the immune response is accomplished by unique antigenpresenting cells, called dendritic cells (DCs) that rest until
encountering foreign microorganisms or inflammatory
stimuli. Early-activated DCs trigger innate immune
responses that represent the first line of defence against
invading pathogens. Activated DCs subsequently prime
antigen-specific immune responses, clearing the infection
and giving rise to immunological memory.
AIMS
The projects aims were to:
develop novel vaccine technologies. Early clinical
trials indicated that antigen-pulsed DCs have great
potential for treating cancer. In situ DC targeting
was developed for use as vaccines in infectious
diseases and cancer;
create tools and methodologies for the development
of DC vaccine technology, including construction
of viral and bacterial vectors, modification of RNA,
peptides and proteins, and antibody development
for targeting of the DC receptor repertoire. A
comparison was undertaken for peptides, proteins,
RNA, DNA and antigen modifications that allow
presentation via MHC molecules;
define optimal reagents and protocols for
maturation and activation of mouse and
human DC in vitro for use in vaccination. The
optimisation of protocols for both species was
compared in order to facilitate information from
preclinical models being transferred to clinical
trials for future projects. The aim was to obtain
a clear understanding of how DCs induce proinflammatory versus anti-inflammatory cytokines,
chemokines and their receptors, and how they
activate CD4+ and CD8+ T cells. It was essential
that such activation signals were tested for their
36
Project Coordinator:
Anne OGarra
E-Mail: [email protected]
Partners:
Leonardo Biondi
Biopolo Scrl - c/o IFOM
Milan, Italy
Vicenzo Cerundolo
University of Oxford
Oxford, England
UK
Carl Figdor
Muriel Moser
Universit Libre de Bruxelles
Gosseliers, Belgium
Paola Ricciardi Castagnoli
University of Milano-Bicocca
Milan, Italy
Gerold Schuler
University Hospital of Erlangen
Erlangen, Germany
Anna Ranghetti
SEKMED S.R.L.
Milan, Italy
Catherine De Greef
Brucells SA
Brussels, Belgium
37
THERAVAC
Acronym: THERAVAC
BACKGROUND
AIMS
The projects aims are as follows:
production of a GMP batch of ACT carrying the
melanoma tyrosinase epitope suitable for a phase
1/2 clinical trial;
the detailed toxicology assessment of this GMP
batch of ACT;
a phase 1/2 clinical trial in melanoma patients of
this GMP batch of ACT carrying the tyrosinase
melanoma CD8+ T cell epitope;
the understanding of the interaction of ACT with
dendritic cells at molecular and atomic details;
the detailed understanding of the mechanism of
PPV-VLP specificity and efficacy;
the engineering of improved vaccine delivery
molecules based on the improved understanding of
their functional mechanism.
38
Project Coordinator:
Claude Leclerc
Institut Pasteur
25 Rue du Dr Roux
Paris, France
Tel: +33-1 45 68 86 18
E-Mail: [email protected]
Partners:
Anita Lewit-Bentley
CNRS
Cachan, France
Rino Rappuoli
Novartis
Sienna, Italy
Paloma Rueda
INGENASA
Madrid, Spain
Peter Sebo
Institute of Microbiology
Prague, Czech Republic
Daniel Ladant
Institut Pasteur
Paris, France
Benot Van den Eynde
ICP
Brussels, Belgium
Dorothy Xing
NIBSC
Enfield, England
UK
39
AIDS-CoVAC
Acronym: AIDS-CoVAC
Generation of a
Coronavirus-Based
Multigene AIDS Vaccine
and Evaluation in a
Preclinical SIV Model
BACKGROUND
AIMS
The projects aims are as follows:
to develop a coronavirus-based multigene vaccine
that specifically targets DCs;
to evaluate the novel approach through preclinical
testing in a simian model;
to expand the understanding of the molecular
biology of coronavirus and DC interaction, and
exploit this knowledge to improve the novel virus
vector system.
40
Project Coordinator:
Burkhard Ludewig
Kantonsspital St Gallen
St Gallen, Switzerland
E-Mail: [email protected]
Partners:
Seighart Sopper
Deutsches Primatenzentrum GmbH
Gttingen, Germany
Maria Foti
Universit degli Studi di Milano-Bicocca
Milan, Italy
41
HIVAB
Generation of Broadly
Cross-Neutralising
Antibodies for Innovative
Active-Passive
HIV Vaccination Strategies
Based on Modified Ig-Gene
Transgenic Mice
BACKGROUND
AIMS
The projects aims were as follows:
to develop transgenic mice with distinct genetic
alterations;
to develop antigens based on synthetic HIV
envelope genes;
to identify and later produce novel neutralising
antibodies following humanisation.
This project used two experimental strategies, separately
and in combination: the first was used to produce mice
with germ line-modified immunoglobulin genes, which
introduce extended mobility into the immunoglobulin protein
backbone; the second used HIV receptor-transgenic mice
for immunisation, to favour receptor mediated transitory
stages of the viral envelope.
42
Acronym: HIVAB
Project Coordinator:
Hans Wolf
University of Regensburg
Regensburg, Germany
E-Mail: [email protected]
Partners:
Ralf Wagner
University of Regensburg
Regensburg, Germany
Thomas Hehlgans
University of Regensburg
Regensburg, Germany
Simon Jeffs
Imperial College of Science,
Technology and Medicine (ICSTM)
London, England
UK
Renate Kunert
Institute for Applied Microbiology
(IAM/BOKU))
Vienna, Austria
Frank Notka
Geneart GmbH
Regensburg, Germany
Hermann Katinger
Polymun Scientific GmbH
Vienna, Austria
43
INNOVAC
Acronym: INNOVAC
Project number: LSHP-CT-2006-036871
Highly Innovative
Strategies for Vaccination
to Poverty-Related
Diseases
Sciences/AcademicStaff/Cutting/
cutting.html
BACKGROUND
AIMS
The projects aim was to develop three platform
technologies that were used for developing innovative
methods of vaccination against tuberculosis (TB) and
malaria. The research platforms included:
bacterial spores robust and heat-stable
bioparticles with proven efficacy as mucosal
vaccines;
intracellular and invasive bacteria including E. coli
strains and Mycobacterium bovis (rBCG);
S-layer protein conjugates and S-layer protein
coated liposomes.
The project tested and evaluated vaccination strategies
using recombinant systems, some in their infancy and
others at a more advanced stage of development. This
included construction of vaccine vehicles, their evaluation
in animal models, challenge experiments and safety tests,
where appropriate, in order to bring potential vaccines to
the clinical evaluation stage.
44
Project Coordinator:
Simon M Cutting
Partners:
Rocky Cranenburgh
Cobra Biomanufacturing
Keele, England
UK
Alexander Matis
Nano S Biotechnologie
Vienna, Austria
Riccardo Manganelli
University of Padova,
Padova, Italy
Giovanni Delogu
Universita Cattolica del Sacro Cuore
Rome, Italy
Volker Heussler
Bernhard Nocht Institute for Tropical Medicine
Hamburg, Germany
Nguyen Thu Van
Vabiotech
Hanoi, Vietnam
45
ImmunoGrid
Acronym: ImmunoGrid
The European
Virtual Human Immune
System Project
BACKGROUND
AIMS
46
Project Coordinator:
Elda Rossi
CINECA
Bologna, Italy
Tel: +39-0 51 61 71 515
E-Mail: [email protected]
Partners:
Vladimir Brusic
University of Queensland
Queensland, Australia
Filippo Castiglione;
Massimo Bernaschi
Consiglio Nazionale delle Ricerche
Rome, Italy
Santo Motta;
Francesco Pappalardo
Universita di Catania
Catania, Italy
PierLuigi Lollini
Alma Mater Studiorum Universita di Bologna
Bologna, Italy
Sren Brunak
Denmark Technical University
Lyngby, Denmark
Marie-Paule Lefranc
Centre National de la Recherche Scientifique
Montpellier, France
David Moss; Adrian Sheperd
Birkbeck College
London, England
UK
47
VaccTIP
Acronym: VaccTIP
Vaccine Strategies
for Combined Targeting
of Innate and Adaptive
Immune Pathways
BACKGROUND
AIMS
The project aimed to:
produce polymeric CD40L (Mega-CD40L) and
bacterial Flagellin as two novel adjuvants targeting
the innate and adaptive immune pathways;
construct novel alphavirus-based vaccine vectors
which stimulate innate immunity and further elevate
humoral and cellular immune responses, and to
bring technology forward in view of future implementation as global vaccines in developing countries;
analyse innate immune reactions both in human
and murine settings;
evaluate the effect on T and B cell responses to
specific antigens in human and murine models;
evaluate the adjuvanticity on recombinant alphaand poxvirus vectors expressing HIV-1 antigens.
48
Project Coordinator:
Peter Liljestrom
Karolinska Institutet
Stockholm, Sweden
Tel: +46-8 45 72 550
E-Mail: [email protected]
Partners:
Giuseppe Pantaleo
University of Lausanne
Lausanne, Switzerland
Stphane Demotz
Apoxis SA
Lausanne, Switzerland
Jean-Claude Sirard
Institut de Biologie
INSERM BP447
Lille, France
Mariano Esteban
Centro Nacional de Biotechnologia
Madrid, Spain
49
MVACTOR
Acronym: MVACTOR
Host Immune
Activation-Optimised
Vaccinia Virus Vectors
for Vaccine Development
BACKGROUND
Poxviruses engineered to express foreign genes are
recognised as potent delivery systems for heterologous
antigens and as candidate vaccines against a wide spectrum of human and animal diseases. One of these vectors,
the safety-tested highly attenuated modified vaccinia virus
Ankara (MVA) (a product of European vaccine research),
serves worldwide as the vaccinia virus strain of choice
for clinical investigations in experimental vaccination
against AIDS, tuberculosis, malaria or tumour diseases.
At present the MVACTOR consortium represents the
only European research network with prime expertise in
poxvirus research. Recent work by the project partners
significantly increased the knowledge based on versatile
poxvirus gene functions, so as to regulate virus-host
interactions and to modulate innate and adaptive host cell
immune responses.
With this project, MVA will serve as the basis to develop
a new generation of potent viral vector vaccines with
optimised host immune activating properties. Specifically,
the projects focus is to identify and/or inactivate MVA
gene functions that have immune inhibitory potential (e.g.
counteracting interferons, interleukins, CC chemokine
functions) and potentiate vector vaccine performance by
virion modification and host cytokine co-expression.
AIMS
The project aims on the development of new generation
potent viral vector vaccines with optimised host immuneactivating properties based on the highly attenuated
modified vaccinia virus Ankara (MVA). Specifically, it will
identify and characterise MVA gene functions that have
unknown and/or potentially specific immune inhibitory
capacity. Basing on these results, it will then potentiate
vector vaccine performance by rational genetic engineering
of the MVA vector through:
precise inactivation of viral immune evasion genes;
specific activation of host innate immune
responses;
50
Project Coordinator:
Gerd Sutter
Paul-Ehrlich-Institut
Langen, Germany
E-Mail: [email protected]
Partners:
Geoffrey L Smith
Imperial College of Science,
Technology and Medicine
London, United Kingdom
Mariano Esteban
Consejo Superior de Investigaciones Cientficas
Madrid, Spain
Rafael Blasco
Institute Nacional de Investigacin y Tecnologia
Madrid, Spain
51
Diseases Specific
Vaccine Research
Introduction
Budget Total = 136,029,199
Non-infectious Diseases (19,836,426)
DISEASE SPECIFIC
VACCINE RESEARCH
COM(2005) 179 of 27.04.2005 FROM THE COMMISSION TO THE COUNCIL AND THE EUROPEAN PARLIAMENT: A European Programme for Action to
Confront HIV/AIDS, Malaria and Tuberculosis through External Action (2007-2011)
53
EMERGING EPIDEMICS
Influenza pandemics such as the Spanish flu of 1918 to
1919, which killed between 50 and 100 million people
around the world, is a reminder that new viral diseases
can suddenly appear or re-appear with short notice
and dramatic consequences for global public health.
A newer example stems from spring 2003, when a
hitherto unknown infectious disease, SARS (Severe
Acute Respiratory Syndrome), was reported in China,
and quickly spread to Hong Kong, Taiwan, Singapore,
Vietnam and Canada before it was contained. The EC
has acknowledged the importance of vaccine research
in emerging infectious diseases by supporting a number
of activities in this area. The largest number of projects
is focused on pandemic influenza, thereby reflecting the
current shortcomings of available flu vaccines. The main
challenge is that flu virus changes so rapidly that vaccines
formulated against one strain may be ineffective the
following year. The largest EC project to address this is
FluVacc, which is developing an improved technology for
quickly producing new live attenuated influenza vaccines
based on reverse genetics.
The other large integrated project in emerging epidemics
is Hepacivac, which aims to develop prophylactic and
therapeutic vaccine candidates that can elicit a strong
and specific T cell response against the hepatitis C virus.
54
NON-INFECTIOUS DISEASES
The possibility of creating vaccines for a host of noninfectious diseases such as cancer, neurodegenerative
diseases and autoimmune diseases provides one of the
most exciting fields for current vaccine research. It holds
the potential to meet a huge unmet medical need for
some diseases, while for other diseases it may provide
a future alternative to traditional treatments that often are
costly, only partially effective and sometimes associated
with several adverse effects. With the exception of a
vaccine project against Alzheimers disease (MIMOVAX),
EC efforts in this area have been concentrated on cancer
vaccines. The projects address identification of specific
tumour antigens that are present in cancer cells, such
as in leukaemia, and thereby provide potential immune
targets for which immunogenic vaccines may possibly be
designed. Other projects are targeting some of the major
cancer types, such as melanoma, carcinoma, and lung
cancer. This includes the CANCERIMMUNOTHERAPY
consortium, which comprises 22 partners that have
received a total EC contribution of EUR 12 million to
develop a safe and efficient therapeutic vaccine against
cancers such as melanoma. This will initially be done by
comparing the effect of various types of vaccines, such as
peptides and RNA with different types of immunological
adjuvants and DCs. The field of vaccines for non-infectious
diseases is highly promising, but it should be kept in mind
that there are still many roadblocks to be overcome. Some
of the basic discoveries and necessary technologies may
not yet be sufficiently matured. As such, some of the EC
funded activities in basic vaccinology will undoubtedly
also benefit the future advancement of vaccines for noninfectious diseases.
55
AVIP
Acronym: AVIP
Project number: LSHP-CT-2004-503487
AIDS Vaccine
Integrated Project
BACKGROUND
Vaccines based on viral structural products (Env/Gag/
Pol) alone have failed to prevent infection by Human
Immunodeficiency Virus (HIV)/Simian Immunodeficiency
Virus (SIV). New strategies have recently been developed
aimed at blocking virus replication and onset of HIV/SIV
in the absence of sterilising immunity. Control of virus
replication may modify the virus-host interaction, favoring
the host immune response, providing protection from
disease progression and reducing virus transmission
to healthy individuals. This strategy, useful for both
preventive and therapeutic interventions, should include
both nonstructural and structural viral products, since
vaccines targeting viral structural products (Env/Gag/Pol),
as well as viral regulatory gene products (Tat/Rev/Nef),
should be superior at inducing immune responses to both
early and late viral products.
AIMS
The aims of the project are:
to generate novel HIV-1 vaccine candidates to be
tested in phase I preventive and therapeutic trials in
Europe within the five-year programme. To achieve
this goal, four novel vaccines have been selected
from a larger pool based on two criteria: the
combination of HIV regulatory (Tat and/or Rev, and/
or Nef) with structural (Env and/or Gag/Pol) genes/
products, and the advanced stage of development
of single components, including efficacy studies in
monkeys and new murine efficacy models;
to conduct parallel preventive and therapeutic
phase I trials in Europe with the four novel
combined vaccines;
to perform feasibility studies and technology
transfer in Developing Countries (DCs) for phase II/
III trials;
to carry out training in EU and in DCs through the
AVIP International School;
to ensure community involvement both in EU and
DCs.
56
PRD-AIDS
Mauro Magnani
University of Urbino
Urbino, Italy
Ioana Stanescu
FIT Biotech Oyj Plc
Tampere, Finland
Volker Erfle
Helmholtz Center Munich
Neuherberg, Germany
Rino Rappuoli / Susan Barnett
Novartis Vaccines and Diagnostics Srl
Siena, Italy
Project Coordinator:
Dr Barbara Ensoli
Partners:
Britta Wahren
Microbiology and Tumorbiology Center, Karolinska Institute
Stockholm, Sweden
Frances Gotch
Imperial College, Chelsea and Westminster Hospital
London, England
UK
Eftyhia Vardas
Perinatal HIV Research Unit of the Wits Health Consortium
Sowetho, South Africa
Richard Glashoff
University of Stellenbosch Medical School
Tygerberg, South Africa
Roger Le Grand
SNV/DRM/SDV/CEA Laboratoire dImmuno-Pathologie
Experimentale (LIPE)
Fontenay-Aux-Roses, France
Riccardo Gavioli
University of Ferrara
Ferrara, Italy
Carlos A Guzman
Helmholtz-Zentrum fr Infektionsforschung GmbH
Braunschweig, Germany
PRD-AIDS
57
RMVHIV
Acronym: RMVHIV
Recombinant Measles
Virus as a Vector
for HIV Vaccines
BACKGROUND
This project aims at demonstrating the safety and
immunogenicity of a novel recombinant measles virus
(MV) vector for use as an AIDS vaccine. The vector is
replication competent in vivo and is derived from a widely
used measles vaccine strain (Schwarz), which is known
to induce very long-lasting immunity. This novel vector
potentially offers a unique combination of safety and
potency. The recombinant HIV MV vectors will express
three relatively conserved HIV proteins (i.e. Gag, Pol, Nef)
from HIV clade B and A strains. A Good Manufacturing
Practices (GMP) compatible production process for the
recombinant MV vector will be developed and a GMP lot
will be produced for two clinical studies.
The first study will evaluate the safety profile of the
MV vector, while the second study will also assess the
immunogenicity in MV-immune volunteers. With these
two clinical studies, the project will specifically address
potential shedding of the recombinant vector into the
environment and the potential negative impact of preexisting MV immunity.
The measles vaccine, a live attenuated strain of MV, is
one of the safest human vaccines available and has been
given to billions of children since the 1960s. However,
because of the inadequate distribution of the vaccine in
developing countries, there are still 45 million cases of
measles and 800 000 child deaths per year worldwide.
The Pasteur Institute has developed an MV vector based
on the Schwarz strain, the safest and most widely used
vaccine strain. The consortium is proposing to construct
recombinant MV vectors that express the HIV-1 clade B
and A Gag, Pol, and Nef proteins. These proteins possess
highly conserved regions that have been shown to be
the target of CD8-positive cells, and thereby constitute a
promising antigenic composition for an HIV vaccine. It has
been demonstrated that CD8-positive cells from individuals
infected with virus strains from different clades are highly
cross-reactive with respect to the Gag, Pol, and Nef
proteins. By assuming a similar cross-reactivity for vaccine-
58
PRD-AIDS
AIMS
The objective of RMVHIV is the demonstration of safety
and immunogenicity of a recombinant HIV MV vector
in adult HIV-uninfected volunteers. This includes the
identification of a suitable dose of recombinant MV, the
demonstration of an acceptable reactogenicity profile,
and the characterisation of potential virus shedding.
Furthermore, the vaccine will have to induce significant
levels of HIV-specific CD8-positive cells in volunteers with
pre-existing immunity, and ideally also measurable CD4positive cell and antibody responses.
Project Coordinator:
Gerald Voss
Gerald Voss
GlaxoSmithKline Biologicals
Rixensart, Belgium
Tel: +32-2 65 68 243
E-Mail: [email protected]
Partners:
Frdric Tangy
Institut Pasteur
Paris, France
Geert Leroux-Roels
Gent Universiteit
Ghent, Belgium
Odile Launay
Centre Cochin-Pasteur dEssais Vaccinaux,
Hpital Cochin
Paris, France
David Lewis
St Georges, University of London
London, England
UK
Neil Almond
National Biological Standards Board
Potters Bar, England
UK
PRD-AIDS
59
AUTO/ALLO CELL-HIV
Development of a Novel
Therapeutic HIV-1 Vaccine:
Horizontal Gene Transfer
by Using Apoptotic
HIV-1 DNA Containing
Activated T Cells
BACKGROUND
AIMS
The project has discovered that genes can be horizontally
transferred to neighbouring cells by the uptake of
apoptotic cells, which also allows transfer of proteins
leading to cross-presentation of antigens. The aim is to
develop a therapeutic HIV vaccine using apoptotic cells
as the antigen delivery system. In order to achieve this the
project is performing the following actions:
carrying out safety and immunogenicity studies in
macaques;
optimising the techniques for production of
AutoCell/AlloCell in a good management practice
(GMP) certified Cell Therapy Centre;
producing individualised prototype AutoCell/AlloCell
compositions;
launching a phase I/II clinical trial.
Anti-retroviral treatment leads to reconstitution of immune
responses to many pathogens, but it does so without
the emergence of HIV-specific responses. Anti-retroviral
treatment also allows patients to respond to immunisation
using recall antigens and neo-antigens. Hence, it may be
feasible to induce a novel adaptive HIV-1-specific immune
response by therapeutic vaccination, which would also
allow the patient to stop anti-retroviral medication.
60
PRD-AIDS
Project Coordinator:
Anna-Lena Spetz
Karolinska Institutet
Stockholm, Sweden
E-Mail: [email protected]
Partners:
Gerrit Koopman
Biomedical Primate Research Center
Rijswijk, Netherlands
Eva-Maria Fenyo
Lund University
Luc Perrin
Geneva University Hospital/Geneva Medical School
Geneva, Switzerland
Jan Andersson
Brigitte Autran
INSERM U543
Paris, France
PRD-AIDS
61
Pox-gene
A Combined Pox-Virus/
Lentiviral Vector System
to Treat HIV Infection;
Immunisation and Direct
In Vivo Gene Transfer
in T-Lymphocytes
Acronym: Pox-gene
Project number: LSHP-CT-2005-018680
EC contribution: 1 180 000
Duration: 36 months
Type: STREP
Starting date: 1 December 2005
Project website: www.bprc.nl
BACKGROUND
AIMS
The project aims to develop a combined vaccination/
gene therapy protocol for the treatment of HIV infection.
Currently available technology for genetically modifying
MVA pox-viruses was exploited to create a vector that
expresses both HIV-1 proteins and an HIV inhibitory
lentiviral construct. Target cells infected with MVA therefore
express not only HIV-1 proteins capable of stimulating
antigen specific T cells, thereby boosting anti-HIV-1
immune responses, but will also release lentiviral particles
capable of transducing antigen stimulated T cells with an
anti-viral gene that protects them from HIV-1 infection.
62
PRD-AIDS
Project Coordinator:
Gerrit Koopman
Partners:
PRD-AIDS
63
Allomicrovac
Acronym: Allomicrovac
A Combined
Microbicidal-Immunising
Strategy Against SIV
and HIV Infection
BACKGROUND
AIMS
The project aims to utilise one cohort of macaques for
three HIV-1/SIV preventive methodologies in microbicide,
preventive and therapeutic immunisation. Macaques
that are not infected are to be evaluated for preventive
immunity; infected animals are utilised for therapeutic
immunisation two months after infection.
Emphasis is on determining whether a trimolecular
construct of MHC antigens combined with microbial
HSP70 and extracellular CCR5 peptides can be utilised
in microbicidal, preventive and therapeutic immunisation.
Combining a short term microbicide with a long term
preventive vaccine will deal with the problem of repeated
applications of microbicides before sexual intercourse.
64
PRD-AIDS
By early 2008, the HSP70-CCR5 construct for immunization was produced, consisting of dextramers of macaque
MHC A1, A4 and A8 alleles and dextramers linked to
HSP70 and CCR5 N terminal and loop 1 and 2 peptides.
The final vaccine constructs was assembled in the molar
ratio of 1 mol Dextran / 10 mol MHC / 8 mol HSP70CCR5tripeptide. Two groups of macaques were treated vaginally
by the dextramers MHC-HSP70-CCR5 peptide construct
at 2-weekly intervals, followed 30min. later by 50TCID of
SIVmac251. A 3rd group of macaques was treated with
saline prior to treatment with SIVmac251 and a 4th was
a naive control group. The macaques are monitored for
innate immune factors, vaginal and serum antibodies, CC
chemokines, cytokines and T cell functions. The investigation should be completed by the end of the year, when
the results will be analysed and published.
Project Coordinator:
Thomas Lehner
Guys Hospital
London, England
UK
Telephone: +44 207 188 3072
E-Mail: [email protected]
Partners:
Gunnel Biberfeld
Karolinska Institute
Solna, Sweden
Jrgen Schller,
DakoCytomation,
Glostrup, Denmark
Mahavir Singh
Lionex Diagnostics and Therapeutics
Braunschweig, Germany
Rigmor Thorstensson
Swedish Institute
for Infectious Disease Control
Solna, Sweden
Robert Vaughan,
Guys Hospital
London, England
UK
Yufei Wang,
Guys Hospital
London, England
UK
PRD-AIDS
65
VIAV
Acronym: VIAV
Project number: LSHP-CT-2004-012188
Very Innovative
AIDS Vaccine
BACKGROUND
There are indications that the HIV Tat protein increase HIV
cell absorption, infectivity and tropisms by interacting with
components of the HIV membrane and envelope protein
(Env). Because the interaction between Tat and Env is
believed to increase the generation of complex-specific,
neutralisation-sensitive epitopes and/or the stabilisation
of cryptic and/or transiently exposed Env epitopes, a
vaccine based on Tat-Env complexes was considered
likely to generate protective immune responses against
vulnerable viral targets.
AIMS
The intention of this project was to develop a highly innovative Tat-Env complex-based vaccine capable of inducing
cross-clade neutralising antibodies against novel, neutralisation sensitive Env epitopes to prevent HIV infection and/
or AIDS progression. This was achieved using new antigen
design as a result of novel virological, immunological and
modelling data from the VIAV consortium.
66
PRD-AIDS
Project Coordinator:
Flavia Ferrantelli
Partners:
Banci Lucia
Consorzio Interuniversitario
Risonanze Magnetiche di Metalloproteine
Florence, Italy
Eva Maria Fenyo
Lunds Universitet
Lund, Sweden
Dag Helland
Universitetet i Bergen
Bergen, Norway
Elisa Gargiulo
DIATHEVA
Fano, Italy
PRD-AIDS
67
HIV VIROSOMES
Development of
a New Vaccine
against HIV:
Virosomes
Incorporating
HIV Proteins
BACKGROUND
AIMS
The objective of the project was to develop a preventive
HIV vaccine using a focused straight-forward approach.
The aim was to incorporate native proteins derived from
primary HIV strains into liposomes generated by a novel
large scale liposomal technology. Alternatively, recombinant
HIV proteins should be used. This project included:
a. stabilisation of the native structure and
conformation of native and recombinant HIV
envelope proteins in liposomes
b. selection of candidate vaccines in small animal
immunisation studies
c. establishment of immunogenicity, and eventually,
efficacy in the rhesus macaque model
d. establishment of a GMP compliant process suitable
for production of clinical material
68
PRD-AIDS
Project Coordinator:
Gabriela Stiegler
Partners:
Biomedical Primate Research Centre (BPRC)
Department of Virology
The Netherlands
European Molecular Biology Laboratory (EMBL)
Grenoble Outstation
France
Institute of Applied Microbiology (IAM)
University of Natural Resources
and Applied Life Sciences
Austria
Polymun Scientific Immunbiologische
Forschung GmbH
Austria
PRD-AIDS
69
TIP-VAC
Acronym: TIP-VAC
Explaining and
Improving Efficacy of
Targeted Immunodeficiency
Virus-like Particle
Vaccines against AIDS
BACKGROUND
AIMS
The aims of the project were to:
determine the efficacy of the VLPs in a larger number of
animals;
understand better the requirements for and the
mechanisms of protection;
further improve the targeted VLPs.
The project developed a targeted immunodeficiency viruslike particle vaccine (VLP), which has a heterologous
viral surface protein incorporated in the membrane of the
particle. This should increase uptake and presentation
of VLPs by dendritic cells. A pilot vaccination experiment
in the SIV/macaque model provided strong protection
against challenge with a pathogenic SIV.
70
PRD-AIDS
Project Coordinator:
Klaus berla
Ruhr-University
Bochum, Germany
Tel: +49-2 34 32 23 189
E-Mail: [email protected]
Partners:
Paul Racz
Bernhard-Nocht-Institute for Tropical Medicine
Hamburg. Germany
Ralph Steinman
Henry G Kunkel
The Rockefeller University
New York, USA
Heribert Stoiber
Medical University Innsbruck
Innsbruck, Austria
Ralf Ignatius
Charit University Medicine Berlin
Berlin, Germany
Mariagrazia Uguccioni
Institute for Research in Biomedicine
Bellinzona, Switzerland
Christiane Stahl-Hennig
Deutsches Primatenzentrum (DPZ) GmbH
Gttingen, Germany
PRD-AIDS
71
EPI-VAC
Acronym: EPI-VAC
Identification of Novel
Epitopes as HIV-1
Vaccines Candidates
BACKGROUND
AIMS
This project aimed to select epitopes that mimic
neutralisation-sensitive domains of HIV-1 envelope and
may function as candidate HIV-1 vaccines and. It carried
out the following strategies to meet its objective:
screening of random peptide libraries with novel
MAbs that neutralise primary HIV-1 isolates
assigned to distinct clades;
designing 30 to 40 amino acid peptides that mimic
discontinuous regions of gp120 and gp41 that are
sensitive to neutralisation by antibodies and are
conserved among HIV strains of distinct clades.
In order to meet the challenge, EPI-VAC developed pools
of innovative immunogens that mimic conserved regions
of the viral envelope and are shared by a substantial
percentage of primary viral isolates assigned to distinct
clades from disease progression, if volunteers are
exposed to heterologous isolates (1-3).
72
PRD-AIDS
Project Coordinator:
Giuseppe Scala
E-Mail: [email protected]
Partners:
Vincenzo Pavone
University of Naples Federico II
Naples, Italy
David Davis
Foundation Biomedical
Primate Research Center
Rijswijk, Netherlands
Gabriela Stiegler
Polymun Scientific
Immunbiologische Forschung GmbH HIV Research
Vienna, Austria
Boris Ferko
University of Applied Life Sciences
and Natural Resources
Institute of Applied Microbiology
Vienna, Austria
PRD-AIDS
73
EMVDA
Acronym: EMVDA
BACKGROUND
There is considerable optimism that a malaria vaccine can
be developed. This is based on the fact that acquisition
of immunity induced by natural infection does eventually
prevent mortality and provide protection against clinical
disease. There has been interest in the development of
malaria vaccines for over 30 years, with the initial research
emphasis on attenuated or killed whole organisms,
and more recently on subunit-based approaches. The
malaria parasite is a complex organism with an elaborate
lifecycle; as a consequence much effort has been devoted
to identifying molecules that stimulate host immunity
and identifying protective components of that immune
response. In parallel, research on delivery technologies
has sought to develop ways to evoke protective immune
responses by active immunisation.
Three stages of the malaria parasite lifecycle are targeted
for vaccine development: the pre-erythrocytic, the asexual
and the sexual blood stages. This project is very largely
focused on the asexual blood stage that is responsible for
the disease. This is the area in which European laboratories
are probably most globally competitive, and it allows them to
focus relatively limited resources to the greatest advantage.
The projects strategy includes the development of new
engineered and improved synthetic antigens, for example
with amino acid sequences from two or more antigens or
with modifications to improve immunogenicity.
AIMS
The goal of the project is to systematically develop and
test malaria vaccines by comparative and continuous
evaluation of candidates. The best malaria vaccine
candidates are being selected through collaborations
with two SMEs, eight European malaria vaccine research
centres, the European Malaria Vaccine Initiative (EMVI)
and the African Malaria Network (AMANET). These
vaccines will be developed further within a process that
includes antigen validation, as well as the creation of a
vaccine development rationale and early proof-of-principle
clinical trials.
74
PRD-Malaria
To develop a vaccine for malaria a scientific and technological structure supported by effective management
has to be established in order to move candidate malaria parasite antigens through five stages of preclinical
and clinical testing. Individual candidates are at different
stages of development. Stringent go/no-go criteria are
being used to assess and compare competing antigens,
and delivery systems to focus resources on to the most
credible vaccine candidates. Emphasis and resources
are being focused on moving candidate vaccines into
clinical trials.
Project Coordinator:
Dr Odile Leroy
Partners:
Michael Theisen
Statens Serum Institut
Copenhagen, Denmark
Reinhard Glck
CSO Etna Biotech
Bern, Switzerland
Rinaldo Zurbriggen
Pevion Biotech
Bern, Switzerland
Adrian Hill
The Wellcome Trust Centre for Human Genetics
Oxford, England
UK
Anthony Holder
National Institute for Medical Research (NIMR)
London, England
UK
Roma Chilengi
African Malaria Network Trust (AMANET)
Tanzania, Africa
Gerd Pluschke
Swiss Tropical Institute
Basel, Switzerland
Klavs Berzins
The Wenner-Gren Institute, Stockholm University
Stockholm, Sweden
Franois Spertini
Centre Hospitalier Universitaire Vaudois
Lausanne, Switzerland
Peter Kremsner
Eberhard-Karls Universitt Tbingen
Tbingen, Germany
Alan Thomas
Biomedical Primate Research Centre
Rijswijk, Netherlands
David Cavanagh
University of Edinburgh
Edinburgh, Scotland
UK
Robert Sauerwein
University Hospital Faculty of Medical Sciences
Nijmegen, Netherlands
PRD-Malaria
75
PRIBOMAL
Preclinical Studies
towards an Affordable,
Safe and Efficacious
Two-Component
Paediatric Malaria
Vaccine
Acronym: PRIBOMAL
Project number: LSHP-CT-2006-037494
EC contribution: 2 345 358
Duration: 36 months
Type: STREP
Starting date: 1 January 2006
Project website: www.crucell.com
BACKGROUND
AIMS
The consortium aims to show in preclinical studies
the safety and efficacy of a novel and affordable twocomponent paediatric malaria vaccine.
76
PRD-Malaria
Project Coordinator:
Jaap Goudsmit
Crucell Holland
Leiden, Netherlands
Tel: +31-0 71 52 48 755
E-mail: [email protected]
Partners:
Stefan Kaufmann
Max Planck Institute for Infection Biology
Berlin, Germany
Marita Troye-Blomberg
Stockholm University
Stockholm, Sweden
PRD-Malaria
77
SME-Malaria
Acronym: SME-Malaria
Project number: LSHP-CT-2006-018918
BACKGROUND
The most promising P. falciparum antigen-vector combinations will be used in monkey immunisation studies (these
animals can be efficiently infected with MeV) to determine
various aspects of potential safety, as well as humoral and
cellular immunity.
This project is investigating new malaria vaccine candidates with the objective of taking at least one through to
Good Laboratory Practices (GLP) pilot scale up. The most
promising candidates will undergo in vitro and in vivo testing, lead optimisation, and safety and toxicology testing
according to GLP standards.
The two selected antigens, MSP-1 and AMA-1, are leading vaccine candidates associated with the surface of
merozoite, the parasite form that invades uninfected erythrocytes. Both were originally identified as targets of antibody-based immunity directed against asexual blood stage
parasite multiplication. It has subsequently become clear
that both antigens are also expressed during the development of liver stage schizonts (the prelude to blood stage
development). They represent potential targets for cellular
immunity-based mechanisms of immunity and protection.
AIMS
The project aims to obtain at least one malaria vaccine
candidate stably expressed in MeV that induces humoral
and cellular immune responses.
78
PRD-Malaria
Project Coordinator:
Reinhard Glck
E-Mail: [email protected]
Partners
Hermann Bujard
University of Heidelberg
Heidelberg, Germany
David Arnot
Edinburgh University
Edinburgh, Scotland
UK
Yves Barbier
Texcell
Evry, France
Alan Thomas
Biomedical Primate Research Centre
Rijswijk, Netherlands
PRD-Malaria
79
MALINV
Acronym: MALINV
Differential Expression
of Malaria InvasionAssociated Proteins
in the Porozoite: Novel
Vaccination Strategy
BACKGROUND
In humans, sterile immunity against malaria was only
obtained after exposure to irradiated sporozoites
inoculated by mosquitoes. Recently, antigens by invasion
blood stage parasites have been shown to be expressed by
sporozoites, which leads to the hypothesis that they might
also be involved in sporozoite invasion of hepatocytes.
The three antigens investigated to date may not be
responsible for induction of optimal protective responses.
This would account for the difficulties encountered in
reproducing this sterile long-lasting immunity by current
sub-unit vaccines. It would clearly be desirable to
investigate other pre-erythrocytic antigens.
AIMS
The projects aims were:
a. to list all potential genes within the ebl and rh
families in P. falciparum, P.
b. berghei and P. yoelii. Real-time polymerase chain
reaction (RT-PCR) was performed for each gene on
ribonucleic acid (RNA) purified from all the stages
of these parasites, and the expression profile in
sporozoites determined;
c. to obtain recombinant proteins, peptides and
immunological reagents specific to each gene
product expressed in sporozoites;
d. to characterise the proteins expressed in the
sporozoite using reagents obtained in item 2, above;
e. to obtain transgenic parasite lines in which
sporozoite-expressed genes identified in item 1 are
individually disrupted or knocked out, and to purify
the corresponding sporozoites;
f. to employ the reagents from item 2, to determine the
functional role of each protein in sporozoite invasion;
g. to employ the sporozoites obtained in item 4, to
assess the functional role of each protein during the
lifecycle;
h. to employ the reagents from item 2 and item 4, to
conduct immunisation studies;
i. to conduct a detailed analysis of immune responses
80
PRD-Malaria
Project Coordinator:
Laurent Rnia
Hpital Cochin
Paris, France
Tel: +33-1 40 51 65 07
E-Mail: [email protected]
Partners:
Dominique Mazier
Hpital Piti-Salptrire
Paris, France
Robert Sauerwein
University Medical Centre
Nijmegen, Netherlands
Maria M Mota
Instituto Gulbenkian de Ciencia
Oeiras, Portugal
Alan Cowman
Walter and Eliza Hall Institute of Medical Research
Melbourne, Australia
PRD-Malaria
81
TB-VAC
Acronym: TB-VAC
An Integrated Project
for New Vaccines
against Tuberculosis
BACKGROUND
AIMS
The main aims of the project are:
the discovery and development of new tuberculosis vaccine
candidates effective in the young adult population;
the development of tests that predict vaccine
efficacy in humans;
the clinical evaluation of lead candidates in small
initial trials in Europe and Africa;
capacity building in developing countries for clinical
evaluation of vaccines;
liaising with other consortia to coordinate specific
activities;
liaising with the European and Developing
Countries Clinical Trials Partnership (EDCTP) to
enable further large clinical trials in Africa.
82
PRD-TB
PRD-TB
83
Project Coordinator:
Jelle Thole
Animal Sciences Group
Lelystad, Netherlands
Tel: +31-3 20 23 85 08
Email: [email protected]
E-Mail: [email protected]
S H E Kaufmann
MaxPlanck Institute for Infection Biology
Munich, Germany
E-Mail: [email protected]
F Poccia
National Institute for Infectious Diseases
Rome, Italy
Partners:
PRD-TB
H Wagner / R Lang
Technical University of Munich
Munich, Germany
Paul Lambert
University of Geneva
Geneva, Switzerland
84
S Stenger
University of Erlangen
Erlangen, Germany
F Mascart
Universit Libre de Bruxelles
Brussels, Belgium
F Dieli
University of Palermo
Palermo, Italy
P. Mettens
GSK Biologicals
Rixensart, Belgium
S Mboup
Hpital le Dantec
Dakar, Senegal
P Andersen
Statens Serum Institut
Copenhagen, Denmark
C Martin
Universidad de Zaragoza
Zaragoza, Spain
H. Engers
Armauer Hansen Research Institute
Addis Ababa, Ethiopia
F Sallusto
Institute for Research in Biomedicine
Bellinzona, Switzerland
C A Siegrist
University of Geneva
Geneva, Switzerland
G De Libero
University Hospital of Basel
Basel, Switzerland
J Westermann
University of Lbeck
Lbeck, Germany
F Spertini
Centre Hospital Universitaire Vaudois
Lausanne, Switzerland
B Gicquel / S Cole
Institut Pasteur
Paris, France
T Ottenhoff
Leiden University Medical Centre
Leiden, Netherlands
A Thomas / F Verreck
Biomedical Primate Research Center
Rijswijk, Netherlands
R Brookes
MRC Gambia
Banjul, Gambia
M Ota
University of Birmingham
Birmingham, England
UK
Oxford, England
UK
H Dockrell
London School of Hygiene and Tropical Medicine
London, England
UK
A Rawkins
Health Protection Agency Porton Down
Salisbury, England
UK
G. D Hussey
University of Cape Town
Cape Town, South Africa
S Stenger
Universitatsklinikum Ulm
Ulm, Germany
Y Perrie
Aston University
Birmingham, England
UK
E Bell
Manchester University Medical School
Manchester, England
UK
B D Robertson
Imperial College of Science
London, England
UK
K B Walker
National Institute for Biological Standards and Control
Potters Bar, Herts, England
UK
AVS Hill/ H McShane
University of Oxford
PRD-TB
85
Neotim
Acronym: Neotim
BACKGROUND
AIMS
The Neotim project aims to compare neonates/infants
and adults in terms of protective responses generated
during mycobacterial infections or vaccination with novel
mycobacterial antigens in murine experimental systems
and in humanised mice (reconstituted with human
lymphoid and myeloid cellular populations). The following
activities are being conducted:
comparing innate and adaptive immune responses,
with an emphasis on dendritic cells and T-cells;
investigating neonatal and adult mice during
mycobacterial infection and following inoculation
of a novel and promising candidate vaccine,
methylated HBHA;
investigating such responses using mice
reconstituted de novo with human lymphoid and
myeloid hemopoietic-derived cell lineages (allowing,
for the first time, an experimental dissection of
human immunity to mycobacteria); monitoring and
comparing the immune responses of naturally
infected humans in the corresponding age groups;
investigating the human molecular basis of hypersusceptibility to live BCG in rare neonates with
disseminated BCG disease in order to discover
novel mycobacterial susceptibility genes, which will
then be tested in the humanised mouse model.
86
PRD-TB
Project Coordinator:
Prof. Martin Rottenberg
Partners:
Carmen Fernandez
Stockholm University
Stockholm, Sweden
PRD-TB
87
Vaccines4TB
Acronym: Vaccines4TB
Project number: LSHP-CT-2004-012175
EC contribution: 1 053 445
Duration: 24 months
Type: STREP
Starting date: 1 January 2005
Project website: www.biocompetence.eu/index.php/
kb_1/io_3555/io.html
BACKGROUND
AIMS
The specific aims of the project were to:
evaluate the CD8 cytotoxic T cell (CTL) response
repertoire in the human population;
test if expression libraries representing the whole
M. tuberculosis genome can be used for CTL
antigen discovery;
test that the use of immuno-bioinformatics is a fast
and rational approach to CTL epitope identification.
88
PRD-TB
Project Coordinator:
Ole Lund
E-mail: [email protected]
Website: www.cbs.dtu.dk
Partners:
Sren Buus
Institute for Medical Microbiology and Immunology Panum
Copenhagen, Denmark
Tom H M Ottenhoff
Leiden University Medical Center
Leiden, Netherlands
Ugur Sahin
Pharmaceuticals AG, Germany
Mainz, Germany
PRD-TB
89
ImmunoVacTB
Acronym: ImmunoVacTB
A New Approach
for Developing a Less
Immunosuppressive
Vaccine for Tuberculosis
BACKGROUND
AIMS
The project aims to find a new strategy to overcome
these known problems of inefficacy, and will design less
immunosuppressive BCG strains, lacking PGL and/or
(parts of) the mannose cap. Immuno VacTB will isolate
BCG strains that lack the LAM mannose cap (or parts
thereof). This will be done in BCG with an intact as well
as with an interrupted pks15/1 gene. These recombinant
single or double mutant BCG strains will be evaluated
in vitro and in vivo for their ability to induce cytokine
production and to protect against tuberculosis in a murine
infection model.
The project will construct novel BCG strains that lack (part
of) the mannose cap of lipoarabinomanan and/or cannot
produce phenolic glycolipid. These strains will be an interesting platform from which to introduce M. tuberculosis
genes encoding important antigens, or non-mycobacterial
genes that enhance an immunoprotective response.
90
PRD-TB
Project Coordinator:
Bernard A M van der Zeijst
Netherlands Vaccine Insitute
Bilthoven, Netherlands
Tel: +31-3 02 74 41 64
E-mail: [email protected]
Partners:
B J Appelmelk
VU University Medical Center
Amsterdam, Netherlands
Rui Appelberg
Institute for Molecular and Cell Biology
Porto, Portugal
Germain Puzo
Institut de Pharmacologie et Biologie Structurale
Toulouse, France
PRD-TB
91
FLUVACC
Acronym: FLUVACC
Project number: LSHB-CT-2005-518281
Live Attenuated
Replication-Defective
Influenza Vaccine
BACKGROUND
AIMS
The aim of this project is to develop a novel vaccine
against influenza. This vaccine is a novel component of
European systemic efforts to prevent and control influenza,
based on a replication deficient virus that is generated by
a specialised technique known as reverse genetics. The
vaccine will be produced in cell culture.
Another important aim is to improve core technology for live
attenuated vaccine production, using a technique called
reverse genetics. The project has developed a master
strain that is lacking the NS1 gene which is essential
for productive viral replication. Candidate vaccines for
emerging influenza subtypes can be quickly produced by
inserting their genes into this master strain so that they
express the immunogenic surface proteins, but remain
replication-deficient. This master strain was adapted to
grow to high titers in tissue culture, making it possible to
produce large quantities in the case of a pandemic.
92
EE-FLU
Project Coordinator:
Joachim Seipelt
Partners:
Bert Klebl
GPC Biotech AG
Munich, Germany
Ale trancar
BIA Separations d.o.o.
Ljubljana, Slovenia
Martin lais
BioTest Ltd
Konrovice, Czech Republic
Oleg I Kiselev
Russian Academy of Medical Sciences
St Petersburg, Russia
Michael Bergmann
Vienna University Medical School
Vienna, Austria
Thorsten Wolff
Robert Koch Institute
Berlin, Germany
Maria Weidinger-Moser
Weikom & Network Communications Agency
Vienna, Austria
EE-FLU
93
FluVac
Dose Sparing
and Increased
Immunogenicity
for Vaccination
against Pandemic
Influenza with
CoVaccine HT
BACKGROUND
AIMS
This project is aiming at a novel influenza vaccine
formulation by combining CoVaccine HT and H5N1.
Feasibility studies with the adjuvant have indicated that
CoVaccine HT is a promising candidate for emergency
vaccines to establish high levels of immunity and to
compensate for the limited availability of antigen. The
ultimate goal is to prove the safety and efficacy of a
CoVaccine HT adjuvanted pandemic whole H5N1 virus
vaccine in humans, and to gain insight in its performance
in animal models.
Another objective is to exploit cell culture technology
for antigen production, as this method is more flexible,
independent of supply of animal-derived materials and
more consistent.
It will also target the delivery of a prototype emergency
vaccine to control or prevent a pandemic. In preclinical
and clinical studies, the project will test optimal doses
of inactivated, cell culture-derived whole influenza virus
(H5N1) and CoVaccine HT as adjuvant. The novel
adjuvant CoVaccine HT has been shown to elicit high
humoral and cellular responses against different types
of antigens (including the inactivated influenza virus) in
different animal species. It is considered to be a promising
candidate for a pandemic influenza vaccine.
94
Acronym: FluVac
EE-FLU
Project Coordinator:
Luuk A.Th. Hilgers
Nobilon International B.V.
Boxmeer, Netherlands
Tel: +31-0 48 55 87 276
Email: [email protected]
Project Manager:
Jacco G M Heldens
Nobilon International B.V.
Boxmeer, Netherlands
Partners:
James Glover
Protherics plc
The Heath Business and Technical Park
Runcorn, England
UK
Ingileif Jnsdttir
University of Iceland
Reykjavik, Iceland
John S Oxford
Retroscreen Virology Ltd
London, England
UK
Guus Rimmelzwaan
Erasmus Medical Center
Rotterdam, Netherlands
EE-FLU
95
PANFLUVAC
Acronym: PANFLUVAC
Efficacious Vaccine
Formulation System
for Prophylactic
Control of Influenza
Pandemics
BACKGROUND
AIMS
PANFLUVAC aims to produce an efficacious vaccine
formulation to meet immediate and future needs for
controlling influenza epidemics and pandemics. The
project is constructing vaccine delivery systems for
intranasal and parenteral vaccines. New H5N1 vaccines
are to be based on well-established virosome technology
- which has proven its worth for efficacious interpandemic
vaccines - as well as whole virus vaccines. This will permit
a comparison of the intranasal virosomal vaccine with the
whole virus vaccine.
96
EE-FLU
Project Coordinator:
Kenneth McCullough
E-mail: [email protected]
Partners:
Ronald Kompier
Crucell NV
Leiden, Netherlands
John Wood
National Institute for Biological Standards and Control
Hertfordshire, England
UK
John Oxford
Retroscreen Virology Ltd
London, England
UK
Lars R. Haaheim
University of Bergen
Bergen, Norway
Maria Zambon
Central Public Health Laboratory
London, England
UK
Isabella Donatelli
Istituto Superiore di Sanit
Rome, Italy
Carlos Guzman
Hemholtz Centre for Infection Research
Braunschweig, Germany
Vronique Gobry
SCIPROM
St Sulpice, Switzerland
EE-FLU
97
Intranasal H5vaccine
Acronym: Intranasal H5vaccine
Project number: P5B-CT-2007-044512
Protective Efficacy of
Intranasal del NS1
(H5N1) Influenza Vaccine
BACKGROUND
AIMS
The Intranasal H5vaccine project is developing a novel vaccine against avian influenza. It is assessing immunological
properties; conducting preclinical experiments and establishing novel, sensitive methods for the assessment of the
antibody response against viral proteins. These assays will
allow for the selection of an optimal vaccine candidate that
will be subsequently evaluated in clinical trials.
The project will also test the protective properties of the
vaccine against homologous and heterologous influenza
strains in ferret challenge experiments, using different H5
strains. In order to close the gap between the ferret model
and human studies, the project will use the macaque
model to evaluate the immune response. These preclinical
studies will allow the selection of the most potent vaccine
candidate, which will be produced according to cGMP
(current Good Manufacturing Practice) guidelines on Vero
cells. After a toxicological evaluation of the vaccine, clinical
phase I/II studies will be performed. A human challenge
study using the attenuated H5 virus will be carried out,
if permitted by the authorities. Alternatively, for proof of
principle of the del NS technology, an H1-del NS1 vaccine
will be produced and used in an H1 challenge study.
98
EE-FLU
Project Coordinator:
Joachim Seipelt
Partners:
Ivana urov
BioTest Ltd
Konrovice, Czech Republic
Volker Wacheck
Vienna University Medical School
Vienna, Austria
Jindrich Cinatl
Clinics of J W Goethe-University
Frankfurt, Germany
Nicolai Bovin
Shemyakin Institute of Bioorganic Chemistry
Moscow, Russia
John Oxford
Retroscreen Virology Ltd
London, England
UK
EE-FLU
99
CHIMERIC VACCINES
Acronym: CHIMERIC VACCINES
Project number: COOP-CT-2004-512864
BACKGROUND
AIMS
This project developed a novel approach for a vaccine.
The technology was based on the insertion of selected
epitopes into a genetically modified influenza virus that is
apathogenic. To achieve these results, the project identified
both a stable vector and promising antigens. Antigen
selection was based on bioinformatics methods that were
substantiated by experimental validation. The properties
of the backbone vector in terms of safety and stability
were assessed in preclinical experiments and gave highly
satisfactory results. To bring the proposed chimeric vaccine
into clinical trials, a production process in small scale was
established, using a novel purification technology.
100
EE-FLU
Project Coordinator:
Joachim Seipelt
Partners:
Emergentec Biodevelopment GmbH
Vienna, Austria
BioTest Ltd
Konrovice, Czech Republic
BIA Separations d.o.o.
Ljubljana, Slovenia
Clinics of J W Goethe-University
Frankfurt, Germany
Vienna University Medical School
Vienna, Austria
State Institution Research
Institute of Influenza
St Petersburg, Russia
EE-FLU
101
Universal Vaccine
Acronym: Universal Vaccine
BACKGROUND
AIMS
The project aimed to:
utilise the M2e peptide as antigenic determinant;
develop a powerful, new, safe and easilyadministered nasal vaccine for humans that
provides lifelong protection against influenza;
covalently fuse the M2e-peptide with the CTA1-DD
adjuvant, creating a strongly immunogenic influenza
vaccine suitable for mucosal delivery;
further improve vaccine efficacy by incorporating
the fusion protein in proprietary liposomes;
increase the in vivo maintenance of the antigen
by using blocked or constrained peptides or
peptidomimetics;
determine the in vivo mechanism of action of the
mucosal influenza vaccine;
demonstrate the safety and efficacy of the vaccine
in animal challenge models.
102
EE-FLU
Project Coordinator:
Anna Ramne
Biovitrum AB
Gteborg, Sweden
Tel: +46-0 31 74 91 118
E-Mail: [email protected]
Partners:
Hans Langedijk
Pepscan Systems BV
Lelystad, Netherlands
Roger New
Royal Veterinary College
London, England
UK
Doriano Cingolani
Eurogentec SA
Seraing, Belgium
Walter Fiers/Xavier Saelens
Flanders Interuniversity/Ghent University
Ghent, Belgium
Nils Lycke
Gteborg University (UGOT)
Gteborg, Sweden
EE-FLU
103
HEPACIVAC
Acronym: HEPACIVAC
New Preventative
and Therapeutic
Hepatitis C Vaccines:
From Preclinical
to Phase I
BACKGROUND
AIMS
104
EE-Hepatitis
Project Coordinator:
Riccardo Cortese
Krystina Bienkowska-Szewczyk
University of Gdansk
Gdask, Poland
E-mail: [email protected]
Scientific Coordinator:
Sayed F Abdelwahab
Sergio Abrignani
Novartis Vaccine & Diagnostics
Siena, Italy
Cristiana Tozzi
ALTA S.r.l.
Siena, Italy
E-mail: [email protected]
Adrian Hill
University of Oxford
Oxford, England
UK
Partners:
Alfredo Nicosia
Istituto di Ricerche di Biologia Molecolare P. Angeletti
Rome, Italy
Ferruccio Bonino
Jane McKeating
University of Birmingham
Birmingham, England
UK
Stefan Zeuzem
University of Saarland
Saarland, Germany
EE-Hepatitis
105
DISSECT
Acronym: DISSECT
Development
of Intervention
Strategies against
SARS in a EuropeanChinese Taskforce
BACKGROUND
Severe Acute Respiratory Syndrome (SARS) was
initially detected in Guangdong province, China. It has
been proven that the etiological agent of SARS is a new
coronavirus (CoV) with the acronym SARS-CoV.
The recent outbreak constitutes an important challenge to
the capabilities of EU Member States, and confirms that
new communicable disease outbreaks require a specific
focus. Actions should attempt to address longer term
research commitments, define appropriate sustainable
priorities and encourage collaboration with partners
from SARS-affected areas in third countries. There is
an urgent need to develop strategies for preventive
therapy for healthcare workers, who could eventually be
vaccinated, and then for other target groups who should
be vaccinated.
The emergence of SARS has created serious economic
and societal problems in China, reducing human and
commercial interaction. The project created a EuropeanChinese task force to combat SARS, and create essential
links between two parts of the world.
AIMS
This project encompasses all the complementary aspects
necessary for the development of intervention strategies,
including vaccination, immunotherapy, and antivirals
to protect against SARS. In addition, techniques and
materials will be generated to develop diagnostic kits that
will lead to the identification of infected individuals at very
early steps of the disease, and to differentiate between
vaccinated and naturally infected people. The first focus
of the project is vaccine development. Accordingly, a set
of complementary strategies is proposed to guarantee the
success. These strategies include classical vaccines
(such as whole inactivated virus), subunit vaccines,
and state of the art-recombinant technology derived
vaccines. The second focus of the project is on the state
of the art-therapeutic approaches, ranging from the use of
monoclonal antibodies to specific antivirals. For achieving
106
EE-SARS
Partners:
Qi Xie
Institute of Genetics and Developmental Biology
Beijing, China
Peter J. M. Rottier
Utrecht University
Utrecht, Netherlands
Albert DME Osterhaus
Erasmus University Rotterdam
Rotterdam, Netherlands
Anlong Xu
Sun Yat-sen University
Guangzhou, China
Zhihong Hu
Wuhan Institute of Virology
Wuhan, China
Pilar Perez Brea
Centro Nacional de Microbiologia
Madrid, Spain
Juan Plana-Durn
Fort Dodge Veterinaria SA
Girona, Spain
Project Coordinator:
Luis Enjuanes
EE-SARS
107
SARSVAC
Acronym: SARSVAC
Immunoprevention
and Immunotherapy
of SARS Infection
BACKGROUND
AIMS
The project aimed to produce an efficacious vaccine
formulation to meet immediate and future needs
for protection against SARS infections as well as
immunotherapeutic measures for SARS.
108
EE-SARS
Project Coordinator:
Mariagrazia Pizza
Partners:
Stephan Becker
Philipps-Universitat Marburg
Marburg, Germany
Antonio Lanzavecchia
Istituto di Ricerca in Biomedicina
Bellinzona, Switzerland
Peiying Ouyang Peiying
Fudan University
Shanghai, China
EE-SARS
109
SARS/FLU VACCINE
Acronym: SARS/FLU VACCINE
Project number: LSHB-CT-2004-512054
Development of a
Combined Influenza/
SARS Vaccine
BACKGROUND
In 2002, an atypical pneumonia, characterised by progressive respiratory failure, emerged in southern China. The
causative agent was rapidly identified as a new coronavirus designated as Severe Acute Respiratory Syndromeassociated virus SARS-CoV. The disease swept rapidly to
neighbouring regions and led to several international cases, including Canada. By the end of the epidemic, in July
2003, about 8 000 SARS cases and almost 800 deaths
due to SARS were recorded worldwide. Since then, the
world is in an inter-epidemic period, as no new cases have
been reported.
AIMS
The aim of the project was to test constructs for
immunogenicity. The ones that provoke the best immune
response without compromising safety in animal testing
will be selected for a full preclinical testing programme.
110
EE-SARS+FLU
Project Coordinator:
Thomas Muster
Partners:
Jindrich Cinatl
Clinics of J W Goethe-University
Frankfurt, Germany
Ale trancar
BIA Separations d.o.o.
Ljubljana, Slovenia
Bernd Mayer
Emergentec Biodevelopment GmbH
Vienna, Austria
Ivana urov
BioTest Ltd
Konrovice, Czech Republic
Joachim Seipelt
University of Vienna Medical School
Vienna, Austria
EE-SARS+FLU
111
NOVADUCK
Acronym: NOVADUCK
Novel AI DIVA
Recombinant Vaccines
for Duck
BACKGROUND
AIMS
112
EE-Avian FLU
Project Coordinator:
Dr Michel Bublot
Merial SAS
254 Rue Marcel Mrieux
69007 Lyon, France
Tel:+33 4 72 72 59 73
Email: [email protected]
www.novaduck.eu
Partners:
Dr Vronique Jestin
Agence Franaise de Scurit Sanitaire
des Aliments (AFSSA)
Ploufragan, France
Dr Thierry van den Berg
Veterinary and Agrochemical Research Centre (VAR)
Brussels, Belgium
Dr Csaba Dren
Veterinary Medical and
Research Institute of the HAS (VMRI)
Budapest, Hungary
Dr Vilmos Palfi
Central Veterinary Institute
Budapest, Hungary
Dr Jill Banks
Veterinary Laboratory Agency
Kent, England, UK
Fabienne Mathieu
Biosource Europe
Nivelles, Belgium
Dr Charlotte Dalba
Epixis S A
Paris, France
EE-Avian FLU
113
Vaccine, Diagnostic
Test Development and
Immunology Aspects
of Avian nfluenza
BACKGROUND
AIMS
The primary aim of this project is to develop better
avian influenza vaccines through live or vector vaccines
that could be mass applicable through spray, drinking
water or eye drops. These vector vaccines would offer
considerable advantages - mass applicable, less labour
intensive and animal friendly application, protection by
local and systemic immunity and less interference with
eventual maternal antibodies, more complete protection
through cellular and humoral immunity, faster onset of
immunity when used in face of an outbreak and cheaper
production methods.
114
EE-Avian FLU
Project Coordinator:
Dr Danny Goovaerts
Intervet International bv
Wim de Krverstraat 35
P.O. Box 31
Boxmeer, Netherlands
Tel: +31-4 85 58 77 27
Email: [email protected]
Partners:
Dr Thomas Mettenleiter
Friedrich-Loeffler-Institut
Federal Research Institute for Animal Health
Greifswald-Insel Riems, Germany
Dr Christian Schelp
Bommeli Diagnostics
Leibefeld-Bern, Switzerland
EE-Avian FLU
115
SCOOTt
Acronym: SCOOTT
Sustainable Control
of Onchocerciasis
Today and Tomorrow
BACKGROUND
AIMS
The aims of the project are to improve sustainable control
of onchocerciasis through:
refinement of existing chemotherapeutic regimes
by use of doxycycline to complement ivermectin
treatment and further screening of existing drugs;
assessment of immunological sequelae of
ivermectin intervention and their implications for
improved control strategies;
identification of new targets, including vaccine
candidates, and approaches for integrated control.
116
NID-Helminth
Project Coordinator:
Prof. David W Taylor
Partners:
NID-Helminth
117
TRANCHI
Acronym: TRANCHI
T Cell Regulation
and the Control of
Helminth Infections
BACKGROUND
AIMS
The project will establish whether Regulatory T cells,
lymphocytes with immunosuppressive properties, are
active in chronic helminth infections, and if so, whether
targeting such cells may offer an immunological cure to the
major tropical diseases of filariasis and schistosomiasis. It
will achieve these by carrying out the following:
profiling the type and functions of Tregs in filariasis
and schistosomiasis infected humans;
comparing Treg activity in patient groups of differing
infection status/levels of pathology;
establishing if polymorphisms for regulatory genes
are linked to Treg profiles in humans;
demonstrating the role of Tregs in helminthassociated hyporesponsiveness;
testing whether neutralisation of Tregs restores
immune responsiveness in human cells;
testing whether neutralisation of Tregs restores
immunity to infection in animal models;
characterising human Treg gene expression and T
cell receptor usage;
assessing community and health system issues for
new immunological interventions.
118
NID-Helminth
Project Coordinator:
Prof. Rick Maizels
Partners:
NID-Helminth
119
BOVAC
Development of a
Prophylactic Vaccine
and Diagnostic Markers
to Diagnose and Prevent
Lyme Borreliosis
Specific to Europe and
North America
BACKGROUND
Lyme disease or borreliosis (LB) is the commonest tickborne disease in Europe. It can be transmitted to humans
when they are bitten by ticks carrying species of the
Borrelia bacterium. Infection is often diagnosed by a rash
(Erythema migrans) that spreads out from the site of the
tick bite. If caught early, the disease can be prevented with
oral antibiotics. However, if left untreated, the bacteria can
spread through the bloodstream, access various tissues
and cause severe diseases including meningitis, arthritis
and carditis. Official incidence rates in Europe range
from 0.3 to 150 cases per year per 100 000 population.
However, few countries have made LB a notifiable
disease, so these rates are only an approximation. Many
experts believe that actual rates of LB infection could be
up to seven times higher.
AIMS
The aim of the project was to select novel vaccine
candidates suitable to develop a vaccine to prevent
LB. The consortium was also interested in using the
identified antigens to evaluate their use in developing
novel diagnostic tools for LB as well as assessing the
importance of LB in Austria and the Czech Republic by a
thorough epidemiology study.
120
Acronym: BOVAC
NID-Lyme disease
Project Coordinator:
Dr Andreas Meinke
Intercell AG
Campus Vienna Biocenter 5
Vienna, Austria
Tel: +43-1 20 62 02 10
www.intercell.com
Email: [email protected]
Partners:
Dr Martin lais
BioTest S.R.O.
Konrovice, Czech Republic
MWG Biotech AG
Ebersberg, Germany
Prof. Bohumir Kri
National Institute of Public Health
Prague, Czech Republic
Prof. Sven Bergstrom
Ume University
Ume, Sweden
NID-Lyme disease
121
SAVINMUCOPATH
Acronym: SAVINMUCOPATH
Novel Therapeutic
and Prophylactic
Strategies to Control
Mucosal Infections
by South American
Bacterial Strains
BACKGROUND
AIMS
The main aims of the project are to gain understanding
of the host-pathogen interaction and to develop novel
mucosa-specific therapeutics and vaccines to control
bacterial infections. Innate defences are up-regulated
at mucosal sites upon detection of conserved microbial
molecules and contribute both to the immediate barrier
function to mucosal colonisation and the long lived
antigen-specific mucosal immune responses. These
molecules thus have early immunostimulatory activities
and adjuvant properties on mucosa. The projects strategy
is to take advantage of these natural mechanisms to
improve clearance and immunoprophylaxis against the
selected life-threatening pathogens.
Bacterial strains are being studied in experimental animal
mucosal infection models in order to (i) characterise the
innate mechanisms of early elimination of pathogens and
the concomitant mechanisms of induction of mucosal
adaptive immunity, (ii) define the proof of concept that
conserved microbial molecules can activate mucosal
immunity, and (iii) identify novel mucosa-specific pathogen
molecules with biological activities on mucosal innate and
adaptive immunity using purified bacterial components
and screening on cell and animal models.
122
NID-Respiratory
Project Coordinator:
Dr Jean-Claude Sirard
INSERM U801
Institut Pasteur de Lille - Institut de Biologie
Equipe dImmunit Anti-Microbienne des Muqueuses
1 Rue du Pr Calmette
Lille, France
Tel: +33-3 20 87 10 76
E-mail : [email protected]
Partners:
Martin Rumbo
Laboratorio de Investigaciones
en el Sistema Inmune (LISIN)
La Plata, Argentina
Daniela Hozbor
Instituto de Bioqumica
y Biologa Molecular (IBBM)
La Plata, Argentina
Tracy Hussell
Kennedy Institute of Rheumatology - Imperial College
London, England
UK
Wolf-Dietrich Hardt
ETHZ, Institute of Microbiology
Zurich, Switzerland
Jos Alejandro Chabalgoity
Instituto de Higiene
Montevideo, Uruguay
Alexis Kalergis
Pontificia Universidad Catolica de Chile
Santiago, Chile
Patricia Lopez Biscayart / Augusto Pich Otero
BIOL Instituto Biolgico Argentino
Buenos Aires, Argentina
NID-Respiratory
123
OMVac
Novel Prevention
and Treatment
Possibilities for
Otitis Media through
the Comprehensive
Identification of
Antigenic Proteins
BACKGROUND
AIMS
The main objective of this project is the identification of
novel vaccine candidates from NTHi and M. catarrhalis
to develop a prophylactic vaccine against OM. Bacterial
surface display libraries will be constructed and screened
with human sera from exposed individuals to define the
ANTIGENome of both pathogens. Following thorough
in vitro validation selected antigens will be evaluated in
experimental animal models of OM.
The project is also addressing the comprehensive
characterisation of natural immune responses against
proteineacous antigens of the major three bacterial
pathogens causing OM. Moreover, the function of
protective antigens during pathogenesis and biofilm
formation will be investigated.
124
Acronym: OMVac
NID-Otitis media
Project Coordinator:
Andreas Meinke
Intercell AG
Campus Vienna, Biocenter 6
Vienna, Austria
Tel: +43-1 20 62 02 10
Email: [email protected]
Partners:
Andreas J. Kungl
University of Graz
Graz, Austria
Wolfgang Zimmermann
AGOWA GmbH
Berlin, Germany
va Bn
Semmelweis University
Budapest, Hungary
John Hays
Erasmus MC
Rotterdam, Netherlands
Peter W.M. Hermans
Radboud University Nijmegen Medical Centre
Nijmegen, Netherlands
Birgitta Henriques-Normark
Karolinska Institutet
Solna, Sweden
NID-Otitis media
125
HEVAR
Acronym: HEVAR
Herpesvirus-based
Vaccines against
Rotavirus Infections
BACKGROUND
AIMS
The projects aim is to contribute to a better understanding
of the immune biology of rotavirus infections using a novel
generation of gene transfer vectors, as a first step towards
the development of innovative genetic vaccines to fight
against these pathogens. To this end, HEVAR will develop
herpes simplex virus type 1 (HSV-1)-based vectors for the
generation and analysis of rotavirus-specific expression
and display model vaccines. This approach is based
on the possibility of engineering HSV-1-based vectors
expressing and/or displaying rotavirus antigens, either
individually or in combination, alone or together with
immune-modulator genes.
126
NID-Diarrhoea
Project Coordinator:
Dr Alberto Epstein
Partners:
Ludwig-Maximilians-Universitaet-Muenchen
Muenchen, Germany
NID-Diarrhoea
127
SUPASALVAC
Salmonella-Free Broilers
by Live Vaccine-Induced
Innate Resistance
to Colonisation
and Invasion and Novel
Methods to Eliminate
Vaccine and Field Strains
BACKGROUND
The European poultry industry urgently needs a costeffective way of dealing with salmonella in broilers (young
chickens) in order to produce quality-assured, pathogenfree meat at a competitive price. The immune system
of broilers is insufficiently developed when they are
slaughtered (before six weeks) and, as a consequence,
they frequently succumb to infection by pathogenic
salmonella. Strategies are required for the biological
control of the food borne infection in broilers, including
further investigation of promising designer vaccines for
newly hatched chicks.
AIMS
This project aims to use biotechnology to enhance the
effects of live salmonella vaccines. The activities include
the following:
testing strains of salmonella to see which are best
at inhibiting colonisation by similar strains;
discovering which genes are involved in inhibition;
identifying the genes that draw chick immune cells
to the gut in response to the live vaccine;
identifying the role of intestinal defensin peptides in
controlling colonisation.
The project will also investigate other methods of preventing
colonisation of chick intestines by salmonella, exploring
dietary additives or methods of inhibiting bacterial genes
active during colonisation. Furthermore, bacteria-killing
viruses (known as bacteriophages) will be used to target
particular bacteria. The project is developing phages to
destroy salmonella, both in broilers and in carcasses.
128
Acronym: SUPASALVAC
NID-Diarrhoea
Project Coordinator:
Dr Paul Barrow
Institute for Animal Health
Compton Laboratory
Newbury
Berkshire RG20 7NN
England, UK
Tel: +44-1 63 55 78 411
Dr V Allen
University of Bristol
Bristol, England
UK
Dr M Schellekens
EBI BV
Den Haag, Netherlands
Email: [email protected]
Partners:
Prof. R Ducatelle
Universiteit Gent
Gent, Belgium
Dr U Methner
Federal Research Center for Virus Diseases of Animals
Tbingen, Germany
Dr I Rychlik
Veterinary Research Institute
Brno, Czech Republic
Prof. B Nagy
Veterinary Medical Research Institute
Budapest, Hungary
Dr I Schroder
Lohmann Animal Health
Cuxhaven, Germany
Dr A C Lalmanach
Institut National de la Recherche Agronomique
Paris, France
Dr J Wagenaar
Institut voor Dierhouderij en Diergezondheid
Lelystad, Netherlands
NID-Diarrhoea
129
TRYPADVAC2
Acronym: TRYPADVAC2
Development of an
Anti-Disease Vaccine
and Diagnostic Tests for
African Trypanosomosis
BACKGROUND
Trypanosomosis, or African sleeping sickness, is a disease
that affects both humans and livestock. Today nearly a third
of Africa is affected by Trypanosomosis which is caused by a
parasite transmitted by the bite of the tsetse fly. The disease
is having a detrimental effect on the development of African
rural communities as it restricts the keeping of farm animals
which affects supplies of meat and milk and also limits the
development of mixed arable and livestock farming.
Previous studies of trypanosome infections have focused
on congopain, an immunosuppressive cysteine protease
(CP) of Trypanosoma congolense. As the effects of
immunisation with congopain are limited, association with
other antigens is required.
Recent developments in the field of proteomics and
progress in the genome mapping of trypanosomes have
provided tools for the study of new pathogenic pathways.
In order to improve diagnosis of the disease, procedures
for antibody detection will be developed and/or validated.
They are based on recombinant technology, which
circumvent problems associated with the current use of
parasite extracts. Recombinant and synthetic peptides
from CPs and heat shock proteins, both previously
identified as major antigens, as well as newly described
molecules, will be assessed for their diagnostic potential.
Techniques for detection of parasite antigens in the host
tissues will be re-examined using recently developed
monoclonal antibodies.
AIMS
The aim of the project is to contribute to the eradication
of Trypanosomosis in humans and livestock through
limitation of trypanosome-associated pathology and
accurate diagnostics of trypanosome infections. It is also
proposed to develop immunisation strategies against
pathogenic factors of trypanosomes.
The current proposals will include screening and
characterisation of other pathogenic molecules,
130
NID-Trypanosome
Project Coordinator:
Dr Alain Boulange
Partners:
Prof. J Mottram
Wellcome Centre for Molecular Parasitology
Glasgow, Scotland
UK
Prof. G Lubega
University of Makerere
Kampala, Uganda
Dr L Neves
Dr P Jacquier
Diamed AG
Cressier-sur-Morat, Switzerland
Prof. V Rosario
Institute of Hygiene and Tropical Medicine
Lisbon, Portugal
Prof. M Gonzatti
Prof. T Coetzer
University of Kwazulu-Natal
Westville, South Africa
Prof. S Magez
Flanders University Institute of Biology
Ghent, Belgium
Prof. T Baltz
Centre National de la Recherche Scientifique
Paris, France
Dr S Thevenon
Centre International de Recherche
Developpement sur lElevage en Zone Subhumide
Bobo Dioulasso, Burkina Faso
NID-Trypanosome
131
CANCERIMMUNOTHERAPY
Acronym: CANCERIMMUNOTHERAPY
Project number: LSHC-CT-2006-518234
Cancer Immunology
and Immunotherapy
BACKGROUND
Cancer is a major life-threatening disease and the second
greatest cause of mortality in Europe after cardiovascular
diseases. Classical cancer treatment still relies on surgery,
chemotherapy and radiotherapy. Despite clear progress in
some cancer types, cancer therapy in general often fails
to prevent progression to metastatic disease. In addition,
these approaches are very toxic in themselves, imposing
a heavy burden of side effects on the patient. There is
clearly a need for new therapeutic approaches that would
be more efficient and less toxic.
AIMS
The first part of the project consists of vaccination clinical
trials for the comparison of various vaccines, such as
peptides and RNA, with different types of immunological
adjuvants and dendritic cells. Safety and clinical efficacy
are the primary endpoints of these trials. A huge effort is
being made to monitor the anti-vaccine T cell responses,
as examining the correlation between immunological
and clinical responses to the vaccines is crucial to
understanding which factor(s) limit tumour regression. A
second part of the project, tightly connected to the clinical
trials since it uses biological material from the vaccinated
patients, consists of optimising tumour vaccines and
combating immune evasion.
Prospective mechanisms of tumour escape will be analysed and correlated with the clinical results. Improved
modalities of vaccination will be tested and new target
antigens will be identified. All these results will help the
project partners to design improved vaccines. Finally, considering the complexity of mechanisms that may lead to
or prevent tumour regression in vaccinated patients, the
project proposes exploring more fundamental aspects of
the anti-tumour immune response. This includes the crosspresentation of tumour antigens by dendritic cells, recruitment of cells of the innate immune system, involvement of
suppressor T cells, and development of murine models of
inducible tumours. If new concepts emerge from this work,
they will also help in the design of better vaccines.
132
Non-inf-cancer
Project Coordinator:
Thierry Boon
de Duve Institute
Brussels, Belgium
E-mail: [email protected]
Partners:
Hans-Georg Rammensee
Eberhard-Karls-Universitt Tbingen
Tbingen, Germany
Sebastian Amigorena
Institut Curie
Paris, France
Marc Bonneville
Institut Nationale de la Sant
et de la Recherche Mdicale
Paris, France
Pedro Romero
Ludwig Institute for Cancer Research
Epalinges, Switzerland
Cornelis Melief
Leiden University Medical Center
Leiden, Netherlands
Chiara Castelli
Fondazione Istituto Nazionale Tumori
Milan, Italy
Gerold Schuler
Friedrich Alexander Universitt Erlangen-Nrnberg
Erlangen, Germany
Thomas Wlfel
Johannes Gutenberg-Universitt Mainz
Mainz, Germany
Eric Tartour
Universit Ren Descartes
Paris, France
Vincenzo Cerundolo
The Chancellor, Masters and Scholars
of the University of Oxford
Oxford, England
UK
Muriel Moser
Universit Libre de Bruxelles
Brussels, Belgium
Alexander Eggermont
Erasmus MC Rotterdam
Rotterdam, Netherlands
Kris Thielemans
Vrije Universiteit Brussel
Brussels, Belgium
Carl Figdor
Stichting Katholieke Universiteit
Nijmegen, Netherlands
Hlne Sicard
Innate Pharma SA
Marseille, France
Federico Garrido
Fundacion Virgen de las Nieves
Granada, Spain
Vincenzo Russo
Fondazione Centro San Raffaele del Monte Tabor
Milan, Italy
Gnter Hmmerling
Deutsches Krebsforschungszentrum
Heidelberg, Germany
Brigitte Dreno
Centre Hspitalier Universitaire de Nantes
Nantes, France
Ulrich Keilholz
Charite Universitatsmedizin Berlin
Berlin, Germany
Jochen Probst
CureVac - the RNA people
Tbingen, Germany
Non-inf-cancer
133
DENDRITOPHAGES
Acronym: DENDRITOPHAGES
Project number: LSHP-CT-2003-503583
Therapeutic Cancer
Vaccines
BACKGROUND
AIMS
The project aimed to demonstrate the immune and clinical
efficacy, reproducibility and feasibility of anticancer cell
vaccine by sequential steps: (i) the best dendritic cell (DC)
vaccination strategy via preclinical studies was selected;
(ii) the immune response was monitored in correlation to
the clinical response after identifying the most relevant
immuno-monitoring techniques, demonstrating the
immunological efficacy of DC immunotherapy in prostate
cancer, which was performed after loading ex vivo DCs
with proteic antigen; (iii) a clinical trial was carried out
to evaluate the cell drug on patients with progressing
prostate cancer. The patients blood monocytes were
transformed into effector monocyte-derived DCs, which
fight the disease. The therapeutic cell drug comprised
DCs loaded with cancer-specific antigens, activating the
patients immune system after re-injection.
134
Non-inf-cancer
Project Coordinator:
Jacques Bartholeyns
Partners:
Andreas Mackensen
University of Regensburg
Regensburg, Germany
Miles Prince
Centre for Blood Cell Therapies
Victoria, Australia
Reinhard Glueck
Universit di Catania
Catania, Italy
Thomas Felzmann
Childrens Cancer Research Institute
Vienna, Austria
Filippo Belardelli
Istituto Superiore di Sanit
Rome, Italy
Non-inf-cancer
135
LCVAC
Acronym: LCVAC
New Vaccination
Therapies for
Lung Cancer
BACKGROUND
AIMS
LCVACs aims were:
to identify and characterise novel lung cancer
specific antigens using proteome analysis;
to develop an effective and safe vaccination
protocol for antigens and tumor cells;
to characterise and generate human lung cancer
cell lines to be used for an allogeneic cellular
cancer vaccine.
136
Non-inf-cancer
Project Coordinator:
Ren Vleugels
Partners:
Onyvax Ltd
St. Georges Hospital Medical School
London,
UK
Medical Proteome Center
Ruhr-Universitt Bochum
Bochum Germany
Universiteit Maastricht
Department of Molecular Cell Biology
The Netherlands
Non-inf-cancer
137
VITAL
Development of
Optimised Recombinant
Idiotypic Vaccines
for Subset-Specific
Immunotherapy of
B Cell Lymphomas
Acronym: VITAL
Project number: LSHC-CT-2006-037874
EC contribution: 2 050 000
Duration: 36 months
Type: STREP
Starting date: 1 January 2007
Project website: www.cro.sanita.fvg.it/progetti/
vital/index.htm
BACKGROUND
AIMS
A database is being built of Id sequences expressed by
various B-NHL so as to identify subgroups of tumours
expressing molecularly correlated Id proteins. VITAL will
also do the following:
characterise the selected Id proteins for their
immunogenicity and for the ability to induce crossreactive immune responses against related Id
proteins;
identify B and T cell epitopes;
develop dedicated assays for immunomonitoring;
produce optimised versions of selected Id vaccines
(by using new strategies and validated in animal
models);
assess and validate new adjuvants and delivery
systems for improved Id vaccine formulations and
administration;
generate and filter, based on GMP standards,
the most promising Id proteins, which will then be
included in new vaccine formulations for crossreactive immunotherapy trials;
perform preclinical characterisation of the
immunogenicity of selected natural Id proteins that
can induce immune responses against lymphoma
cells expressing molecularly correlated Id proteins.
138
Non-inf-cancer
Project Coordinator:
Riccardo Dolcetti
Partners:
Bjarne Bogen
University of Oslo
and Rikshospitalet University Hospital
Oslo, Norway
Maria Masucci
Karolinska Institutet
Stockholm, Sweden
Antonio Rosato
University of Padova
Padova, Italy
Non-inf-cancer
139
MimoVax
Acronym: MimoVax
BACKGROUND
There are 12 million cases of Alzheimers disease
worldwide and 3.5 million people in Europe are afflicted
with it. It is believed that these figures will double in
the next 20 years as the European population ages.
Alzheimers mostly affects people over 65 years of age
and it is the most common form of dementia. There is
currently no cure for the progressive neuro degeneration
it causes. More research is urgently needed into its
cause both for social and economic reasons. This project
is focusing on the use of immune reactions to fight
Alzheimers, which is caused by deposits of amyloidbeta () peptides which form into clumps (known as
plaques). They develop when parts of a human protein
detach from the cell membrane of nerve cells and stick
together. The project is focusing on developing a vaccine
that breaks down these cell clumps.
AIMS
The aim of MimoVax is to find a vaccine that will cure
Alzheimers. To do this the following objectives will
be carried out:
novel monoclonal antibodies directed against neoepitopes on N-terminally truncated derivatives of
A will be generated and their specificities will be
evaluated;
mimotope peptides will be identified by screening
peptide libraries followed by testing their suitability
by assessing their in vivo immunogenicity in mice;
mimotope-based vaccines will be formulated
and evaluated for efficacy in animal models of
Alzheimers;
in vivo imaging systems will be set up to analyse
anti-A antibody distribution and turnover in mice
as well as effects of the novel Alzheimers vaccine
on plaque deposition in living transgenic animals;
SOPs for manufacturing GMP material will be
determined;
vaccine conjugate formulation for subsequent
analysis of toxicity and for the phase I clinical trial
140
Non-Inf-Alzheimer
Project Coordinator:
Dr Frank Mattner
AFFiRiS GmbH
Campus Vienna Biocenter 2
Viehmarktgasse 2A
Vienna, Austria
Tel: +43-1 79 81 57 51 5
E-mail: [email protected]
Partners:
Manfred Windisch
JSW
Graz, Austria
Antn Alvarez
EuroEspes
Coruna, Spain
Fritz Andreae
piCHEM
Graz, Austria
Richard Dodel
Philipps University Marburg
Marburg, Germany
Alexander Drzezga
Technical University Munich
Munich, Germany
Iris Grnert
Biolution
Vienna, Austria
Non-Inf-Alzheimer
141
Clinical Research
and capacity Building
Introduction
144
1.
2.
3.
4.
5.
6.
7.
8.
9.
EDCTP
Acronym: EDCTP
European and
Developing Countries
Clinical Trials
Partnership
BACKGROUND
Disease prevention in Africa is a top priority, not only in
order to help prevent millions of unnecessary deaths,
but also to begin to free the continent from its perpetual
disease-poverty-disease cycle, so it can begin to grow
economically. The principal diseases devastating Africa
are HIV/AIDS (human immunodeficiency virus/acquired
immune deficiency syndrome), malaria and tuberculosis
(TB), which are currently killing about 5 million people
every year. About 2 million of these die from HIV/AIDS,
for which there is as yet no cure. AIDS also forms a
deadly partnership with TB, which is the leading cause
of mortality in people who are HIV positive. Malaria kills
about 1 million people in Africa every year and 500 million
are infected annually.
These diseases put a huge economic burden on African
families. People on very low wages incur huge debts
and use up to a quarter of their annual income to pay for
treatment for sick relations. If they are unable to save,
they can never escape the poverty-disease trap. The high
economic toll of the main diseases is also contributing
to a decline in per capita income in many sub-Saharan
African countries. When large numbers of the population
are unable to continue working because of severe illness,
harvests are not gathered, factories are less productive
and children are put to work to replace sick adults, again
perpetuating the poverty cycle.
Reducing the disease burden in Africa will take years of
dedicated hard work and cooperation between European
and African scientists and medical specialists. Proper
organisation is also of key importance. Malaria and TB are
both preventable and curable, but need long-term courses
of treatment that must be followed strictly. However, this
procedure is not always implemented and many patients do
not complete their designated treatments. For there to be a
serious reduction in curable diseases such as malaria and
TB, health centres must be established with fully trained
staff who keep registers of patients receiving treatment.
If the disease burden is to be lightened in Africa, longterm and coordinated programmes of action must be
146
AIMS
The Commission has funded research on HIV/AIDS, TB
and malaria for many years. Over EUR 100 million was
spent on research under the Fifth Framework Programme,
for example, but now is the time for a more coordinated
and integrated approach. Increasing the health of the
populations of AIDS-stricken countries, thereby enabling
them to break out of the poverty-disease cycle, and
consequently creating more robust economic and social
conditions are the overall aims of the project. These will
be developed through networking and coordination of both
European and African programmes and activities carried
out in developing countries.
This is truly a joint programme, with European and African
experts and institutes working together. African scientists
and doctors are on the programmes steering committee
and will take a vital part in the process, from the opening
phases until the clinical trials stage. The drugs to be tested
in the clinical trials will be selected taking into account the
needs and the financial means of the recipients in terms of
ease of use and affordability, and trials will be carried out
in full accordance with ethical best practice.
Pharmaceutical companies provide over 60% of drugs
used in developing countries, and many drug and vaccine
candidates originate from private laboratories. However,
a longstanding problem involving health programmes
in developing countries is that it is not feasible for
pharmaceutical companies to commit themselves fully to
Coordinator:
Prof. Charles Mgone
147
EUROPRISE
Acronym: EUROPRISE
Project number: LSHP-CT-2006-037611
BACKGROUND
The successful development of preventative strategies
against HIV-1 infection (microbicides, vaccines or their
combined effects) would provide a pivotal turning point
in global efforts to combat the pandemic spread of
AIDS, with an impact of incalculable value on societal
problems associated with this disease. The principal
aim of this project is to bring together EU scientists
from the microbicide and vaccine fields to embrace
a coordinated approach to HIV-1 infection prevention
research.
Vaccines delivering non-replicating antigens mostly
fail to induce sufficient mucosal responses and
immunological memory to provide protection against
high viral challenge. In contrast, while it may be
technically easier to develop microbicides that prevent
transmission when applied before intercourse, their
duration of protection is likely to be short-lived and
their efficacy will be critically dependent upon user
compliance. To date, both fields have been slow to work
together in the development of products that provide
multiple levels of protection.
This network is focusing on the premise that
microbicides and vaccines targeting multiple stages
of mucosal transmission will have the best chance
of success. Indeed, there are many reasons why the
two fields should collaborate in developing effective
strategies to prevent mucosal vaginal or rectal
transmission, including the facts that approximately 80
% of new HIV infections are now heterosexual, and the
main entry of infection is across the vaginal or rectal
mucosa. Topically applied products are likely to have
a high level of acceptance, and importantly, their use
would be female-initiated. This is an important factor
for women who have no means to protect themselves
if their partners do not use condoms. Also, effective
microbicides are likely to become available before
effective vaccine candidates. Thus it is likely that in the
near future all vaccine efficacy trials will be carried out
in an environment where there is widespread use of
148
AIMS
The project has the following aims:
standardisation and harmonisation of research
tools;
identification of new anti-HIV infection/AIDS vaccine
and microbicide candidates and combinations to
prevent HIV infection/AIDS;
establishment of a clinical development pathway
for vaccines and microbicides within a European
framework;
provision of scientific training in microbicide and
vaccine development;
facilitating access to information relevant to HIV-1
microbicides and vaccines;
provision of a single focus for European HIV-1
microbicide and vaccine research.
Project Coordinator:
Robin Shattock
Scientific Coordinator:
Hans Wigzell
Karolinska Institutet
Stockholm, Sweden
E-mail: [email protected]
Rino Rappuoli
Novartis Vaccines and Diagnostics srl
Siena, Italy
E-mail: [email protected]
149
Partners:
M Cranage; D Lewis
St Georges Hospital University of London
London, England
UK
H Wigzell; Dr B Wahren; Dr F Chiodi; A L Spetz
Karolinska Institutet
Stockholm, Sweden
D Medaglini; G Pozzi
Universita di Siena
Siena, Italy
R Weiss
University College London
London, England
UK
F Gotch; S Patterson
Imperial College of Science, Technology & Medicine
London, England
UK
A Tagliabue
ALTA
Siena, Italy
V Jespers
Prince Leopold Institute for Tropical Medicine
Antwerp, Belgium
G Voss
GlaxoSmith Kline
Rixensart, Belgium
H Katinger; G Steiglere
Polymun
Vienna, Austria
150
K Uberla
Ruhr Universitat
Bochum, Germany
R Le Grand
Commissariat a lEnergie Atomique
Paris, France
G Scalae
Universita di Napoli
Naples, Italy
B Verrier
Institut National de la Sante et de la Medecine
Paris, France
C Lacey
University of York
York, England
UK
Q Sattentau
University of Oxford
Oxford, England
UK
P La Colla
Universit di Cagliari
Cagliari, Italy
Christiane Stahl-Hennig
Deutsches Primatenzentrum GmbH
Goettingen, Germany
Feny
Lunds Universitet
Lund, Sweden
F Tangy
Institut Pasteur
Paris, France
D Zipeto
Universit di Verona
Verona, Italy
S Norley
Robert Koch-Institut
Berlin, Germany
E Karamov
Russian Academy of Medical Science
Moscow, Russia
K Nihlmark
Mabtech AB
Stockholm, Sweden
N Dedes
European AIDS Treatment Group e.V.
Dusseldorf, Germany
C Moog
Universite Louis Pasteur
Strasbourg, France
H Schuitemaker
Sanquin
Amsterdam, Netherlands
J Alcami; R Najera
Instituto de Salus Carlos III
Majadahonda, Spain
G Leroux-Roels
Ghent University
Ghent, Belgium
151
DC-THERA
Acronym: DC-THERA
Project number: LSHB-CT-2004-512074
BACKGROUND
Dendritic cells (DC) form part of the immune system and
are found in most body tissues, particularly in the skin and
lining of the gastro-intestinal tract. Their job is to process
and ingest antigen material including foreign bacteria that
invades the body. They control many types of immune
response and consequently there is growing interest in
using DC in HIV/AIDS and cancer research and many other
diseases. Dendritic cell immunobiology has enormous
potential for the development of new immunotherapies for
cancer and infectious disease and this project focuses on
these potentials.
AIMS
The projects aim is to facilitate the translation of genomic,
proteomic and bioinformatic information with knowledge
from molecular cell biology and pre-clinical models towards
clinical trials of DC-based therapies for cancer and HIV. It
will do this by an ambitious joint programme of activities, with
the intention of restructuring the field of immunotherapy.
Project Coordinator:
Jonathan Austyn
University of Oxford
Oxford, England
UK
E-mail: [email protected]
Partners:
Gordon G MacPherson
University of Oxford
Oxford, England
UK
152
Vincenzo Cerundolo
University of Oxford
Oxford, England, UK
Carl G Figdor/Gosse Adema
Radboud University Medical Centre
Nijmegen, Netherlands
Muriel Moser
Universite Libre de Bruxelles
Brussels, Belgium
Anne OGarra
Medical Research Council
London, England
UK
Francesca Granucci
University of Milano-Bicocca
Milan, Italy
Gerold Schuler/Alexander Steinkasserer
Friedrich-Alexander-Universitt Erlangen-Nrnberg
Erlangen, Germany
Robert Coffin
Biovex
Abingdon, England
UK
Catherine De Greef
Brucells
Brussels, Belgium
Ugo DOro
Chiron
Siena, Italy
Andrea Splendiani
Leaf Bioscience
Milan, Italy
Charles Nicolette
Argos Therapeutics
Durham, USA
Philippe Pierre
CNRS-INSERM-Univ Med
Marseille, France
Duccio Cavalieri
University of Florence
Florence, Italy
Sandra Gessani
Istituto Superiore di Sanit
Rome, Italy
Nicolas Glaichenhaus
Universit de Nice-Sophia Antipolis
Valbonne, France
Benedita Rocha
INSERM U591 Institut Necker
Paris, France
Federica Sallusto/Antonio Lanzavechia/Markus Manz/
Mariagrazia Uguccioni
Institute for Research in Biomedicine
Bellinzona, Switzerland
Mark Suter
University of Zurich
Zurich, Switzerland
Kris Thielemans
Medical School of the Vrije Universiteit Brussel
Brussels, Belgium
Filippo Petralia
SEKMED
Milan, Italy
Sebastian Amigorena
Institut Curie, Paris, France
Alberto Mantovani
Istituto Clinico Humanitas
Milan, Italy
C J M Melief
Leiden University Medical Center
Leiden, Netherlands
Hermann Wagner
Technische Universitt Mnchen
Munich, Germany
Laurence Zitvogel
ERM0208 INSERM
Villejuif, France
Paola Ricciardi-Castagnoli
Universit degli Studi di Milano-Bicocca
Milano, Italy
Matthias Mann
Max-Planck-Institut fr Biochemie
Martinsried, Germany
153
ENACT
European Network
for the Identification
and Validation
of Antigens and
Biomarkers in Cancer
and their Application
in Clinical Cancer
Immunotherapy
BACKGROUND
AIMS
This project aimed to identify markers of response and
tumour antigens associated with ovarian, breast and
prostate cancer, as well as melanoma progression and
resistance to immunotherapy. A technological base was
established for vaccine development (not necessarily
restricted to cancer vaccines), and a better understanding
of the basic biological mechanisms underlying antigen
presentation and recognition of tumours by CD8+ and
CD4+ T lymphocytes and NK cells emerged.
154
Acronym: ENACT
Project Coordinator:
Prof. R Rees
Partners:
Italo Dodi
Nottingham Trent University
Nottingham, England
UK
Elissaveta Naumova
University Hospital
Sofia, Bulgaria
Graham Pawelec
Abt.Innere Medizin II Zentrum
fr Medizinische Forschung
Tbingen, Germany
Rolf Kiessling
Karolinska Institute
Stockholm, Sweden
Federico Garrido
Hospital Universitario
Granada, Spain
Dirk Schadendorf
Gustav Gaudernack
The Norwegian Radium Hospital
Oslo, Norway
Graham Ball
Loreus Ltd
Nottingham, England
UK
Costas Baxevanis
Hellenic Anticancer Institute
Athens, Greece
Mike Whelan
St Georges Hospital Medical School
London, England
UK
Francine Jotereau
INSERM U463
Nantes, France
Piotr Laidler
Jagiellonian University
Krakw, Poland
Aija Line
University of Latvia
Riga, Latvia
155
ADVAC-EC
Advanced Vaccinology
Training for Scientists
from ACC and Developing
Countries: From
Genomics to Vaccination
Strategies for
Communicable Diseases
Linked to Poverty
BACKGROUND
AIMS
The overall objective of the ADVAC-EU project was
to create a critical mass of people, in Europe and in
developing countries, with a sufficiently broad knowledge
of vaccinology to be able to play a leading role in decisionmaking processes concerning the following:
156
Acronym: ADVAC-EC
Project Coordinator:
Paul-Henri Lambert
Foundation Mrieux
Lyon, France
Tel: +41 22 37 95 783/777
E-mail: [email protected]
Partner:
Claire-Anne Siegrist
CMU Centre of Vaccinology
Geneva, Switzerland
157
NeutNet
Acronym: NeutNet
Standardisation of
HIV Neutralisation
Assays to be Used
in Vaccine Research
and Clinical Trials
BACKGROUND
It is well established that antibodies play an important
part in protection against viral diseases such as measles
and influenza, but the role of neutralising antibodies in
HIV-1 protection and pathogenesis remains to be further
defined. Production of an antibody response with a broad
neutralising activity against primary isolates of multiple
HIV-1 subtypes continues to be a desired characteristic
for candidate HIV vaccines. To support the evaluation
of phase I, II and III human vaccine trials testing new
HIV immunogens, it is important to standardise as far
as possible and apply high-throughput specific and
reproducible HIV neutralisation assays. Standardised
HIV neutralisation assays make it possible to compare all
vaccine efforts throughout the world.
AIMS
The project primarily aims to coordinate activities
to standardise methods for the measurement of
neutralising antibodies to HIV-1 to be used in vaccine
research and clinical trials. Other aims are:
developing and centralising the necessary reagents
to undertake the study;
preparing a draft protocol and guidelines for the
study;
organising an initial study to compare different
neutralisation methods using a number of wellknown monoclonal antibodies against a panel of
well-characterised viruses;
defining the methods for data analysis and
statistical comparisons of assay results from the
different participants;
organising a subsequent study to compare
polyclonal serologic reagents and defining the best
conditions to determine neutralising activity;
defining the needs of the scientific community
involved in both preventive and candidate materials
for evaluation;
organising a workshop in collaboration with WHO/
UNAIDS to discuss the results of the actions listed
158
Project Coordinator:
Gabriella Scarlatti
Vicky Polonis
Henry Jackson Foundation
Rockville, Maryland
USA
Terri Wrin
MonogramBio Inc
San Francisco, California
USA
Partners:
Susan Zolla-Pazner
New York University School of Medicine
New York City
USA
Harvey Holmes
National Institute for Biological Standards and Control
Pottersbar, England,
UK
David C Montefiori
Duke University Medical Center
Durham, North Carolina
USA
Christiane Moog
Saladin Osmanov
Quentin Sattentau
University of Oxford
Oxford, England,
UK
Lynn Morris
National Institute for Communicable Diseases
Johannesburg, South Africa
Ruengpung Sutthent
Mahidol University
Bangkok, Thailand
Vera Bongertz
Oswaldo Cruz Foundation
Rio de Janeiro, Brazil
159
COINFECT
Acronym: COINFECT
Malaria and
Coinfection:
New Insights
for Malaria Control
BACKGROUND
AIMS
The overall aim of the project is to deliver the information
and tools necessary for researchers in Asian countries to
produce robust data on malaria and helminth coinfection
and their immunological interaction. COINFECT will be
used to transfer knowledge and technology between
European, Malaysian and Indonesian experts in the area
of malarial and helminth coinfection, in order to prepare a
team with the ability to carry out high quality research in
this important area. This will be achieved by:
training in the setting up of specific epidemiological
studies;
developing questionnaires;
training medical staff in clinical assessment;
training in malarial diagnosis quality control;
training in helminth diagnosis quality;
standardised procedures for collection of blood by
finger prick;
demonstration of immunological methods that
measure antibodies and cytokines;
demonstration of molecular biological methods that
measure gene expression by quantitative PCR and
determine genetic polymorphisms.
Pilot study:
The trained personnel will carry out pilot studies to determine
the prevalence of malarial and helminth coinfections and the
immunological interaction between these two infections.
160
Project Coordinator:
Prof. Dr Maria Yazdanbakhsh
Leiden University Medical Center
Laboratory for Parasitology
Albinusdreef 2
Leiden, Netherlands
Tel:+31-7 15 26 50 67
E-mail: [email protected]
Partners
161
EURHAVAC
Acronym: EURHAVAC
European Network
for Harmonisation
of Malaria Vaccine
Development
BACKGROUND
AIMS
The projects aims are as follows:
the definition and implementation of decisionmaking processes for pharmaceutical development;
the definition and implementation of a clinical
development strategy;
the harmonisation of the evaluation criteria for
malaria vaccines;
the dissemination of knowledge and information
yielded by the three first action plans.
162
Project Coordinator:
Sren Jepsen
163
PAHPV-1
Acronym: PAHPV-1
BACKGROUND
AIMS
The aims of the project were as follows:
to collect epidemiological data on the incidence
of HPV16 and the immune status against the
virus in preparation for a future HPV vaccination
programme;
to include HPV education and screening in current
STD campaigns in Jakarta and Bali and share
the experiences with HPV researchers in other
developing countries.
164
Project coordinator:
Prof. Dr G G Kenter
E-mail: [email protected]
Partners:
165
REBAVAC
Novel Opportunities
to Develop Vaccines
to Control Antibiotic
Resistant Bacteria:
from the Trials Back
to the Laboratory
Acronym: REBAVAC
Project number: LSHM-CT-2006-037163
EC contribution: 121 200
Duration: 12 months
Type: SSA
Starting date: 1 September 2006
Project website: www.altaweb.eu/rebavac
BACKGROUND
AIMS
At the workshop the worlds most important experts in
vaccinology and immunology met healthcare providers,
industry representatives and public health experts and
discussed the key issues outlined above, including
how to use and improve currently available conjugate
polysaccharide vaccines to fight antibiotic resistant bacteria,
and how better to understand the contribution of innate and
adaptive components of the immune system in eliciting and
boosting immune responses to conjugate vaccines.
166
Project Coordinator:
Aldo Tagliabue
CEO ALTA Srl
Siena, Italy
Tel: +39-0 57 72 43 508
E-mail: [email protected]
167
Index by acronym
ADVAC-EC.................................................................... 156
FLUVACC........................................................................ 92
AIDS-CoVAC.................................................................. 40
HEPACIVAC.................................................................. 104
HEVAR......................................................................... 126
Allomicrovac.................................................................. 64
HIVAB............................................................................ 42
AUTO/ALLO CELL-HIV.................................................. 60
HIV VIROSOMES.......................................................... 68
AVIP.............................................................................. 56
ImmunoGrid................................................................... 46
BacAbs........................................................................... 30
ImmunoVacTB................................................................ 90
BOVAC......................................................................... 120
INNOVAC....................................................................... 44
CANCERIMMUNOTHERAPY........................................ 132
Intranasal H5vaccine.................................................... 98
LCVAC.......................................................................... 136
COINFECT.................................................................... 160
MALINV......................................................................... 80
CompuVac...................................................................... 28
MICROBEARRAY........................................................... 32
DC-THERA................................................................... 152
MimoVax..................................................................... 140
DC-VACC........................................................................ 36
MuNanoVac................................................................... 26
DEC-VAC........................................................................ 34
MUVAPRED................................................................... 20
DENDRITOPHAGES...................................................... 134
MVACTOR..................................................................... 50
DISSECT....................................................................... 106
Neotim........................................................................... 86
EDCTP.......................................................................... 146
NeutNet....................................................................... 158
EMVDA.......................................................................... 74
NOVADUCK................................................................. 112
ENACT.......................................................................... 154
OMVac......................................................................... 124
EPI-VAC......................................................................... 72
PAHPV-1..................................................................... 164
EPIVAC.......................................................................... 24
PANFLUVAC................................................................... 96
EURHAVAC.................................................................. 162
Pox-gene....................................................................... 62
EUROPRISE................................................................. 148
PRIBOMAL.................................................................... 76
FluVac............................................................................ 94
REBAVAC..................................................................... 166
RMVHIV........................................................................ 58
SARS/FLU VACCINE.................................................... 110
SARSVAC..................................................................... 108
SAVINMUCOPATH...................................................... 122
SCOOTT....................................................................... 116
SME-Malaria................................................................. 78
SUPASALVAC............................................................... 128
TB-VAC.......................................................................... 82
THERAVAC..................................................................... 38
TIP-VAC......................................................................... 70
TRANCHI..................................................................... 118
TRYPADVAC2.............................................................. 130
Universal Vaccine........................................................ 102
Vaccines4TB.................................................................. 88
VaccTIP.......................................................................... 48
VIAV.............................................................................. 66
VITAL........................................................................... 138
Index by coordinator
Austyn, Jonathan
DC-THERA....................................................................................... 152
Bartholeyns, Jacques
SUPASALVAC................................................................................... 128
Goudsmit, Jaap
PRIBOMAL........................................................................................ 76
Hilgers, Luuk A Th
FluVac................................................................................................ 94
DENDRITOPHAGES.......................................................................... 134
Boon, Thierry
CANCERIMMUNOTHERAPY............................................................ 132
Barrow, Paul
Boulange, Alain
TRYPADVAC2.................................................................................. 130
Huet, Jacqueline
MuNanoVac....................................................................................... 26
Jepsen, Soren
EURHAVAC...................................................................................... 162
Kenter, G G
PAHPV-1......................................................................................... 164
Bublot, Michel
Klatzmann, David
Cortese, Riccardo
Koopman, Gerrit
Cutting, Simon M
Lambert, Paul-Henri
Crisanti, Andrea
Leclerc, Claude
Daura, Xavier
Lehner, Thomas
Dolcetti, Riccardo
Leroy, Odile
Enjuanes, Luis
Liljestrom, Peter
Ensoli, Barbara
Ludewig, Burkhard
Epstein, Alberto
Lund, Ole
Ferrantelli, Flavia
Maizels, Rick
Glck, Reinhard
Mattner, Frank
Goovaerts, Danny
McCullough, Kenneth
NOVADUCK..................................................................................... 112
HEPACIVAC...................................................................................... 104
INNOVAC........................................................................................... 44
MICROBEARRAY............................................................................... 32
BacAbs............................................................................................... 30
VITAL............................................................................................... 138
DISSECT........................................................................................... 106
AVIP.................................................................................................. 56
HEVAR............................................................................................. 126
VIAV.................................................................................................. 66
SME-Malaria..................................................................................... 78
CompuVac.......................................................................................... 28
Pox-gene........................................................................................... 62
ADVAC-EC........................................................................................ 156
THERAVAC......................................................................................... 38
Allomicrovac...................................................................................... 64
EMVDA.............................................................................................. 74
VaccTIP.............................................................................................. 48
AIDS-CoVAC...................................................................................... 40
Vaccines4TB...................................................................................... 88
TRANCHI......................................................................................... 118
MimoVax......................................................................................... 140
PANFLUVAC....................................................................................... 96
Meinke, Andreas
OMVac............................................................................................. 124
BOVAC............................................................................................. 120
Mgone, Charles
EDCTP.............................................................................................. 146
Muster, Thomas
OGarra, Anne
DC-VACC............................................................................................ 36
Pizza, Mariagrazia
SARSVAC......................................................................................... 108
Ramne, Anna
Rappuoli, Rino
MUVAPRED....................................................................................... 20
Rees, R
Spetz, Anna-Lena
AUTO/ALLO CELL-HIV...................................................................... 60
Stanescu, Ioana
EPIVAC.............................................................................................. 24
Stiegler, Gabriela
HIV VIROSOMES.............................................................................. 68
Sutter, Gerd
MVACTOR......................................................................................... 50
Tagliabue, Aldo
REBAVAC......................................................................................... 166
Taylor, David W
SCOOTT........................................................................................... 116
Thole, Jelle
TB-VAC.............................................................................................. 82
berla, Klaus
ENACT.............................................................................................. 154
DEC-VAC............................................................................................ 34
TIP-VAC............................................................................................. 70
Rnia, Laurent
Rossi, Elda
Rottenberg, Martin
Voss, Gerald
Scala, Giuseppe
Wolf, Hans
Scarlatti, Gabriella
Yazdanbakhsh, Maria
MALINV............................................................................................. 80
ImmunoGrid....................................................................................... 46
Neotim............................................................................................... 86
EPI-VAC............................................................................................. 72
NeutNet........................................................................................... 158
Seipelt, Joachim
FLUVACC............................................................................................ 92
Intranasal H5vaccine........................................................................ 98
CHIMERIC VACCINES...................................................................... 100
Shattock, Robin
EUROPRISE..................................................................................... 148
Sirard, Jean-Claude
SAVINMUCOPATH.......................................................................... 122
ImmunoVacTB.................................................................................... 90
LCVAC.............................................................................................. 136
RMVHIV............................................................................................ 58
HIVAB................................................................................................ 42
COINFECT........................................................................................ 160
European Commission
Vaccines for Humans Research funded by the European Union
Luxembourg: Office for Official Publications of the European Communities
2008 176 pp. 21.0 x 29.7 cm
ISBN
DOI
978-92-79-09455-2
10.2777/84341
KI-81-08-337-EN-C
Europe has a long and successful tradition for vaccine research in both public and private institutions. Two-thirds of
global vaccine R&D is being conducted by European organisations and almost 90% of all vaccine production takes
place in Europe. Europe is therefore well positioned to take on new challenges in vaccine research, and exploit the
immense opportunities that are opening up in this field of science. The European Commissions Sixth Framework
Programme for Research (FP6) has been an important catalyst in this direction and has provided substantial
momentum to further advance the European activities in vaccine research.
This publication compiles the projects on human vaccine research that were initiated during FP6 (2002-2006). Most
of the projects have been funded through the Health theme of FP6, although important contributions came from
the Food theme, the Information Society theme (IST) and from cross-cutting activities on international cooperation
(INCO), and the support activities to small and medium-sized enterprises (COOP). Many of the projects will not
conclude before 2009 or later, but they are already beginning to deliver important results. Taken together they
provide evidence of a vibrant, visionary and ambitious research community, where hundreds of scientists are working
together to discover and develop new vaccines for the world.