G Model

ARTICLE IN PRESS

JVAC 16139 1–7

Vaccine xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Vaccine journal homepage: www.elsevier.com/locate/vaccine

HPV Monograph Series: General Overview

1

TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development

2

3

4 5 6 7 8 9 10 11 12 13 14

Q1

Mark J. Geels a,1 , Regitze Louise Thøgersen a,1 , Carlos A. Guzman b , Mei Mei Ho c , Frank Verreck d , Nicolas Collin e , James S. Robertson c,2 , Samuel J. McConkey f , Stefan H.E. Kaufmann g , Odile Leroy a,∗ a

European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Im Neuenheimer Feld 326 - 3.OG, 69120, Heidelberg, Germany Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany c National Institute for Biological Standards and Control, Department of Health-Medicines and Healthcare Products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire, EN6 3QG, United Kingdom d Department of Parasitology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, Netherlands e Vaccine Formulation Laboratory, University of Lausanne, Chemin des Boveresses 155, Epalinges 1066, Switzerland f Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, 123 St. Stephens Green Dublin 2, Ireland g Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany b

15

16 32

a r t i c l e

i n f o

a b s t r a c t

17 18 19

Article history: Available online xxx

20 21 22 23 24 25 26 27 28 29 30 31

Keywords: Vaccine Research and Development European Research Infrastructures Services Standardisation Harmonisation Animal Models Adjuvants Biomarkers FP7

TRANSVAC was a collaborative infrastructure project aimed at enhancing European translational vaccine research and training. The objective of this four year project (2009–2013), funded under the European Commission’s (EC) seventh framework programme (FP7), was to support European collaboration in the vaccine field, principally through the provision of transnational access (TNA) to critical vaccine research and development (R&D) infrastructures, as well as by improving and harmonising the services provided by these infrastructures through joint research activities (JRA). The project successfully provided all available services to advance 29 projects and, through engaging all vaccine stakeholders, successfully laid down the blueprint for the implementation of a permanent research infrastructure for early vaccine R&D in Europe. © 2015 Published by Elsevier Ltd.

1. Introduction

33

1.1. Background and rationale of TRANSVAC

34

Q2 Abbreviations: AIDS, Acquired immunodeficiency syndrome; BCG, Mucobacterium bovis bacille calmette–guérin; DTH, Delayed type hypersensitivity; EC, European Commission; ELISpot, Enzyme-linked immunospot; EVRI, European Vaccine Research and Development Infrastructure; FP7 , Seventh framework programme (FP7); HIV, Human immunodeficiency virus; ICS, Intracellular cytokine staining; IFN␥, Interferon gamma; IGRA, Interferon gamma release assays; IL, Interleukin; JRA, Joint research activities; NHP, Non-human primate; pCD, Plasmacytoid dendritic cells; PPD, Purified protein derivative; R&D, Research and development; RI, Research infrastructure; RNA, Ribonucleic acid; SAC, Scientific Advisory Committee; SIV, Simian immunodeficiency virus; SOP, Standard operating procedures; TB, Tuberculosis; Th, T helper cells; TLR, Toll-like receptor; TNA, Transnational access; USP, User Selection Panel; VFL, Vaccine Formulation Laboratory; WP, Work package. ∗ Corresponding author. Tel.: +49 6221 565890; fax: +49 6221 565727. E-mail addresses: [email protected], [email protected] (O. Leroy). 1 Contributed equally. 2 Independent Vaccine Expert now retired from this affiliation.

Through the natural legacies of Edward Jenner (1749–1823), Louis Pasteur (1822–1895) and Emil von Behring (1854–1917), vaccine development in Europe has been deeply ingrained in life science research. A key role in the TRANSVAC network was played by state-owned R&D and production facilities, alongside academic and private partners. However, in the last decade this environment has been changing by the decision of European governments, mostly for economic reasons, to withdraw funding from stateowned vaccine production facilities [1,2]. This shift jeopardises the European vaccine R&D network and has increased the risk of a slowdown in innovative vaccine research and in products progressing through the development pipeline. This has an immediate effect on knowledge and competencies, and also increases the fragmentation of expertise and facilities. In addition, there is a lack of synergy

http://dx.doi.org/10.1016/j.vaccine.2015.01.079 0264-410X/© 2015 Published by Elsevier Ltd.

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

35 36 37 38 39 40 41 42 43 44 45 46 47 48

G Model JVAC 16139 1–7 2

ARTICLE IN PRESS M.J. Geels et al. / Vaccine xxx (2015) xxx–xxx

Fig. 1. PERT diagram of project.

49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88

between the private and public sector, and a lack of commonly defined strategies by the public sector of European Union member states. A supra-national action plan for the vaccine development community is required to combat this situation. The European vaccine development community could benefit from the establishment of an efficient, sustainable and collaborative research infrastructure (RI) based on shared and goals in order to create a common agenda, address existing and future challenges, pool resources, and develop cost- and time-efficient strategies [3]. RIs play an important role in the advancement and exploitation of knowledge and technology. Aside from researchers and institutions, large RIs are often a vital requirement for a more effective scientific system in which, for example, overcapacity can be more easily distributed and regulatory compliant quality systems for products and processes can be implemented and maintained. RIs can provide quick leverage for new applications that extend beyond their original purpose and thus act as a structuring force in the scientific system. Altogether, a complex process like vaccine development can be most effectively supported by a multi-disciplinary RI. TRANSVAC, a collaborative RI project funded under the EC FP7, operated between 2009 and 2013 as a joint effort of European groups in the field of vaccine R&D with a total budget of Euro 9.9 million. It was aimed at accelerating the development of promising vaccine candidates by developing, optimising and standardising state-of-the-art processes and facilities available to vaccine developers to bridge the gap between bench research and clinical assessment of novel vaccines. This improvement occurred at three levels (Fig. 1): (1) TNA: provided researchers with free access to vaccine development services such as adjuvant formulation, animal models, reference reagents/standards and global analyses (e.g. transcriptomics). As core services of the consortium, these were made available free of charge to European vaccine R&D groups. Through a competitive peer-review process, European groups working in vaccine development could benefit from the expertise, reagents, and facilities of the TRANSVAC consortium. (2) JRA: within the consortium the focus of internal research was on improving the use of global analysis platforms, adjuvants and animal models. All project efforts in JRA were designed and executed on the basis of applicable

regulatory requirements. Therefore, inter-laboratory harmonisation, qualification and standardisation through the development of standard operating procedures (SOPs) and provision of standardised reagents were key objectives. (3) Networking: the consortium provided training in vaccine development, harmonisation of assays and global analyses (e.g. microarrays), and optimal use of animal models. Finally, to ensure leverage of the scientific and operational lessons learned, the consortium engaged in stakeholder consultations with key representatives from academia, public health institutions, pharmaceutical industry, funding organisations and regulators, and developed a European roadmap for vaccine R&D infrastructures [4]. The programme work plan included work packages (WPs) in the following six activities: 1. Coherent development of novel and improved vaccine formulations (WP 1, 8 & 11). 2. Development and production of recombinant vaccine candidates and cell substrates for the manufacture of vaccines (WP 5, 6 & 8). 3. Evaluation of vaccine candidates in different animal models (WP 2, 8, 9 & 12). 4. Definition of biomarkers of protective immunity through global analyses of host responses after vaccination (WP 3, 8, 10 & 13). 5. Harmonisation of immunological assays for preclinical studies and clinical trials (WP 4, 7, 8 & 14). 6. Project management; TRANSVAC stakeholder consultation (WP15).

89 90 91 92 93 94 95 96 97 98 99 100 101 102

103 104 105 106 107 108 109 110 111 112 113 114 115

2. Results and milestones reached

116

2.1. Transnational access

117

The core of the TRANSVAC project consisted of providing access to services crucial to early-phase vaccine development, such as adjuvants, animal models, reference reagents and global gene expression platforms (e.g. microarrays and next generation deep sequencing) to vaccine R&D projects from European research groups (Table 1).

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

118 119 120 121 122 123

G Model JVAC 16139 1–7

ARTICLE IN PRESS M.J. Geels et al. / Vaccine xxx (2015) xxx–xxx

3

Table 1 Overview of TRANSVAC service providers and servicesa . Service provider

Services

Access provided in % (number of studies or samples)

Biomedical Primate Research Centre, The Netherlands Helmholtz Centre for Infection Research, Germany

In vivo vaccine safety and immunogenicity profiling in NHP models Pre-clinical studies in the murine system to assess the immunogenicity, safety and efficacy of specific vaccine formulations Deep sequencing and comparative transcriptome analysis service based on the Illumina platform in the form of samples (RNA/DNA) In vivo safety and/or immunogenicity evaluation of vaccines in pigs Vaccine safety and immunogenicity profiling in NHP models To prepare specified standards (vials) as well as curatorship and shipping of specified reference standards

100% (1 study) 100% (1 study)

Helmholtz Centre for Infection Research, Germany Central Veterinary Institute, The Netherlands Public Health England, United Kingdom National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, United Kingdom Max Planck Institute for Infection Biology, United Kingdom University of Regensburg, Germany University of Lausanne, Vaccine Formulation Laboratory, Switzerland a

Full service and experimental realisation for global transcriptome analysis of gene regulation using the Agilent microarray platform Full service and experimental realisation for global transcriptome analysis of gene regulation using the Affymetrix microarray platform Vaccine formulation with adjuvants, which are free of intellectual property and patent issues (or which are accessible through material transfer agreements)

142

143

2.2. Joint research activities

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141

152

TRANSVAC JRAs were designed to enhance the development, harmonisation, standardisation and qualification of vaccine development technologies and services existing within the consortium. The developed services and activities performed during the project were numerous, and are described in detail elsewhere (www.transvac.org). Here, we highlight TNA and JRA activities and services that were in highest demand. Fig. 2 gives an overview of the budget according to different disease areas for TNA and JRA combined.

153

2.3. Adjuvants and vaccine formulation

144 145 146 147 148 149 150 151

154 155 156 157 158 159 160 161 162 163

100% (1 study) 100% (1 study) 100% (164 vials)

117% (840 samples) 100% (420 samples) 106% (50 formulations)

Not all partners in TRANSVAC were service providers. Full list of partners is given in acknowledgements.

TRANSVAC has set up two bodies for the selection of proposals for access to services: first, the Scientific Advisory Committee (SAC) consisting of independent external scientists and secondly, the User Selection Panel (USP) composed of both independent external experts and the TRANSVAC TNA WP leaders. The SAC reviewed and ranked proposals based on scientific and experimental quality. The USP, which had the ultimate responsibility for the selection of the applications, considered opinions of the SAC and completed the peer-review process according to the feasibility of the access proposal with regard to provision of available infrastructure. Between January 2011 and April 2013 the consortium received 55 eligible applications and provided access to 29 projects from European research groups in both the public and private sectors. Table 2a and b provides a breakdown of the eligible TNA applications and TNA grantees per disease area and geographical origin of grantees. TNA included logistical, technological and scientific support as well as any specific on-site training needed from the service provider to the applicant. The type of projects that were granted TNA ranged from discovery to clinical in nature.

124

96% (146 samples)

Access to modern adjuvant systems and to the associated formulation know-how is often considered as a major challenge in vaccine development. TRANSVAC provided co-funding for the establishment of the Vaccine Formulation Laboratory (VFL), a World Health Organization Collaborating Centre, at the University of Lausanne. The VFL facilitates access to adjuvants that are not covered by intellectual property rights or are available under licence agreements, and provides support for vaccine formulation through advice, training, preclinical evaluation of adjuvanted vaccines, and technology transfer. In the context of the TRANSVAC project,

several adjuvant-vaccine formulations using malaria, tuberculosis (TB) or hepatitis B antigens were developed and tested in mice for their immunogenicity. SOPs were also developed for future validation assays to characterise the vaccine formulations. 2.4. Global molecular analysis of vaccine responses

Table 2 a. Overview of eligible TNA applications and TNA grantees per disease area. Disease area

Number of eligible projects received

Number of projects funded

Cancer Dengue Hepatitis Human immunodeficiency virus Human papiloma virus Immunopotenciating technologies Influenza Lyme disease Malaria Mumps Bordetella pertussis Streptococcus pneumoniae Tuberculosis Total

3 3 2 8 1 13 4 1 5 1 2 1 11 55

2 1 0 4 0 9 2 1 2 1 2 1 4 29

Table 2b Overview of eligible TNA applications and TNA grantees per European member state/associated country Number of Country Number of eligible projects received projects funded 3 1 1 1 4 10 3 2 1 11 1 6 4 7 55

165 166 167

168

Global molecular analyses are exploited for a better understanding of the human response to infection and vaccination [5]. An understanding of underlying molecular events in patients

Austria Belgium Croatia Czech Republic Denmark France Germany Ireland Poland Spain Sweden Switzerland The Netherlands United Kingdom Total

164

1 1 1 1 3 8 0 2 1 3 0 1 3 4 29

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

169 170 171

G Model

ARTICLE IN PRESS

JVAC 16139 1–7

M.J. Geels et al. / Vaccine xxx (2015) xxx–xxx

4

€49,073

€8,215

Cancer Dengue

€634,254 Human immunodeficiency virus Immunopotenciating technologies €3,232,683

€1,437,069

Influenza Lyme disease

€567,150

Malaria Mumps

€1,179,358

Bordetella pertussis €49,287

€7,668 €29,572

Streptococcus pneumoniae Tuberculosis

Fig. 2. Total TRANSVAC budget per disease combined for TNAs and JRAs.

200

with different forms of disease (e.g. subclinical, mild, severe), as well as in responders and poor/non-responders to vaccination, is critical for future improved intervention strategies. In this regard, high-throughput technologies, including microarray analyses and ribonucleic acid (RNA) deep sequencing, which allow global gene expression profiling within different study groups [6] were harmonised and provided by TRANSVAC. Side-by-side comparisons using different technologies have been carried out. For example, a first stand-alone data analysis based on Agilent microarrays and a phase I clinical trial with Mucobacterium bovis bacille calmette–guérin (BCG), using two different tuberculin skin test groups [purified protein derivative (PPD)-positive and -negative] was completed by Max Planck Institute for Infection Biology in Germany. The results indicate that although only a small number of genes were differentially regulated between the PPD+ and PPD− study groups, the group-specific signatures were readily measureable with a more profound response in the PPD+ group as compared to the PPD– group. The consortium made considerable efforts to standardise research on biomarkers of host responses post-vaccination against TB, malaria and human immunodeficiency virus (HIV). The research focussed on next-generation sequencing and microarrays (Agilent, Affymetrix). The main conclusion was that platforms for use in preclinical and clinical studies are insufficiently standardised. Coordinated efforts should continue to harmonise the experimental design of these studies, as well as the establishment of internal standards and controls. This will ensure comparability and efficiency of the global analyses performed on preclinical and clinical datasets [6].

201

2.5. Model and technology development

172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199

202 203 204 205 206 207 208 209 210 211 212 213 214 215 216

Immunological assays are commonly used for new vaccine development, from discovery, preclinical to clinical studies [7,8]. In TRANSVAC several rounds of harmonisation of immunological assays [flow cytometry intracellular cytokine staining (ICS), enzyme-linked immunospot (ELISpot), and antigen-specific interferon gamma release assays (IGRA)], which are applicable for use in preclinical studies and clinical trials, were performed. During the harmonisation process, critical steps and reagents contributing to variability in results were identified and optimal experimental conditions were refined in each of these assays. Finally, SOPs for harmonised assays, which take into account regulatory considerations, were established (free access on www.transvac.org/RTD). The coefficient of variation of results between laboratories performing these assays was reduced considerably using harmonised SOPs compared to data using protocols from earlier rounds [Steven

Smith, Manuscript in Preparation]. These SOPs will be used for further qualification of assays across the laboratories involved. Preclinical evaluation of new candidates and strategies is a key element in vaccine R&D, especially in disease areas (e.g. malaria, TB, HIV) where a clear and unambiguous understanding of the protective immune mechanisms is missing. Under the programme’s JRA instrument, TRANSVAC undertook efforts to refine and improve small and large animal models for vaccine evaluation. These activities aimed at in-depth definition of specific model characteristics and extension of the available reagents for mouse, guinea pig, pig and non-human primate (NHP) macaque models. Technologies and tools were developed for generic immunological and clinical evaluations in support of vaccine safety, immunogenicity and efficacy analyses. From in vivo animal experiments materials have been banked and fed into the global molecular analysis platform. Wherever applied, these developments were implemented and impacted immediately on the TNA provided by TRANSVAC. The generation of humanised mice represents a promising tool to facilitate the development of new immune interventions and drugs. Within TRANSVAC two types of system were established at the Helmholtz Centre for Infection Research in Germany. The first system is based on mice reconstituted with human hepatocytes, which can be infected with hepatotropic agents (e.g. HCV, HBV). This model enables the analysis of diverse therapeutics (e.g. anti-viral drugs, biologicals) to perform a screening and selection of most promising candidates. Second, mice were reconstituted with a human immune system. Different model systems were established based on mouse strains engrafted with hematopoietic progenitor cells obtained from human cord blood or fetal liver. The resulting animals harbour a full repertoire of human innate and adaptive immune cells. These mice can be exploited for the characterisation of distinct immune cells infected with pathogens, as well as for the analysis of immune modulators targeting specific leukocyte sub-populations. To thoroughly assess the effect of adjuvants on immune cells within TRANSVAC highly sophisticated multiparametric immune monitoring approaches based on flow cytometry and multiplex analysis were also established. The models can be harnessed, among other applications, to test anti-infectives and biologicals against human tropic pathogens (e.g. HIV). Due to the lack of fully functional lymph nodes, additional efforts are needed before these systems can be used for vaccine evaluation studies. However, preliminary studies demonstrated that lymph node formation can be improved by co-engraftment of lymphoid tissue inducer cells. Species of macaques, rhesus monkeys (Macaca mulatta) and cynomolgus monkeys (Macaca fascicularis) in particular, are the most widely used NHP models in vaccine research. However, it is deceptive to consider NHP macaques as a single model entity as data in literature suggest that cynomolgus versus rhesus macaques, as well as different spectrotypes within these species, present with different susceptibility to infectious diseases including malaria, TB and simian immunodeficiency virus (SIV) infection [9–12]. Recognising the possible implication of macaque population heterogeneity on vaccine research, TRANSVAC JRA activities aimed at investigating innate and adaptive immune response patterns in macaque species (cynomolgus versus rhesus) and rhesus spectrotypes (Chinese versus Indian). While grossly showing similar response patterns, cynomolgus as well as rhesus macaque plasmacytoid dendritic cells (pDC) showed interleukin (IL) 12 expression upon specific toll-like receptor (TLR) stimulation, which is absent in their human counterparts (V. Sommandas et al., manuscript in preparation). Relevant for TB, a head-to-head comparison under TRANSVAC demonstrated a higher susceptibility phenotype of Indian over Chinese rhesus macaques supported by various readouts of disease (F.A.W. Verreck et al., manuscript in preparation). BCG-induced immunity in both cohorts has been investigated as a

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282

G Model JVAC 16139 1–7

ARTICLE IN PRESS M.J. Geels et al. / Vaccine xxx (2015) xxx–xxx

5

Table 3 Strategic goals of EVRI. -Establish a comprehensive platform for European vaccine R&D, providing scientific and technical services to European vaccine stakeholders and addressing major challenges in vaccine development. -Coordinate joint research activities and stimulate technological innovation derived from the basic research it fosters. -Create a reference vaccine R&D platform for the European Medicines Agency (EMA) performing ‘regulatory science’ activities. -Build world-wide linkages between centres of excellence to address the global threat of infectious diseases. -Build capacity by promoting theoretical and practical education in vaccinology.

292

prototypical vaccine inducing a Th1 type cellular immune response, and while transcriptome data from global molecular analyses still need to be mined, the segregation of biological and immunological responses in these NHP rhesus populations is underpinned by these studies and provides a perspective on possible identification of signatures of disease/protective immunity. The results of the head-to-head approach enabled by the TRANSVAC infrastructure (JRA) programme are of great importance for cohort stratification, and provide another milestone in preclinical vaccine evaluations in outbred NHP.

293

2.6. Networking and capacity building

283 284 285 286 287 288 289 290 291

323

Capacity building and networking activities in TRANSVAC were presented in several workshops encompassing different areas. Four workshops examined the use of animal models ranging from statistical analyses in animal studies to hands-on training using case studies in the in vivo setting. This included a symposium on the pig animal model which was held at the Central Veterinary Institute in The Netherlands bringing together participants from 18 countries from Europe, North America, Africa and Asia. The pig immune system is more comparable to humans than that of rodents and provides a crucial link between small animals and NHPs. Increasing knowledge about pigs provides opportunities for an improved understanding of vaccines and vaccine development [13]. During the symposium relevant issues such as genomics, immunology, model infectious diseases, and vaccine delivery were addressed. Secondly, capacity building activities involved two training courses on practical approaches to vaccine development, held in September 2012 and March 2013 at the Vaccine Formulation Laboratory in Lausanne, Switzerland. For each course, 15 participants working in vaccine development were selected through a competitive process. Successful applicants received a week-long course on topics ranging from antigen selection, pre-clinical evaluation of vaccines in animal models, vaccine formulation and clinical development. All participants have evaluated the courses with high ratings and positive feedbacks, highlighting the increasing demand of training in vaccine development within the European vaccine community. However, due to the limitations of funds it was not possible to enrol all eligible candidates and the end of the project threatened the continuation of this high in demand course. Therefore, flexible funding and the possibility of (private sector)-sponsoring should be explored to sustain the course.

324

2.7. Stakeholder series and road mapping exercise

294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322

325 326 327 328 329 330 331 332 333 334 335

The TRANSVAC consortium, in collaboration with European vaccine stakeholders, designed a roadmap aimed at securing sustainable vaccine R&D RIs in Europe beyond the lifetime of the TRANSVAC project. Using a bottom–up approach the needs and priorities of European vaccine R&D were identified through a series of stakeholder consultations and four workshops attended by representatives from industry, academia, biotech, regulatory agencies, funders and patient organisations. This three year consultation led to a wealth of critical information compiled in the blueprint for a permanently funded RI in vaccine R&D, the European Vaccine Research and Development Infrastructure (EVRI) [4] (Table 3).

The EVRI strives to be a pan-European infrastructure that accelerates product development and at the same time reduces cost through the optimal use of existing national research capacities. It will build on existing networks, capacities and platforms such as those developed by TRANSVAC. The EVRI’s activities will focus on basic research, discovery, support of bioinformatics, preclinical research, early clinical research, and technology and expertise that are transferrable to other projects. Both human and veterinary vaccines will be within the scope of EVRI, including prophylactic as well as therapeutic vaccines for disease targets in humans. Given appropriate political and financial commitment from relevant national and European entities, as well as private sector and other stakeholders can be secured, the EVRI could be fully operational by 2016. 3. Outlook and conclusion To foster endurable European leadership and innovation in vaccine R&D for both human and animal diseases, research activities and infrastructures across the European research area need to be harmonised in a mutually supportive, world-class European RI. The success of the FP7-funded TRANSVAC project proved to be a valuable exercise by demonstrating the capability of vaccine stakeholders to share their competencies, facilities and knowledge, by collectively pooling resources, by building a sustainable network, by providing European scientists with access to expert services, and by addressing major challenges in vaccine R&D. TRANSVAC reached virtually all of the R&D goals planned within its four year life-time, including its core targets: the delivery of services to vaccine research groups and the continued development, optimisation and qualification of those services. It has additionally set up an excellent policy basis for further development of a European vaccine RI, the EVRI. TRANSVAC was a short-term project, which did not include income-generating activities to make it self-sustainable. However, the need for continuation and further integration of the European vaccine research community remains critical. RIs that consist of a functional network of partners–compared to single-site infrastructures–need time to develop, to acquire knowledge of new processes and to capitalise on investments, both financial and human. The lack of follow-up funding, e.g. European framework funding, despite meeting all primary endpoints, is the greatest risk to the successful implementation of RIs as they are vitally dependent on EC and national research funding. The JRAs of TRANSVAC demonstrated that through collaborative research the quality of vaccine development services can be improved substantially. Moreover, strong international relationships needed for addressing state-of-the-art vaccine research issues are forged. Numerous high-priorities, cross-sectional topics in vaccine R&D remain to be addressed to accelerate the development of novel technologies into practical use. This is in turn critical to access and exploit the innovation potential within the EC. Services offered by TRANSVAC to European vaccine developers were in high demand and contributed to the advancement of over 29 research projects at the (pre-)clinical stage of development. One of the key experiences from TRANSVAC was that the services, which were unique and in high value were most in demand compared to services, which were more ubiquitously available

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

336 337 338 339 340 341 342 343 344 345 346 347 348

349

350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390

G Model JVAC 16139 1–7

M.J. Geels et al. / Vaccine xxx (2015) xxx–xxx

6 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445

446

447 448 449 450 451 452 453

ARTICLE IN PRESS

within the research community. In the design of EVRI one should widen the provision of access to cutting-edge services and by improving both their quantity and quality. In EVRI, the portfolio of services provided through the transnational access should be subjected to periodical review and, if needed, be adjusted based on the recommendation of experts from the vaccine industry to ensure maximum alignment with the needs of all relevant stakeholders. This versatile element (both in process and administration) was lacking in TRANSVAC. Finally the application and evaluation process for transnational access should be streamlined reducing the time from application to service provision. Designing a comprehensive one-step evaluation process and different evaluation tracks could enable this. Additionally, a simplified track for the less expensive and less complex services, and a more detailed track for more expensive and more complex services should be implemented. Future RIs in vaccine R&D should expand their scope of activities to include additional topics, such as alternative in vitro models for screening, selection and prioritisation of vaccine candidates, as well as for assessing vaccine safety and efficacy. The 3Rs (replacement, reduction and refinement) in animal experimentation aims to reduce number of animals used is a crucial focal point in worldwide vaccine R&D and public policy. This can in turn adequately be addressed by multidisciplinary RIs. To ensure success and sustainable collaboration of future vaccine RIs, it is crucial that their mandates, scope and objectives are adopted by all relevant stakeholders, including private and public funders as well as regulatory authorities. By proposing the establishment of EVRI, the TRANSVAC consortium and collaborating European vaccine stakeholders declare their willingness to continue investing efforts into the integration and implementation of existing national and regional vaccine RIs and initiatives of panEuropean interest through collaborative projects. Accessible to the entire European vaccine development community, EVRI can especially help researchers from EU member states in which access to specialised vaccine RIs is limited. It will maximise synergy, optimise the use of existing RIs and limit duplication of efforts across Europe. Since vaccine R&D challenges are common worldwide and they would be best addressed through global, international collaborations, EVRI must maintain a permanent networking and collaboration with international organisations such as WHO and national regulators as well as international funding organisations to ensure that the services provided are relevant and are catering to the global vaccine R&D challenges. International stakeholder consultations should prove a first step in this process. TRANSVAC has proved to be an endeavour in vaccine R&D supported by all stakeholders with significant output and outcome. RIs have been demonstrated to make increasingly significant contributions to boosting Europe’s research and innovation potential. If sufficient political and financial commitment can be secured from relevant national and European policy-makers, funders and other stakeholders, the establishment of an EVRI will improve European competitiveness in this crucial health area and will provide a model for collaborative efforts in the development of international platforms in other research areas.

Disclaimer This article summarises the outcome, discussion and opinions of scientists/experts involved in the TRANSVAC project. This article does not necessarily represent the positions or the stated policy of the authors or the regulatory and other government organisations that were contributed in the preparation of this manuscript. This project has been funded with support from the European Commission. The information and views set out in this article are those of

the author(s) and do not necessarily reflect the official opinion of the European Union. Neither the European Union institutions and bodies nor any person acting on their behalf may be held responsible for the use which may be made of the information contained therein.

Conflict of Interest Stefan H. E. Kaufmann has been advisor to AERAS on TB vaccine development and to Qiagen on TB diagnostics.

Acknowledgments We would like to thank all the external experts of both the TRANSVAC SAC (Prof Jonathan Heeney, United Kingdom; Prof Claire Boog, the Netherlands; Dr Allen Saul, Italy; Dr Barry Walker, United States) and the TRANSVAC USP (Prof Martin Cranage, United Kingdom; Prof G. Davies, United Kingdom; Dr Michel Klein, Canada; Prof Roger LeGrand, France) for their continued efforts and support in the period 2010-2013. The TRANSVAC consortium: European Vaccine Initiative, Germany; Biomedical Primate Research Centre, The Netherlands; Helmholtz Centre for Infection Research, Germany; Vakzine Projekt Management GmbH, Germany; LIONEX GmbH, Germany; Central Veterinary Institute, The Netherlands; UK Department of Health, Public Health England, Centre for Emergency Preparedness and Response (CEPR, formerly under Health Protection Agency), United Kingdom; UK Department of Health, Medicines and Healthcare Products Regulatory Agency, National Institute for Biological Standards and Control (formerly under Health Protection Agency), United Kingdom; Max Planck Institute for Infection Biology, Germany; University of Regensburg, Germany; London School of Hygiene & Tropical Medicine, United Kingdom; University of Oxford, The Jenner Institute, United Kingdom; University of Lausanne, Switzerland; TuBerculosis Vaccine Initiative, The Netherlands. The TRANSVAC interested parties: European Molecular Biology Laboratory Grenoble, France; Instituto de Biologia Experimental e Tecnológica, Portugal; Institute for Translational Vaccinology, The Netherlands; Leids Universitair Medisch Centrum, The Netherlands. TRANSVAC was supported by the EU FP7 project TRANSVAC Q3 (FP7-INFRASTRUCTURES-2008-228403).

References [1] Thoegersen RL, Holder AA, Hill AV, Arnot DE, Imoukhuede EB, Leroy O. Comparative decline in funding of European Commission malaria vaccine projects: what next for the European scientists working in this field? Malar J 2011;10:255. [2] Geels MJ, Imoukhuede EB, Imbault N, van Schooten H, McWade T, TroyeBlomberg M, et al. European Vaccine Initiative: lessons from developing malaria vaccines. Expert Rev Vaccines 2011;10:1697–708. [3] Stephens P, Vaccine RD. Past performance is no guide to the future. Vaccine 2014. [4] Leroy O, Geels M, Korejwo J, Dodet B, Imbault N, Jungbluth S. Roadmap for the establishment of a European vaccine R&D infrastructure. Vaccine 2014;32:7021–4. [5] Weiner J, Kaufmann SHE. Recent advances towards tuberculosis control: vaccines and biomarkers. J Intern Med 2014;275:467–80. [6] Dutruel C, Thole J, Geels M, Mollenkopf H-J, Ottenhoff T, Guzman CA, et al. TRANSVAC workshop on standardisation and harmonisation of analytical platforms for HIV, TB and malaria vaccines: How can big data help? Vaccine 2014. [7] Goonetilleke N, Moore S, Dally L, Winstone N, Cebere I, Mahmoud A, et al. Induction of multifunctional human immunodeficiency virus type 1 (HIV-1)-specific T cells capable of proliferation in healthy subjects by using a prime-boost regimen of DNA- and modified vaccinia virus Ankara-vectored vaccines expressing HIV-1 Gag coupled to CD8+ T-cell epitopes. J Virol 2006;80:4717–28. [8] McShane H, Pathan AA, Sander CR, Keating SM, Gilbert SC, Huygen K, et al. Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat Med 2004;10:1240–4.

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

454 455 456 457 458

459

460 461

462

463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490

491

492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516

G Model JVAC 16139 1–7

ARTICLE IN PRESS M.J. Geels et al. / Vaccine xxx (2015) xxx–xxx

517 518 519 520 521 522 523 524

[9] Langermans JA, Andersen P, van Soolingen D, Vervenne RA, Frost PA, van der Laan T, et al. Divergent effect of bacillus Calmette-Guérin (BCG) vaccination on Mycobacterium tuberculosis infection in highly related macaque species: implications for primate models in tuberculosis vaccine research. Proc Natl Acad Sci USA 2001;98:11497–502. [10] Reimann KA, Parker RA, Seaman MS, Beaudry K, Beddall M, Peterson L, et al. Pathogenicity of simian-human immunodeficiency virus SHIV-89.6P and SIVmac is attenuated in cynomolgus macaques and associated with early Tlymphocyte responses. J Virol 2005;79:8878–85.

7

[11] Flynn S, Satkoski J, Lerche N, Kanthaswamy S, Smith DG. Genetic variation at the TNF-alpha promoter and malaria susceptibility in rhesus (Macaca mulatta) and long-tailed (Macaca fascicularis) macaques. Infect Genet Evol 2009;9: 769–77. [12] Ling B, Veazey RS, Luckay A, Penedo C, Xu K, Lifson JD, et al. SIV(mac) pathogenesis in rhesus macaques of Chinese and Indian origin compared with primary HIV infections in humans. Aids 2002;16:1489–96. [13] Meurens F, Summerfield A, Nauwynck H, Saif L, Gerdts V. The pig: a model for human infectious diseases. Trends Microbiol 2012;20:50–7.

Please cite this article in press as: Geels MJ, et al. TRANSVAC research infrastructure – Results and lessons learned from the European network of vaccine research and development. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.01.079

525 526 527 528 529 530 531 532 533

TRANSVAC research infrastructure - Results and lessons learned from the European network of vaccine research and development.

TRANSVAC was a collaborative infrastructure project aimed at enhancing European translational vaccine research and training. The objective of this fou...
565KB Sizes 0 Downloads 7 Views