1
|
Luo X, Liang R, Liang L, Tang A, Hou S, Ding J, Li Z, Tang X. Advancements, challenges, and future perspectives in developing feline herpesvirus 1 as a vaccine vector. Front Immunol 2024; 15:1445387. [PMID: 39328406 PMCID: PMC11424437 DOI: 10.3389/fimmu.2024.1445387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
As the most prevalent companion animal, cats are threatened by numerous infectious diseases and carry zoonotic pathogens such as Toxoplasma gondii and Bartonella henselae, which are the primary causes of human toxoplasmosis and cat-scratch disease. Vaccines play a crucial role in preventing and controlling the spread of diseases in both humans and animals. Currently, there are only three core vaccines available to prevent feline panleukopenia, feline herpesvirus, and feline calicivirus infections, with few vaccines available for other significant feline infectious and zoonotic diseases. Feline herpesvirus, a major component of the core vaccine, offers several advantages and a stable genetic manipulation platform, making it an ideal model for vaccine vector development to prevent and control feline infectious diseases. This paper reviews the technologies involved in the research and development of the feline herpesvirus vaccine vector, including homologous recombination, CRISPR/Cas9, and bacterial artificial chromosomes. It also examines the design and effectiveness of expressing antigens of other pathogens using the feline herpesvirus as a vaccine vector. Additionally, the paper analyzes existing technical bottlenecks and challenges, providing an outlook on its application prospects. The aim of this review is to provide a scientific basis for the research and development of feline herpesvirus as a vaccine vector and to offer new ideas for the prevention and control of significant feline infectious and zoonotic diseases.
Collapse
Affiliation(s)
- Xinru Luo
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) and Key Laboratory of Veterinary Biological Products and Chemical Drugs of Ministry of Agriculture and Rural Affairs (MARA), Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruiying Liang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) and Key Laboratory of Veterinary Biological Products and Chemical Drugs of Ministry of Agriculture and Rural Affairs (MARA), Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lin Liang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) and Key Laboratory of Veterinary Biological Products and Chemical Drugs of Ministry of Agriculture and Rural Affairs (MARA), Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Aoxing Tang
- Shanghai Veterinary Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Shaohua Hou
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) and Key Laboratory of Veterinary Biological Products and Chemical Drugs of Ministry of Agriculture and Rural Affairs (MARA), Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiabo Ding
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) and Key Laboratory of Veterinary Biological Products and Chemical Drugs of Ministry of Agriculture and Rural Affairs (MARA), Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zibin Li
- College of Life and Health, Dalian University, Dalian, China
| | - Xinming Tang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) and Key Laboratory of Veterinary Biological Products and Chemical Drugs of Ministry of Agriculture and Rural Affairs (MARA), Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
2
|
Andreacchio G, Longo Y, Moreno Mascaraque S, Anandasothy K, Tofan S, Özün E, Wilschrey L, Ptok J, Huynh DT, Luirink J, Drexler I. Viral Vector-Based Chlamydia trachomatis Vaccines Encoding CTH522 Induce Distinct Immune Responses in C57BL/6J and HLA Transgenic Mice. Vaccines (Basel) 2024; 12:944. [PMID: 39204067 PMCID: PMC11360449 DOI: 10.3390/vaccines12080944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Chlamydia trachomatis remains a major global health problem with increasing infection rates, requiring innovative vaccine solutions. Modified Vaccinia Virus Ankara (MVA) is a well-established, safe and highly immunogenic vaccine vector, making it a promising candidate for C. trachomatis vaccine development. In this study, we evaluated two novel MVA-based recombinant vaccines expressing spCTH522 and CTH522:B7 antigens. Our results show that while both vaccines induced CD4+ T-cell responses in C57BL/6J mice, they failed to generate antigen-specific systemic CD8+ T cells. Only the membrane-anchored CTH522 elicited strong IgG2b and IgG2c antibody responses. In an HLA transgenic mouse model, both recombinant MVAs induced Th1-directed CD4+ T cell and multifunctional CD8+ T cells, while only the CTH522:B7 vaccine generated antibody responses, underscoring the importance of antigen localization. Collectively, our data indicate that distinct antigen formulations can induce different immune responses depending on the mouse strain used. This research contributes to the development of effective vaccines by highlighting the importance of careful antigen design and the selection of appropriate animal models to study specific vaccine-induced immune responses. Future studies should investigate whether these immune responses provide protection in humans and should explore different routes of immunization, including mucosal and systemic immunization.
Collapse
Affiliation(s)
- Giuseppe Andreacchio
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Ylenia Longo
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Sara Moreno Mascaraque
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Kartikan Anandasothy
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Sarah Tofan
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Esma Özün
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Lena Wilschrey
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Johannes Ptok
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| | - Dung T. Huynh
- R&D Department, Abera Bioscience AB, 75184 Uppsala, Sweden
| | - Joen Luirink
- R&D Department, Abera Bioscience AB, 75184 Uppsala, Sweden
| | - Ingo Drexler
- Institute of Virology, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany; (G.A.)
| |
Collapse
|
3
|
Langenmayer MC, Luelf-Averhoff AT, Marr L, Jany S, Freudenstein A, Adam-Neumair S, Tscherne A, Fux R, Rojas JJ, Blutke A, Sutter G, Volz A. Newly Designed Poxviral Promoters to Improve Immunogenicity and Efficacy of MVA-NP Candidate Vaccines against Lethal Influenza Virus Infection in Mice. Pathogens 2023; 12:867. [PMID: 37513714 PMCID: PMC10383309 DOI: 10.3390/pathogens12070867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Influenza, a respiratory disease mainly caused by influenza A and B, viruses of the Orthomyxoviridae, is still a burden on our society's health and economic system. Influenza A viruses (IAV) circulate in mammalian and avian populations, causing seasonal outbreaks with high numbers of cases. Due to the high variability in seasonal IAV triggered by antigenic drift, annual vaccination is necessary, highlighting the need for a more broadly protective vaccine against IAV. The safety tested Modified Vaccinia virus Ankara (MVA) is licensed as a third-generation vaccine against smallpox and serves as a potent vector system for the development of new candidate vaccines against different pathogens. Here, we generated and characterized recombinant MVA candidate vaccines that deliver the highly conserved internal nucleoprotein (NP) of IAV under the transcriptional control of five newly designed chimeric poxviral promoters to further increase the immunogenic properties of the recombinant viruses (MVA-NP). Infections of avian cell cultures with the recombinant MVA-NPs demonstrated efficient synthesis of the IAV-NP which was expressed under the control of the five new promoters. Prime-boost or single shot immunizations in C57BL/6 mice readily induced circulating serum antibodies' binding to recombinant IAV-NP and the robust activation of IAV-NP-specific CD8+ T cell responses. Moreover, the MVA-NP candidate vaccines protected C57BL/6 mice against lethal respiratory infection with mouse-adapted IAV (A/Puerto Rico/8/1934/H1N1). Thus, further studies are warranted to evaluate the immunogenicity and efficacy of these recombinant MVA-NP vaccines in other IAV challenge models in more detail.
Collapse
Affiliation(s)
- Martin C Langenmayer
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 80539 Munich, Germany
| | | | - Lisa Marr
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
- Institute of Clinical Hygiene, Medical Microbiology and Infectiology, Paracelsus Medical University, Klinikum Nürnberg, 90419 Nuremberg, Germany
| | - Sylvia Jany
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
| | - Astrid Freudenstein
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
| | - Silvia Adam-Neumair
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
| | - Alina Tscherne
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
| | - Juan J Rojas
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
- Immunology Unit, Department of Pathology and Experimental Therapies, Faculty of Medicine and Health Sciences, University of Barcelona-Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain
| | - Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum Munich, 85764 Neuherberg, Germany
- Institute for Veterinary Pathology, LMU Munich, 80539 Munich, Germany
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 80539 Munich, Germany
| | - Asisa Volz
- Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- German Center of Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30559 Hannover, Germany
| |
Collapse
|
4
|
Ma J, Ji Q, Wang S, Qiu J, Liu Q. Identification and evaluation of a panel of strong constitutive promoters in Listeria monocytogenes for improving the expression of foreign antigens. Appl Microbiol Biotechnol 2021; 105:5135-5145. [PMID: 34086117 PMCID: PMC8175932 DOI: 10.1007/s00253-021-11374-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/10/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022]
Abstract
Attenuated Listeria monocytogenes could be a potential vaccine vector for the immunotherapy of tumors or pathogens. However, the lack of reliable promoters has limited its ability to express foreign antigens. In the present study, 21 promoters were identified from Listeria monocytogenes through RNA-seq analysis under two pH conditions of pH 7.4 and pH 5.5. Based on the constructed fluorescence report system, 7 constitutive promoters exhibited higher strength than Phelp (1.8-fold to 5.4-fold), a previously reported strong promoter. Furthermore, the selected 5 constitutive promoters exhibited higher UreB production activity than Phelp (1.1-fold to 8.3-fold). Notably, a well-characterized constitutive promoter P18 was found with the highest activity of fluorescence intensity and UreB production. In summary, the study provides a panel of strong constitutive promoters for Listeria monocytogenes and offers a theoretical basis for mining constitutive promoters in other organisms. KEY POINTS: • Twenty-one promoters were identified from L. monocytogenes through RNA-seq. • Fluorescent tracer of L. monocytogenes (P18) was performed in vitro and in vivo. • A well-characterized constitutive promoter P18 could improve the expression level of a foreign antigen UreB in L. monocytogenes.
Collapse
Affiliation(s)
- Junfei Ma
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qianyu Ji
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Shuying Wang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jingxuan Qiu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China. .,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
5
|
Samreen B, Tao S, Tischer K, Adler H, Drexler I. ORF6 and ORF61 Expressing MVA Vaccines Impair Early but Not Late Latency in Murine Gammaherpesvirus MHV-68 Infection. Front Immunol 2019; 10:2984. [PMID: 31921215 PMCID: PMC6930802 DOI: 10.3389/fimmu.2019.02984] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/05/2019] [Indexed: 01/02/2023] Open
Abstract
Gammaherpesviruses (γHV) are important pathogens causing persistent infections which lead to several malignancies in immunocompromised patients. Murine γHV 68 (MHV-68), a homolog to human EBV and KSHV, has been employed as a classical pathogen to investigate the molecular pathogenicity of γHV infections. γHV express distinct antigens during lytic or latent infection and antigen-specific T cells have a significant role in controlling the acute and latent viral infection, although the quality of anti-viral T cell responses required for protective immunity is not well-understood. We have generated recombinant modified vaccinia virus Ankara (recMVA) vaccines via MVA-BAC homologous recombination technology expressing MHV-68 ORF6 and ORF61 antigens encoding both MHC class I and II-restricted epitopes. After vaccination, we examined T cell responses before and after MHV-68 infection to determine their involvement in latent virus control. We show recognition of recMVA- and MHV-68-infected APC by ORF6 and ORF61 epitope-specific T cell lines in vitro. The recMVA vaccines efficiently induced MHV-68-specific CD8+ and CD4+ T cell responses after a single immunization and more pronounced after homologous prime/boost vaccination in mice. Moreover, we exhibit protective capacity of prophylactic recMVA vaccination during early latency at day 17 after intranasal challenge with MHV-68, but failed to protect from latency at day 45. Further T cell analysis indicated that T cell exhaustion was not responsible for the lack of protection by recMVA vaccination in long-term latency at day 45. The data support further efforts aiming at improved vaccine development against γHV infections with special focus on targeting protective CD4+ T cell responses.
Collapse
Affiliation(s)
- Baila Samreen
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany.,Department of Oncology-Pathology, Science for Life Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Sha Tao
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karsten Tischer
- Fachbereich Veterinärmedizin, Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Heiko Adler
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Ingo Drexler
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
6
|
Tao S, Tao R, Busch DH, Widera M, Schaal H, Drexler I. Sequestration of Late Antigens Within Viral Factories Impairs MVA Vector-Induced Protective Memory CTL Responses. Front Immunol 2019; 10:2850. [PMID: 31867011 PMCID: PMC6904312 DOI: 10.3389/fimmu.2019.02850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/20/2019] [Indexed: 01/22/2023] Open
Abstract
Cytotoxic CD8+ T cell (CTL) responses play an essential role in antiviral immunity. Here, we focused on the activation of CTL which recognize epitopes derived from viral or recombinant antigens with either early or late expression kinetics after infection with Modified Vaccinia Virus Ankara (MVA). Late antigens but not early antigens failed to efficiently stimulate murine CTL lines in vitro and were unable to activate and expand protective memory T cell responses in mice in vivo. The reduced or absent presentation of late antigens was not due to impaired antigen presentation or delayed protein synthesis, but was caused by sequestration of late antigens within viral factories (VFs). Additionally, the trapping of late antigens in VFs conflicts with antigen processing and presentation as proteasomal activity was strongly reduced or absent in VFs, suggesting inefficient antigen degradation. This study gives for the first time a mechanistic explanation for the weak immunogenicity of late viral antigens for memory CTL activation. Since MVA is preferentially used as a boost vector in heterologous prime/boost vaccinations, this is an important information for future vaccine design.
Collapse
Affiliation(s)
- Sha Tao
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ronny Tao
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Dirk H Busch
- Institute of Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | - Marek Widera
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Heiner Schaal
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ingo Drexler
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
7
|
Genomic Characterization of Orf Virus Strain D1701-V ( Parapoxvirus) and Development of Novel Sites for Multiple Transgene Expression. Viruses 2019; 11:v11020127. [PMID: 30704093 PMCID: PMC6409557 DOI: 10.3390/v11020127] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
The Orf virus (ORFV; Parapoxvirus) strain D1701 with an attenuated phenotype and excellent immunogenic capacity is successfully used for the generation of recombinant vaccines against different viral infections. Adaption for growth in Vero cells was accompanied by additional major genomic changes resulting in ORFV strain variant D1701-V. In this study, restriction enzyme mapping, blot hybridization and DNA sequencing of the deleted region s (A, AT and D) in comparison to the predecessor strain D1701-B revealed the loss of 7 open reading frames (ORF008, ORF101, ORF102, ORF114, ORF115, ORF116, ORF117). The suitability of deletion site D for expression of foreign genes is demonstrated using novel synthetic early promoter eP1 and eP2. Comparison of promoter strength showed that the original vegf-e promoter Pv as well as promoter eP2 display an up to 11-fold stronger expression than promoter eP1, irrespective of the insertion site. Successful integration and expression of the fluorescent marker genes is demonstrated by gene- and insertion-site specific PCR assays, fluorescence microscopy and flow cytometry. For the first time ORFV recombinants are generated simultaneously expressing transgenes in two different insertion loci. That allows production of polyvalent vaccines containing several antigens against one or different pathogens in a single vectored ORFV vaccine.
Collapse
|
8
|
Marr L, Lülf AT, Freudenstein A, Sutter G, Volz A. Myristoylation increases the CD8+T-cell response to a GFP prototype antigen delivered by modified vaccinia virus Ankara. J Gen Virol 2016; 97:934-940. [PMID: 26864442 DOI: 10.1099/jgv.0.000425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activation of CD8(+)T-cells is an essential part of immune responses elicited by recombinant modified vaccinia virus Ankara (MVA). Strategies to enhance T-cell responses to antigens may be particularly necessary for broadly protective immunization against influenza A virus infections or for candidate vaccines targeting chronic infections and cancer. Here, we tested recombinant MVAs that targeted a model antigen, GFP, to different localizations in infected cells. In vitro characterization demonstrated that GFP accumulated in the nucleus (MVA-nls-GFP), associated with cellular membranes (MVA-myr-GFP) or was equally distributed throughout the cell (MVA-GFP). On vaccination, we found significantly higher levels of GFP-specific CD8(+)T-cells in MVA-myr-GFP-vaccinated BALB/c mice than in those immunized with MVA-GFP or MVA-nls-GFP. Thus, myristoyl modification may be a useful strategy to enhance CD8(+)T-cell responses to MVA-delivered target antigens.
Collapse
Affiliation(s)
- Lisa Marr
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539, Munich, Germany
| | - Anna-Theresa Lülf
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539, Munich, Germany
| | - Astrid Freudenstein
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539, Munich, Germany
| | - Gerd Sutter
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539, Munich, Germany
| | - Asisa Volz
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539, Munich, Germany
| |
Collapse
|