date: 2024-07-09T08:03:33Z pdf:unmappedUnicodeCharsPerPage: 0 pdf:PDFVersion: 1.7 pdf:docinfo:title: Parallel Multifactorial Process Optimization and Intensification for High-Yield Production of Live YF17D-Vectored Zika Vaccine xmp:CreatorTool: LaTeX with hyperref Keywords: live-attenuated vaccine; process intensification; virus yield; high cell density; suspension cells; YF17D; Zika vaccine access_permission:modify_annotations: true access_permission:can_print_degraded: true subject: The live-attenuated yellow fever 17D strain is a potent vaccine and viral vector. Its manufacture is based on embryonated chicken eggs or adherent Vero cells. Both processes are unsuitable for rapid and scalable supply. Here, we introduce a high-throughput workflow to identify suspension cells that are fit for the high-yield production of live YF17D-based vaccines in an intensified upstream process. The use of an automated parallel ambr15 microbioreactor system for screening and process optimization has led to the identification of two promising cell lines (AGE1.CR.pIX and HEKDyn) and the establishment of optimized production conditions, which have resulted in a >100-fold increase in virus titers compared to the current state of the art using adherent Vero cells. The process can readily be scaled up from the microbioreactor scale (15 mL) to 1 L stirred tank bioreactors. The viruses produced are genetically stable and maintain their favorable safety and immunogenicity profile, as demonstrated by the absence of neurovirulence in suckling BALB/c mice and consistent seroprotection in AG129 mice. In conclusion, the presented workflow allows for the rapid establishment of a robust, scalable, and high-yield process for the production of live-attenuated orthoflavivirus vaccines, which outperforms current standards. The approach described here can serve as a model for the development of scalable processes and the optimization of yields for other virus-based vaccines that face challenges in meeting growing demands. dc:creator: Sven Göbel, Ozeir Kazemi, Ji Ma, Ingo Jordan, Volker Sandig, Jasmine Paulissen, Winnie Kerstens, Hendrik Jan Thibaut, Udo Reichl, Kai Dallmeier and Yvonne Genzel dcterms:created: 2024-07-09T07:45:18Z Last-Modified: 2024-07-09T08:03:33Z dcterms:modified: 2024-07-09T08:03:33Z dc:format: application/pdf; version=1.7 title: Parallel Multifactorial Process Optimization and Intensification for High-Yield Production of Live YF17D-Vectored Zika Vaccine Last-Save-Date: 2024-07-09T08:03:33Z pdf:docinfo:creator_tool: LaTeX with hyperref access_permission:fill_in_form: true pdf:docinfo:keywords: live-attenuated vaccine; process intensification; virus yield; high cell density; suspension cells; YF17D; Zika vaccine pdf:docinfo:modified: 2024-07-09T08:03:33Z meta:save-date: 2024-07-09T08:03:33Z pdf:encrypted: false dc:title: Parallel Multifactorial Process Optimization and Intensification for High-Yield Production of Live YF17D-Vectored Zika Vaccine modified: 2024-07-09T08:03:33Z cp:subject: The live-attenuated yellow fever 17D strain is a potent vaccine and viral vector. Its manufacture is based on embryonated chicken eggs or adherent Vero cells. Both processes are unsuitable for rapid and scalable supply. Here, we introduce a high-throughput workflow to identify suspension cells that are fit for the high-yield production of live YF17D-based vaccines in an intensified upstream process. The use of an automated parallel ambr15 microbioreactor system for screening and process optimization has led to the identification of two promising cell lines (AGE1.CR.pIX and HEKDyn) and the establishment of optimized production conditions, which have resulted in a >100-fold increase in virus titers compared to the current state of the art using adherent Vero cells. The process can readily be scaled up from the microbioreactor scale (15 mL) to 1 L stirred tank bioreactors. The viruses produced are genetically stable and maintain their favorable safety and immunogenicity profile, as demonstrated by the absence of neurovirulence in suckling BALB/c mice and consistent seroprotection in AG129 mice. In conclusion, the presented workflow allows for the rapid establishment of a robust, scalable, and high-yield process for the production of live-attenuated orthoflavivirus vaccines, which outperforms current standards. The approach described here can serve as a model for the development of scalable processes and the optimization of yields for other virus-based vaccines that face challenges in meeting growing demands. pdf:docinfo:subject: The live-attenuated yellow fever 17D strain is a potent vaccine and viral vector. Its manufacture is based on embryonated chicken eggs or adherent Vero cells. Both processes are unsuitable for rapid and scalable supply. Here, we introduce a high-throughput workflow to identify suspension cells that are fit for the high-yield production of live YF17D-based vaccines in an intensified upstream process. The use of an automated parallel ambr15 microbioreactor system for screening and process optimization has led to the identification of two promising cell lines (AGE1.CR.pIX and HEKDyn) and the establishment of optimized production conditions, which have resulted in a >100-fold increase in virus titers compared to the current state of the art using adherent Vero cells. The process can readily be scaled up from the microbioreactor scale (15 mL) to 1 L stirred tank bioreactors. The viruses produced are genetically stable and maintain their favorable safety and immunogenicity profile, as demonstrated by the absence of neurovirulence in suckling BALB/c mice and consistent seroprotection in AG129 mice. In conclusion, the presented workflow allows for the rapid establishment of a robust, scalable, and high-yield process for the production of live-attenuated orthoflavivirus vaccines, which outperforms current standards. The approach described here can serve as a model for the development of scalable processes and the optimization of yields for other virus-based vaccines that face challenges in meeting growing demands. Content-Type: application/pdf pdf:docinfo:creator: Sven Göbel, Ozeir Kazemi, Ji Ma, Ingo Jordan, Volker Sandig, Jasmine Paulissen, Winnie Kerstens, Hendrik Jan Thibaut, Udo Reichl, Kai Dallmeier and Yvonne Genzel X-Parsed-By: org.apache.tika.parser.DefaultParser creator: Sven Göbel, Ozeir Kazemi, Ji Ma, Ingo Jordan, Volker Sandig, Jasmine Paulissen, Winnie Kerstens, Hendrik Jan Thibaut, Udo Reichl, Kai Dallmeier and Yvonne Genzel meta:author: Sven Göbel, Ozeir Kazemi, Ji Ma, Ingo Jordan, Volker Sandig, Jasmine Paulissen, Winnie Kerstens, Hendrik Jan Thibaut, Udo Reichl, Kai Dallmeier and Yvonne Genzel dc:subject: live-attenuated vaccine; process intensification; virus yield; high cell density; suspension cells; YF17D; Zika vaccine meta:creation-date: 2024-07-09T07:45:18Z created: 2024-07-09T07:45:18Z access_permission:extract_for_accessibility: true access_permission:assemble_document: true xmpTPg:NPages: 23 Creation-Date: 2024-07-09T07:45:18Z pdf:charsPerPage: 4000 access_permission:extract_content: true access_permission:can_print: true meta:keyword: live-attenuated vaccine; process intensification; virus yield; high cell density; suspension cells; YF17D; Zika vaccine Author: Sven Göbel, Ozeir Kazemi, Ji Ma, Ingo Jordan, Volker Sandig, Jasmine Paulissen, Winnie Kerstens, Hendrik Jan Thibaut, Udo Reichl, Kai Dallmeier and Yvonne Genzel producer: pdfTeX-1.40.25 access_permission:can_modify: true pdf:docinfo:producer: pdfTeX-1.40.25 pdf:docinfo:created: 2024-07-09T07:45:18Z