1
|
Lapras B, Marchand C, Merienne C, Medina M, Kolenda C, Laurent F, Pirot F. Rationalisation of the purification process for a phage active pharmaceutical ingredient. Eur J Pharm Biopharm 2024; 203:114438. [PMID: 39111580 DOI: 10.1016/j.ejpb.2024.114438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024]
Abstract
The resurgence of phage therapy, once abandoned in the early 20th century in part due to issues related to the purification process and stability, is spurred by the global threat of antibiotic resistance. Engineering advances have enabled more precise separation unit operations, improving overall purification efficiency. The present review discusses the physicochemical properties of impurities commonly found in a phage lysate, e.g., contaminants, phage-related impurities, and propagation-related impurities. Differences in phages and bacterial impurities properties are leveraged to elaborate a four-step phage purification process: clarification, capture and concentration, subsequent purification and polishing. Ultimately, a framework for rationalising the development of a purification process is proposed, considering three operational characteristics, i.e., scalability, transferability to various phages and duration. This guide facilitates the preselection of a sequence of unit operations, which can then be confronted with the expected impurities to validate the theoretical capacity of the process to purify the phage lysate.
Collapse
Affiliation(s)
- B Lapras
- Hospices Civils de Lyon, Edouard Herriot Hospital, Pharmacy Department, FRIPHARM®, F-69437 Lyon, France; Claude Bernard Lyon 1 University, French National Centre for Scientific Research (CNRS), Institut de Biologie et de Chimie des Protéines (IBCP), Tissue Biology and Therapeutic Engineering Laboratory (LBTI), UMR 5305, F-69007 Lyon, France.
| | - C Marchand
- Hospices Civils de Lyon, Edouard Herriot Hospital, Pharmacy Department, FRIPHARM®, F-69437 Lyon, France
| | - C Merienne
- Hospices Civils de Lyon, Edouard Herriot Hospital, Pharmacy Department, FRIPHARM®, F-69437 Lyon, France
| | - M Medina
- Hospices Civils de Lyon, Croix Rousse Hospital, Bacteriology Department, French National Reference Centre for Staphylococci, F-69317 Lyon, France; Claude Bernard Lyon 1 University, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR 5308, F- 69365 Lyon, France
| | - C Kolenda
- Hospices Civils de Lyon, Croix Rousse Hospital, Bacteriology Department, French National Reference Centre for Staphylococci, F-69317 Lyon, France; Claude Bernard Lyon 1 University, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR 5308, F- 69365 Lyon, France
| | - F Laurent
- Hospices Civils de Lyon, Croix Rousse Hospital, Bacteriology Department, French National Reference Centre for Staphylococci, F-69317 Lyon, France; Claude Bernard Lyon 1 University, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR 5308, F- 69365 Lyon, France
| | - F Pirot
- Hospices Civils de Lyon, Edouard Herriot Hospital, Pharmacy Department, FRIPHARM®, F-69437 Lyon, France; Claude Bernard Lyon 1 University, French National Centre for Scientific Research (CNRS), Institut de Biologie et de Chimie des Protéines (IBCP), Tissue Biology and Therapeutic Engineering Laboratory (LBTI), UMR 5305, F-69007 Lyon, France
| |
Collapse
|
2
|
Pagallies F, Labisch JJ, Wronska M, Pflanz K, Amann R. Efficient and scalable clarification of Orf virus from HEK suspension for vaccine development. Vaccine X 2024; 18:100474. [PMID: 38523620 PMCID: PMC10958475 DOI: 10.1016/j.jvacx.2024.100474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
The Orf virus (ORFV) is a promising vector platform for the generation of vaccines against infectious diseases and cancer, highlighted by its progression to clinical testing phases. One of the critical steps during GMP manufacturing is the clarification of crude harvest because of the enveloped nature and large size of ORFV. This study presents the first description of ORFV clarification process from a HEK suspension batch process. We examined various filter materials, membrane pore sizes, harvest timings, and nuclease treatments. Employing the Ambr® crossflow system for high-throughput, small-volume experiments, we identified polypropylene-based Sartopure® PP3 filters as ideal. These filters, used in two consecutive stages with reducing pore sizes, significantly enhanced ORFV recovery and addressed scalability challenges. Moreover, we demonstrated that the time of harvest and the use of a nuclease play a decisive role to increase ORFV yields. With these findings, we were able to establish an efficient and scalable clarification process of ORFV derived from a suspension production process, essential for advancing ORFV vaccine manufacturing.
Collapse
Affiliation(s)
- Felix Pagallies
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Jennifer J. Labisch
- Lab Essentials Applications Development, Sartorius, Otto-Brenner-Straße 20, 37079 Göttingen, Germany
| | - Malgorzata Wronska
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- PRiME Vector Technologies, Herrenberger Straße 24, 72070 Tübingen, Germany
| | - Karl Pflanz
- Lab Essentials Applications Development, Sartorius, Otto-Brenner-Straße 20, 37079 Göttingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- PRiME Vector Technologies, Herrenberger Straße 24, 72070 Tübingen, Germany
| |
Collapse
|
3
|
Li F, Liu B, Xiong Y, Zhang Z, Zhang Q, Qiu R, Peng F, Nian X, Wu D, Li X, Liu J, Li Z, Tu H, Wu W, Wang Y, Zhang J, Yang X. Enhanced Downstream Processing for a Cell-Based Avian Influenza (H5N1) Vaccine. Vaccines (Basel) 2024; 12:138. [PMID: 38400122 PMCID: PMC10891636 DOI: 10.3390/vaccines12020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
H5N1 highly pathogenic avian influenza virus (HPAIV) infections pose a significant threat to human health, with a mortality rate of around 50%. Limited global approval of H5N1 HPAIV vaccines, excluding China, prompted the need to address safety concerns related to MDCK cell tumorigenicity. Our objective was to improve vaccine safety by minimizing residual DNA and host cell protein (HCP). We developed a downstream processing method for the cell-based H5N1 HPAIV vaccine, employing CaptoTM Core 700, a multimodal resin, for polishing. Hydrophobic-interaction chromatography (HIC) with polypropylene glycol as a functional group facilitated the reversible binding of virus particles for capture. Following the two-step chromatographic process, virus recovery reached 68.16%. Additionally, HCP and DNA levels were reduced to 2112.60 ng/mL and 6.4 ng/mL, respectively. Western blot, high-performance liquid chromatography (HPLC), and transmission electron microscopy (TEM) confirmed the presence of the required antigen with a spherical shape and appropriate particle size. Overall, our presented two-step downstream process demonstrates potential as an efficient and cost-effective platform technology for cell-based influenza (H5N1 HPAIV) vaccines.
Collapse
Affiliation(s)
- Fang Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Bo Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Yu Xiong
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zhegang Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Qingmei Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Ran Qiu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Feixia Peng
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Dongping Wu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuedan Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Jing Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Ze Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Hao Tu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Wenyi Wu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Yu Wang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
- China National Biotec Group Company Limited, Beijing 100029, China
| |
Collapse
|
4
|
Zhou X, Gao M, De X, Sun T, Bai Z, Luo J, Wang F, Ge J. Bacterium-like particles derived from probiotics: progress, challenges and prospects. Front Immunol 2023; 14:1263586. [PMID: 37868963 PMCID: PMC10587609 DOI: 10.3389/fimmu.2023.1263586] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Bacterium-like particles (BLPs) are hollow peptidoglycan particles obtained from food-grade Lactococcus lactis inactivated by hot acid. With the advantage of easy preparation, high safety, great stability, high loading capacity, and high mucosal delivery efficiency, BLPs can load and display proteins on the surface with the help of protein anchor (PA), making BLPs a proper delivery system. Owning to these features, BLPs are widely used in the development of adjuvants, vaccine carriers, virus/antigens purification, and enzyme immobilization. This review has attempted to gather a full understanding of the technical composition, characteristics, applications. The mechanism by which BLPs induces superior adaptive immune responses is also discussed. Besides, this review tracked the latest developments in the field of BLPs, including Lactobacillus-derived BLPs and novel anchors. Finally, the main limitations and proposed breakthrough points to further enhance the immunogenicity of BLPs vaccines were discussed, providing directions for future research. We hope that further developments in the field of antigen delivery of subunit vaccines or others will benefit from BLPs.
Collapse
Affiliation(s)
- Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingchun Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tong Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhikun Bai
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, China
| | - Jilong Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Fang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin, China
| |
Collapse
|
5
|
Amruta A, Iannotta D, Cheetham SW, Lammers T, Wolfram J. Vasculature organotropism in drug delivery. Adv Drug Deliv Rev 2023; 201:115054. [PMID: 37591370 PMCID: PMC10693934 DOI: 10.1016/j.addr.2023.115054] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/22/2023] [Accepted: 08/13/2023] [Indexed: 08/19/2023]
Abstract
Over the past decades, there has been an exponential increase in the development of preclinical and clinical nanodelivery systems, and recently, an accelerating demand to deliver RNA and protein-based therapeutics. Organ-specific vasculature provides a promising intermediary for site-specific delivery of nanoparticles and extracellular vesicles to interstitial cells. Endothelial cells express organ-specific surface marker repertoires that can be used for targeted delivery. This article highlights organ-specific vasculature properties, nanodelivery strategies that exploit vasculature organotropism, and overlooked challenges and opportunities in targeting and simultaneously overcoming the endothelial barrier. Impediments in the clinical translation of vasculature organotropism in drug delivery are also discussed.
Collapse
Affiliation(s)
- A Amruta
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dalila Iannotta
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Seth W Cheetham
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO-ABCD), 52074 Aachen, Germany
| | - Joy Wolfram
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Lothert K, Harsy YMJ, Endres P, Müller E, Wolff MW. Evaluation of restricted access media for the purification of cell culture-derived Orf viruses. Eng Life Sci 2023; 23:e2300009. [PMID: 37664009 PMCID: PMC10472920 DOI: 10.1002/elsc.202300009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 09/05/2023] Open
Abstract
Recently, multimodal chromatography using restricted access media (RAM) for the purification of nanoparticles, such as viruses has regained increasing attention. These chromatography resins combine size exclusion on the particle shell and adsorptive interaction within the core. Accordingly, smaller process-related impurities, for example, DNA and proteins, can be retained, while larger product viruses can pass unhindered. We evaluated a range of currently available RAM, differing in the shells' pore cut-off and the core chemistry, for the purification of a cell culture-derived clarified model virus, namely the Orf virus (ORFV). We examined impurity depletion and product recovery as relevant criteria for the evaluation of column performance, as well as scale-up robustness and regeneration potential for evaluating a multiple use application. The results indicate that some columns, for example, the Capto Core, enable both a high DNA and protein removal, while others, for example, the Monomix Core 60 (MC60), are more suitable for DNA depletion. Furthermore, column regeneration is facilitated by using columns with larger shell pores (5000 vs. 700 kDa) and weaker binding interactions (anion exchange vs. multimodal). According to these findings, the choice of RAM resins should be selected according to the respective feed sample composition and the planned number of application cycles.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences Mittelhessen (THM)GiessenGermany
| | - Yasmina M. J. Harsy
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences Mittelhessen (THM)GiessenGermany
| | - Patrick Endres
- Tosoh Bioscience GmbH, Separations Business Unit ‐ EuropeGriesheimGermany
| | - Egbert Müller
- Tosoh Bioscience GmbH, Separations Business Unit ‐ EuropeGriesheimGermany
| | - Michael W. Wolff
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences Mittelhessen (THM)GiessenGermany
| |
Collapse
|
7
|
Eilts F, Labisch JJ, Orbay S, Harsy YMJ, Steger M, Pagallies F, Amann R, Pflanz K, Wolff MW. Stability studies for the identification of critical process parameters for a pharmaceutical production of the Orf virus. Vaccine 2023:S0264-410X(23)00722-3. [PMID: 37353451 DOI: 10.1016/j.vaccine.2023.06.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
A promising new vaccine platform is based on the Orf virus, a viral vector of the genus Parapoxvirus, which is currently being tested in phase I clinical trials. The application as a vaccine platform mandates a well-characterised, robust, and efficient production process. To identify critical process parameters in the production process affecting the virus' infectivity, the Orf virus was subjected to forced degradation studies, including thermal, pH, chemical, and mechanical stress conditions. The tests indicated a robust virus infectivity within a pH range of 5-7.4 and in the presence of the tested buffering substances (TRIS, HEPES, PBS). The ionic strength up to 0.5 M had no influence on the Orf virus' infectivity stability for NaCl and MgCl2, while NH4Cl destabilized significantly. Furthermore, short-term thermal stress of 2d up to 37 °C and repeated freeze-thaw cycles (20cycles) did not affect the virus' infectivity. The addition of recombinant human serum albumin was found to reduce virus inactivation. Last, the Orf virus showed a low shear sensitivity induced by peristaltic pumps and mixing, but was sensitive to ultrasonication. The isoelectric point of the applied Orf virus genotype D1707-V was determined at pH3.5. The broad picture of the Orf virus' infectivity stability against environmental parameters is an important contribution for the identification of critical process parameters for the production process, and supports the development of a stable pharmaceutical formulation. The work is specifically relevant for enveloped (large DNA) viruses, like the Orf virus and like most vectored vaccine approaches.
Collapse
Affiliation(s)
- Friederike Eilts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Jennifer J Labisch
- Lab Essentials Applications Development, Sartorius Stedim Biotech GmbH, August-Spindler-Straße 11, 37079 Goettingen, Germany; Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Lower Saxony, Germany
| | - Sabri Orbay
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Yasmina M J Harsy
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Marleen Steger
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15/3.008, 72076 Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15/3.008, 72076 Tuebingen, Germany; Prime Vector Technologies, Herrenberger Straße 24, 72070 Tuebingen, Germany
| | - Karl Pflanz
- Lab Essentials Applications Development, Sartorius Stedim Biotech GmbH, August-Spindler-Straße 11, 37079 Goettingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany.
| |
Collapse
|
8
|
Hillebrandt N, Hubbuch J. Size-selective downstream processing of virus particles and non-enveloped virus-like particles. Front Bioeng Biotechnol 2023; 11:1192050. [PMID: 37304136 PMCID: PMC10248422 DOI: 10.3389/fbioe.2023.1192050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Non-enveloped virus-like particles (VLPs) are versatile protein nanoparticles with great potential for biopharmaceutical applications. However, conventional protein downstream processing (DSP) and platform processes are often not easily applicable due to the large size of VLPs and virus particles (VPs) in general. The application of size-selective separation techniques offers to exploit the size difference between VPs and common host-cell impurities. Moreover, size-selective separation techniques offer the potential for wide applicability across different VPs. In this work, basic principles and applications of size-selective separation techniques are reviewed to highlight their potential in DSP of VPs. Finally, specific DSP steps for non-enveloped VLPs and their subunits are reviewed as well as the potential applications and benefits of size-selective separation techniques are shown.
Collapse
Affiliation(s)
| | - Jürgen Hubbuch
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
9
|
Lothert K, Bagrin E, Wolff MW. Evaluating Novel Quantification Methods for Infectious Baculoviruses. Viruses 2023; 15:v15040998. [PMID: 37112978 PMCID: PMC10141099 DOI: 10.3390/v15040998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Accurate and rapid quantification of (infectious) virus titers is of paramount importance in the manufacture of viral vectors and vaccines. Reliable quantification data allow efficient process development at a laboratory scale and thorough process monitoring in later production. However, current gold standard applications, such as endpoint dilution assays, are cumbersome and do not provide true process analytical monitoring. Accordingly, flow cytometry and quantitative polymerase chain reaction have attracted increasing interest in recent years, offering various advantages for rapid quantification. Here, we compared different approaches for the assessment of infectious viruses, using a model baculovirus. Firstly, infectivity was estimated by the quantification of viral nucleic acids in infected cells, and secondly, different flow cytometric approaches were investigated regarding analysis times and calibration ranges. The flow cytometry technique included a quantification based on post-infection fluorophore expression and labeling of a viral surface protein using fluorescent antibodies. Additionally, the possibility of viral (m)RNA labeling in infected cells was investigated as a proof of concept. The results confirmed that infectivity assessment based on qPCR is not trivial and requires sophisticated method optimization, whereas staining of viral surface proteins is a fast and feasible approach for enveloped viruses. Finally, labeling of viral (m)RNA in infected cells appears to be a promising opportunity but will require further research.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Department Life Science Engineering, University of Applied Sciences Mittelhessen (THM), 35390 Giessen, Germany
| | - Elena Bagrin
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Department Life Science Engineering, University of Applied Sciences Mittelhessen (THM), 35390 Giessen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Department Life Science Engineering, University of Applied Sciences Mittelhessen (THM), 35390 Giessen, Germany
| |
Collapse
|
10
|
Higuchi S, Satou T, Uchida Y. Hand-held virus concentration method using a hollow fiber filter module. MethodsX 2023; 10:102126. [PMID: 36970019 PMCID: PMC10036919 DOI: 10.1016/j.mex.2023.102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023] Open
Abstract
A virus concentration method is required for viral vaccine manufacturing and virus-related research. However, concentration methods, such as ultracentrifugation, often require capital investment. We report a simple and easy-to-use handheld syringe method for virus concentration using a hollow fiber (HF) filter module, which can be applicable to viruses of different sizes, without incorporating any special machines or reagents. This virus concentration method does not use pumps, which might cause shear stress for virus particles; therefore, it is useful for stress-sensitive virus particles, and virus-like particles, as well as other proteins. The clarified harvest of flavivirus (Zika virus) was concentrated using an HF filter module and compared with a centrifugal ultrafiltration device (CUD) for demonstration of the HF filter method. The HF filter method achieved concentration of the virus solution in less time than the CUD. The yield comparison of the recovered virus solution indicated that recovery from the developed method was comparable to using the CUD, and infectivity was maintained throughout.•The Zika virus was concentrated from 200 mL to 5 mL within 45 min using the HF filter and handheld syringe module method.•The handheld HF filter method may be applicable to stress-sensitive viruses and proteins of different sizes.•The virus concentration process should be conducted in a safety cabinet, which is preferred for virus containment.
Collapse
|
11
|
Canova CT, Inguva PK, Braatz RD. Mechanistic modeling of viral particle production. Biotechnol Bioeng 2023; 120:629-641. [PMID: 36461898 DOI: 10.1002/bit.28296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022]
Abstract
Viral systems such as wild-type viruses, viral vectors, and virus-like particles are essential components of modern biotechnology and medicine. Despite their importance, the commercial-scale production of viral systems remains highly inefficient for multiple reasons. Computational strategies are a promising avenue for improving process development, optimization, and control, but require a mathematical description of the system. This article reviews mechanistic modeling strategies for the production of viral particles, both at the cellular and bioreactor scales. In many cases, techniques and models from adjacent fields such as epidemiology and wild-type viral infection kinetics can be adapted to construct a suitable process model. These process models can then be employed for various purposes such as in-silico testing of novel process operating strategies and/or advanced process control.
Collapse
Affiliation(s)
- Christopher T Canova
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Pavan K Inguva
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard D Braatz
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
12
|
Meierrieks F, Pickl A, Wolff MW. A robust and efficient alluvial filtration method for the clarification of adeno-associated viruses from crude cell lysates. J Biotechnol 2023; 367:31-41. [PMID: 36997072 DOI: 10.1016/j.jbiotec.2023.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/01/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Adeno-associated virus (AAV) based vectors have recently been gaining importance as DNA delivery systems. Efficient downstream processing of AAV remains a major challenge as serotypes differ in physicochemical properties, making it difficult to design uniform purification processes. Clarification of AAV is an especially critical step. Harvesting of AAV, like other viruses, often requires cell lysis, resulting in a difficult-to-filter cell lysate. In this study, we evaluated the applicability of diatomaceous earth (DE) as a filter aid for clarification of AAV crude cell lysates. DE filtration proved to be a viable clarification method for AAV2, AAV5 and AAV8. Based on a design of experiment approach, the DE concentration was identified as the main factor influencing AAV particle loss. The loss of AAV during DE filtration was limited to <2% by maintaining the DE quantity below 0.181mg DE/1010 AAV. Use of DE reduced manual handling time 3-fold and increased the filter capacity 3.5-fold compared to filtration combined with a prior centrifugation step. Moreover, we showed that the DE type had only a minor influence on the filtration performance. This study demonstrated that filtration with DE as a filter aid is an efficient clarification method for different AAV serotypes.
Collapse
|
13
|
Lauro ML, Bowman AM, Smith JP, Gaye SN, Acevedo-Skrip J, DePhillips PA, Loughney JW. Overcoming Biopharmaceutical Interferents for Quantitation of Host Cell DNA Using an Automated, High-Throughput Methodology. AAPS J 2022; 25:10. [PMID: 36482268 PMCID: PMC9735023 DOI: 10.1208/s12248-022-00764-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/21/2022] [Indexed: 12/13/2022] Open
Abstract
The rapid development of biologics and vaccines in response to the current pandemic has highlighted the need for robust platform assays to characterize diverse biopharmaceuticals. A critical aspect of biopharmaceutical development is achieving a highly pure product, especially with respect to residual host cell material. Specifically, two important host cell impurities of focus within biopharmaceuticals are residual DNA and protein. In this work, a novel high-throughput host cell DNA quantitation assay was developed for rapid screening of complex vaccine drug substance samples. The developed assay utilizes the commercially available, fluorescent-sensitive Picogreen dye within a 96-well plate configuration to allow for a cost effective and rapid analysis. The assay was applied to in-process biopharmaceutical samples with known interferences to the dye, including RNA and protein. An enzymatic digestion pre-treatment was found to overcome these interferences and thus allow this method to be applied to wide-ranging, diverse analyses. In addition, the use of deoxycholate in the digestion treatment allowed for disruption of interactions in a given sample matrix in order to more accurately and selectively quantitate DNA. Critical analytical figures of merit for assay performance, such as precision and spike recovery, were evaluated and successfully demonstrated. This new analytical method can thus be successfully applied to both upstream and downstream process analysis for biologics and vaccines using an innovative and automated high-throughput approach.
Collapse
Affiliation(s)
- Mackenzie L. Lauro
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| | - Amy M. Bowman
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| | - Joseph P. Smith
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| | - Susannah N. Gaye
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| | - Jillian Acevedo-Skrip
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| | - Pete A. DePhillips
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| | - John W. Loughney
- grid.417993.10000 0001 2260 0793Analytical Research & Development, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486 USA
| |
Collapse
|
14
|
Sonugür FG, Babahan C, Abdi Abgarmi S, Akbulut H. Incubation Temperature and Period During Denarase Treatment and Microfiltration Affect the Yield of Recombinant Adenoviral Vectors During Downstream Processing. Mol Biotechnol 2022:10.1007/s12033-022-00616-8. [PMID: 36451062 PMCID: PMC9713150 DOI: 10.1007/s12033-022-00616-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Adenoviral vectors (AV) are commonly used as vaccine and gene therapy vehicles because of their ease of construction, ability to grow to high titers in the large-scale production process, and safety for human applications. However, the efficiency rate of downstream processes for adenoviral vectors still varies greatly. In the current study, we aimed to investigate the effect of the downstream treatment protocol and microfiltration of the harvested upstream material on viral vector yield. We compared the performance of the repeated freeze-thaw (RFT) and the Tween-20 detergent lysis (DLT) methods. In addition, the effects of the cell lysis method, incubation temperature, and time on viral yield were investigated. The samples were incubated at either room temperature or 37 °C for 1-, 2-, and 4-h periods. Samples were filtered with PES and SFCA membrane. Virus yield and infectivity were assayed by qPCR and immuno-titration. In conclusion, our results suggest that 2-h incubation gives the best results when incubated at 37 °C for denarase activity when Tween-20 is used for virus recovery. If the room temperature is preferred, 4-h incubation could be preferred. A phase 1 clinical trial (NCT05526183, January 21, 2022) was started with the recombinant adenovirus used in the study.
Collapse
Affiliation(s)
- Fatma Gizem Sonugür
- Department of Tumor Biology, Cancer Research Institute, Ankara University, Ankara, Turkey
| | - Cansu Babahan
- Department of Tumor Biology, Cancer Research Institute, Ankara University, Ankara, Turkey
| | - Samira Abdi Abgarmi
- Department of Tumor Biology, Cancer Research Institute, Ankara University, Ankara, Turkey
| | - Hakan Akbulut
- Department of Tumor Biology, Cancer Research Institute, Ankara University, Ankara, Turkey ,Department of Medical Oncology, School of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
15
|
Marchel M, Marrucho IM. Application of Aqueous Biphasic Systems Extraction in Various Biomolecules Separation and Purification: Advancements Brought by Quaternary Systems. SEPARATION & PURIFICATION REVIEWS 2022. [DOI: 10.1080/15422119.2022.2136574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mateusz Marchel
- Faculty of Chemistry, Department of Process Engineering and Chemical Technology, Gdansk University of Technology, Gdansk, Poland
- Centro de Química Estrutural and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Isabel M. Marrucho
- Centro de Química Estrutural and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
16
|
Eilts F, Lothert K, Orbay S, Pagallies F, Amann R, Wolff MW. A Summary of Practical Considerations for the Application of the Steric Exclusion Chromatography for the Purification of the Orf Viral Vector. MEMBRANES 2022; 12:1070. [PMID: 36363625 PMCID: PMC9696199 DOI: 10.3390/membranes12111070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Steric exclusion chromatography (SXC) is a promising purification method for biological macromolecules such as the Orf virus (ORFV) vector. The method's principle is closely related to conventional polyethylene glycol (PEG) precipitation, repeatedly implementing membranes as porous chromatographic media. In the past decade, several purification tasks with SXC showed exceptionally high yields and a high impurity removal. However, the effect of varying process parameters, on the precipitation success and its limitations to SXC, is not yet well understood. For this reason, the precipitation behavior and SXC adaptation for ORFV were investigated for the PEG/ORFV contact time, the membranes pore size, and the type and concentration of ions. All three parameters influenced the ORFV recoveries significantly. A small pore size and a long contact time induced filtration effects and inhibited a full virus recovery. The application of salts had complex concentration-dependent effects on precipitation and SXC yields, and ranged from a complete prevention of precipitation in the presence of kosmotropic substances to increased efficiencies with Mg2+ ions. The latter finding might be useful to reduce PEG concentrations while maintaining high yields. With this knowledge, we hope to clarify several limitations of SXC operations and improve the tool-set for a successful process adaptation.
Collapse
Affiliation(s)
- Friederike Eilts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Sabri Orbay
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
- PRiME Vector Technologies, Herrenberger Straße 24, 72070 Tuebingen, Germany
| | - Michael W. Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr.14, 35390 Giessen, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Ohlebergsweg 12, 35392 Giessen, Germany
| |
Collapse
|
17
|
Yang Z, Xu X, Silva CAT, Farnos O, Venereo-Sanchez A, Toussaint C, Dash S, González-Domínguez I, Bernier A, Henry O, Kamen A. Membrane Chromatography-Based Downstream Processing for Cell-Culture Produced Influenza Vaccines. Vaccines (Basel) 2022; 10:vaccines10081310. [PMID: 36016198 PMCID: PMC9414887 DOI: 10.3390/vaccines10081310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022] Open
Abstract
New influenza strains are constantly emerging, causing seasonal epidemics and raising concerns to the risk of a new global pandemic. Since vaccination is an effective method to prevent the spread of the disease and reduce its severity, the development of robust bioprocesses for producing pandemic influenza vaccines is exceptionally important. Herein, a membrane chromatography-based downstream processing platform with a demonstrated industrial application potential was established. Cell culture-derived influenza virus H1N1/A/PR/8/34 was harvested from benchtop bioreactor cultures. For the clarification of the cell culture broth, a depth filtration was selected as an alternative to centrifugation. After inactivation, an anion exchange chromatography membrane was used for viral capture and further processing. Additionally, two pandemic influenza virus strains, the H7N9 subtype of the A/Anhui/1/2013 and H3N2/A/Hong Kong/8/64, were successfully processed through similar downstream process steps establishing optimized process parameters. Overall, 41.3–62.5% viral recovery was achieved, with the removal of 86.3–96.5% host cell DNA and 95.5–99.7% of proteins. The proposed membrane chromatography purification is a scalable and generic method for the processing of different influenza strains and is a promising alternative to the current industrial purification of influenza vaccines based on ultracentrifugation methodologies.
Collapse
Affiliation(s)
- Zeyu Yang
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Xingge Xu
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Cristina A. T. Silva
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, Montreal, QC H3T 1J4, Canada
| | - Omar Farnos
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Alina Venereo-Sanchez
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Cécile Toussaint
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Shantoshini Dash
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Irene González-Domínguez
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Alice Bernier
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Olivier Henry
- Department of Chemical Engineering, Polytechnique Montreal, Montreal, QC H3T 1J4, Canada
| | - Amine Kamen
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada
- Correspondence:
| |
Collapse
|
18
|
Ma GJ, Yoon BK, Sut TN, Yoo KY, Lee SH, Jeon W, Jackman JA, Ariga K, Cho N. Lipid coating technology: A potential solution to address the problem of sticky containers and vanishing drugs. VIEW 2022. [DOI: 10.1002/viw.20200078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Gamaliel Junren Ma
- School of Materials Science and Engineering Nanyang Technological University Nanyang Singapore
| | - Bo Kyeong Yoon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS) Sungkyunkwan University Suwon Republic of Korea
| | - Tun Naw Sut
- School of Materials Science and Engineering Nanyang Technological University Nanyang Singapore
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS) Sungkyunkwan University Suwon Republic of Korea
| | - Ki Yeol Yoo
- LUCA Health and LUCA AICell, Inc. Anyang Republic of Korea
| | - Seung Hwa Lee
- LUCA Health and LUCA AICell, Inc. Anyang Republic of Korea
| | - Won‐Yong Jeon
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS) Sungkyunkwan University Suwon Republic of Korea
| | - Joshua A. Jackman
- School of Chemical Engineering and Biomedical Institute for Convergence at SKKU (BICS) Sungkyunkwan University Suwon Republic of Korea
| | - Katsuhiko Ariga
- WPI‐MANA National Institute for Materials Science (NIMS) Tsukuba Ibaraki Japan
- Department of Advanced Materials Science, Graduate School of Frontier Sciences The University of Tokyo Kashiwa Chiba Japan
| | - Nam‐Joon Cho
- School of Materials Science and Engineering Nanyang Technological University Nanyang Singapore
| |
Collapse
|
19
|
Shi R, Jia S, Liu H, Nie H. Clinical grade lentiviral vector purification and quality control requirements. J Sep Sci 2022; 45:2093-2101. [PMID: 35247228 DOI: 10.1002/jssc.202100937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 11/10/2022]
Abstract
Lentiviral vectors have been proven to be a powerful tool in gene therapies that includes the ability to perform long-term gene editing in both dividing and non-dividing cells. In order to meet the rising demand of clinical grade lentiviral vectors for future clinical trials and requirements by regulatory agencies, new methods and technologies were developed, including the rapid optimization of production and purification processes. However, gaps still exist in achieving ideal yields and recovery rates in large-scale manufacturing process steps. The downstream purification process is a critical step required to obtain sufficient quantity and high-quality lentiviral vectors products, which is challenged by the low stability of the LV particles and large production volumes associated with the manufacturing process. This review summarizes the most recent and promising technologies and enhancements used in the large-scale purification process step of LV manufacturing and aims to provide a significant contribution towards the achievement of providing sufficient quantity and quality of LVs in scalable processes. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ruina Shi
- Immunochina Pharmaceutical Co., Ltd., Beijing, China
| | - Shenghua Jia
- Immunochina Pharmaceutical Co., Ltd., Beijing, China
| | - Huwei Liu
- College of Life Sciences, Wuchang University of Technology, Wuhan, China.,Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Honggang Nie
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.,Analytical Instrumental Center, Peking University, Beijing, China
| |
Collapse
|
20
|
Bezeljak U. Cancer gene therapy goes viral: viral vector platforms come of age. Radiol Oncol 2022; 56:1-13. [PMID: 35148469 PMCID: PMC8884858 DOI: 10.2478/raon-2022-0002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Since the advent of viral vector gene therapy in 1990s, cancer treatment with viral vectors promised to revolutionize the field of oncology. Notably, viral vectors offer a unique combination of efficient gene delivery and engagement of the immune system for anti-tumour response. Despite the early potential, viral vector-based cancer treatments are only recently making a big impact, most prominently as gene delivery devices in approved CAR-T cell therapies, cancer vaccines and targeted oncolytic therapeutics. To reach this broad spectrum of applications, a number of challenges have been overcome - from our understanding of cancer biology to vector design, manufacture and engineering. Here, we take an overview of viral vector usage in cancer therapy and discuss the latest advancements. We also consider production platforms that enable mainstream adoption of viral vectors for cancer gene therapy. CONCLUSIONS Viral vectors offer numerous opportunities in cancer therapy. Recent advances in vector production platforms open new avenues in safe and efficient viral therapeutic strategies, streamlining the transition from lab bench to bedside. As viral vectors come of age, they could become a standard tool in the cancer treatment arsenal.
Collapse
|
21
|
Bankwitz D, Krey T, Pietschmann T. [Development approaches for vaccines against hepatitis C virus infections]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2022; 65:183-191. [PMID: 35015104 PMCID: PMC8749110 DOI: 10.1007/s00103-021-03477-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/07/2021] [Indexed: 11/04/2022]
Abstract
Mehr als 10 Jahre nach der Zulassung der ersten direkt wirkenden antiviralen Wirkstoffe zur Behandlung der Hepatitis C bleibt die Inzidenz der Hepatitis-C-Virus-(HCV-)Infektion ungebrochen hoch. In manchen Ländern stecken sich mehr Menschen neu mit dem Virus an, als Patienten durch eine erfolgreiche Therapie geheilt werden. Die Entwicklung eines prophylaktischen Impfstoffes könnte die Transmission des Virus unterbinden und dadurch einen wesentlichen Beitrag zur Kontrolle dieser weltweit verbreiteten Infektion leisten. In diesem Artikel werden die besonderen Herausforderungen und die aktuellen Ansätze der HCV-Impfstoffentwicklung dargestellt. HCV ist ein hochgradig diverses und wandlungsfähiges Virus, das zumeist dem Immunsystem entkommt und chronische Infektionen etabliert. Andererseits heilt die HCV-Infektion bei bis zu einem Drittel der exponierten Individuen aus, sodass eine schützende Immunität erreichbar ist. Zahlreiche Untersuchungen zu den Determinanten einer schützenden Immunität gegen HCV zeichnen ein immer kompletteres Bild davon, welche Ziele ein Impfstoff erreichen muss. Sehr wahrscheinlich werden sowohl starke neutralisierende Antikörper als auch wirkungsvolle zytotoxische T‑Zellen gebraucht, um sicher vor einer chronischen Infektion zu schützen. Die Schlüsselfrage ist, welche Ansätze besonders breit wirksame Antikörper und T‑Zellen heranreifen lassen. Dies wird erforderlich sein, um vor der großen Fülle unterschiedlicher HCV-Varianten zu schützen. Die jüngsten Erfolge von mRNA-Impfstoffen öffnen neue Türen auch für die HCV-Impfstoffforschung. Kombiniert mit einem tieferen Verständnis der Struktur und Funktion der viralen Hüllproteine, der Identifizierung kreuzprotektiver Antikörper- und T‑Zellepitope sowie der Nutzung standardisierter Verfahren zur Quantifizierung der Wirksamkeit von Impfkandidaten ergeben sich neue Perspektiven für die Entwicklung eines Impfstoffes.
Collapse
Affiliation(s)
- Dorothea Bankwitz
- Twincore Zentrum für Experimentelle und Klinische Infektionsforschung, Institut für Experimentelle Virologie, Feodor-Lynen-Str. 7, 30625, Hannover, Deutschland
| | - Thomas Krey
- Medizinische Hochschule Hannover, RESIST Exzellenzcluster EXC2155, Hannover, Deutschland.,Zentrum für Strukturbiologie und Zellbiologie in der Medizin, Institut für Biochemie, Universität Lübeck, Lübeck, Deutschland.,Deutsches Zentrum für Infektionsforschung (DZIF), Partnerstandort Hamburg-Lübeck-Borstel-Riems, Braunschweig, Deutschland.,Institut für Virologie, Medizinische Hochschule Hannover, Hannover, Deutschland
| | - Thomas Pietschmann
- Twincore Zentrum für Experimentelle und Klinische Infektionsforschung, Institut für Experimentelle Virologie, Feodor-Lynen-Str. 7, 30625, Hannover, Deutschland. .,Medizinische Hochschule Hannover, RESIST Exzellenzcluster EXC2155, Hannover, Deutschland. .,Deutsches Zentrum für Infektionsforschung (DZIF), Partnerstandort Hannover-Braunschweig, Braunschweig, Deutschland.
| |
Collapse
|
22
|
Production- and Purification-Relevant Properties of Human and Murine Cytomegalovirus. Viruses 2021; 13:v13122481. [PMID: 34960750 PMCID: PMC8706497 DOI: 10.3390/v13122481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022] Open
Abstract
There is a large unmet need for a prophylactic vaccine against human cytomegalovirus (HCMV) to combat the ubiquitous infection that is ongoing with this pathogen. A vaccination against HCMV could protect immunocompromised patients and prevent birth defects caused by congenital HCMV infections. Moreover, cytomegalovirus (CMV) has a number of features that make it a very interesting vector platform for gene therapy. In both cases, preparation of a highly purified virus is a prerequisite for safe and effective application. Murine CMV (MCMV) is by far the most studied model for HCMV infections with regard to the principles that govern the immune surveillance of CMVs. Knowledge transfer from MCMV and mice to HCMV and humans could be facilitated by better understanding and characterization of the biological and biophysical properties of both viruses. We carried out a detailed investigation of HCMV and MCMV growth kinetics as well as stability under the influence of clarification and different storage conditions. Further, we investigated the possibilities to concentrate and purify both viruses by ultracentrifugation and ion-exchange chromatography. Defective enveloped particles were not separately analyzed; however, the behavior of exosomes was examined during all experiments. The effectiveness of procedures was monitored using CCID50 assay, Nanoparticle tracking analysis, ELISA for host cell proteins, and quantitative PCR for host cell DNA. MCMV generally proved to be more robust in handling. Despite its greater sensitivity, HCMV was efficiently (100% recovery) purified and concentrated by anion-exchange chromatography using QA monolithic support. The majority of the host genomic DNA as well as most of the host cell proteins were removed by this procedure.
Collapse
|
23
|
Continuous purification of influenza A virus particles using pseudo-affinity membrane chromatography. J Biotechnol 2021; 342:139-148. [PMID: 34678401 DOI: 10.1016/j.jbiotec.2021.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 08/26/2021] [Accepted: 10/11/2021] [Indexed: 11/22/2022]
Abstract
Robust and flexible continuous unit operations that enable the establishment of intensified bioprocesses is one of the most relevant trends in manufacturing of biopharmaceuticals, including virus-based products. Sulfated cellulose membrane adsorbers (SCMA) are one of the most promising matrices for chromatographic purification of virus particles, like influenza viruses. Here, a three 'column' periodical counter current set-up was used to continuously purify influenza A/PR/8/34 virus particles using SCMA in bind-elute mode. It was possible to recover 67.4% of the HA-activity and to remove 67.4% and 99.8% of the total protein and DNA, respectively. The performance of the continuous process operated over a total of 10 loops, was slightly inferior to was obtained in a comparable batch process. Nevertheless, it was possible to increase the effective usage of binding capacity to 80%, resulting on a productivity of 22.8 kHAU mlmemb-1 min-1. As a proof-of-principle, SCMA were successfully used as matrix for purification of cell-derived influenza virus particles, in continuous mode.
Collapse
|
24
|
Evaluation of a downstream process for the recovery and concentration of a Cell-Culture-Derived rVSV-Spike COVID-19 vaccine candidate. Vaccine 2021; 39:7044-7051. [PMID: 34756612 PMCID: PMC8531466 DOI: 10.1016/j.vaccine.2021.10.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 12/25/2022]
Abstract
rVSV-Spike (rVSV-S) is a recombinant viral vaccine candidate under development to control the COVID-19 pandemic and is currently in phase II clinical trials. rVSV-S induces neutralizing antibodies and protects against SARS-CoV-2 infection in animal models. Bringing rVSV-S to clinical trials required the development of a scalable downstream process for the production of rVSV-S that can meet regulatory guidelines. The objective of this study was the development of the first downstream unit operations for cell-culture-derived rVSV-S, namely, the removal of nucleic acid contamination, the clarification and concentration of viral harvested supernatant, and buffer exchange. Retaining the infectivity of the rVSV-S during the downstream process was challenged by the shear sensitivity of the enveloped rVSV-S and its membrane protruding spike protein. Through a series of screening experiments, we evaluated and established the required endonuclease treatment conditions, filter train composition, and hollow fiber-tangential flow filtration parameters to remove large particles, reduce the load of impurities, and concentrate and exchange the buffer while retaining rVSV-S infectivity. The combined effect of the first unit operations on viral recovery and the removal of critical impurities was examined during scale-up experiments. Overall, approximately 40% of viral recovery was obtained and the regulatory requirements of less than 10 ng host cell DNA per dose were met. However, while 86–97% of the host cell proteins were removed, the regulatory acceptable HCP levels were not achieved, requiring subsequent purification and polishing steps. The results we obtained during the scale-up experiments were similar to those obtained during the screening experiments, indicating the scalability of the process. The findings of this study set the foundation for the development of a complete downstream manufacturing process, requiring subsequent purification and polishing unit operations for clinical preparations of rVSV-S.
Collapse
|
25
|
Makovitzki A, Jayson A, Oren Z, Lerer E, Kafri Y, Dor E, Cherry L, Tzadok H, Levin L, Hazan O, Simon I, Tal A, Girshengorn M, Rosen O. In-Line Monitoring of Downstream Purification Processes for VSV Based SARS-CoV-2 Vaccine Using a Novel Technique. BIOTECH 2021; 10:25. [PMID: 35822799 PMCID: PMC9245488 DOI: 10.3390/biotech10040025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/21/2021] [Accepted: 11/02/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) increases the need for a rapid development of efficient vaccines. Among other vaccines in clinical trials, a recombinant VSV-∆G-spike vaccine was developed by the Israel Institute for Biological Research (IIBR) and is being evaluated. The development of an efficient downstream purification process (DSP) enables the vaccine to be advanced to clinical trials. The DSP must eliminate impurities, either process- or product-related, to yield a sufficient product with high purity, potency and quality. To acquire critical information on process restrictions and qualities, the application of in-line monitoring is vital and should significantly impact the process yield, product quality and economy of the entire process. Here, we describe an in-line monitoring technique that was applied in the DSP of the VSV-∆G-spike vaccine. The technique is based on determining the concentrations of metabolites, nutrients and a host cell protein using the automatic chemistry analyzer, Cobas Integra 400 Plus. The analysis revealed critical information on process parameters and significantly impacted purification processes. The technique is rapid, easy and efficient. Adopting this technique during the purification process improves the process yield and the product quality and enhances the economy of the entire downstream process for biotechnology and bio pharmaceutical products.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Osnat Rosen
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel; (A.M.); (A.J.); (Z.O.); (E.L.); (Y.K.); (E.D.); (L.C.); (H.T.); (L.L.); (O.H.); (I.S.); (A.T.); (M.G.)
| |
Collapse
|
26
|
Highly Efficient Purification of Recombinant VSV-∆G-Spike Vaccine against SARS-CoV-2 by Flow-Through Chromatography. BIOTECH 2021; 10:biotech10040022. [PMID: 35822796 PMCID: PMC9245476 DOI: 10.3390/biotech10040022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 01/10/2023] Open
Abstract
This study reports a highly efficient, rapid one-step purification process for the production of the recombinant vesicular stomatitis virus-based vaccine, rVSV-∆G-spike (rVSV-S), recently developed by the Israel Institute for Biological Research (IIBR) for the prevention of COVID-19. Several purification strategies are evaluated using a variety of chromatography methods, including membrane adsorbers and packed-bed ion-exchange chromatography. Cell harvest is initially treated with endonuclease, clarified, and further concentrated by ultrafiltration before chromatography purification. The use of anion-exchange chromatography in all forms results in strong binding of the virus to the media, necessitating a high salt concentration for elution. The large virus and spike protein binds very strongly to the high surface area of the membrane adsorbents, resulting in poor virus recovery (<15%), while the use of packed-bed chromatography, where the surface area is smaller, achieves better recovery (up to 33%). Finally, a highly efficient chromatography purification process with CaptoTM Core 700 resin, which does not require binding and the elution of the virus, is described. rVSV-S cannot enter the inner pores of the resin and is collected in the flow-through eluent. Purification of the rVSV-S virus with CaptoTM Core 700 resulted in viral infectivity above 85% for this step, with the efficient removal of host cell proteins, consistent with regulatory requirements. Similar results were obtained without an initial ultrafiltration step.
Collapse
|
27
|
Li G, Ma W, Mo J, Cheng B, Shoda SI, Zhou D, Ye XS. Influenza Virus Precision Diagnosis and Continuous Purification Enabled by Neuraminidase-Resistant Glycopolymer-Coated Microbeads. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46260-46269. [PMID: 34547894 DOI: 10.1021/acsami.1c11561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Rapid diagnosis and vaccine development are critical to prevent the threat posed by viruses. However, rapid tests, such as colloidal gold assays, yield false-negative results due to the low quantities of viruses; moreover, conventional virus purification, including ultracentrifugation and nanofiltration, is multistep and time-consuming, which limits laboratory research and commercial development of viral vaccines. A rapid virus enrichment and purification technique will improve clinical diagnosis sensitivity and simplify vaccine production. Hence, we developed the surface-glycosylated microbeads (glycobeads) featuring chemically synthetic glycoclusters and reversible linkers to selectively capture the influenza virus. The surface plasmon resonance (SPR) evaluation indicated broad spectrum affinity of S-linked glycosides to various influenza viruses. The magnetic glycobeads were integrated into clinical rapid diagnosis, leading to a 30-fold lower limit of detection. Additionally, the captured viruses can be released under physiological conditions, delivering purified viruses with >50% recovery and without decreasing their native infectivity. Notably, this glycobead platform will facilitate the sensitive detection and continuous one-step purification of the target virus that contributes to future vaccine production.
Collapse
Affiliation(s)
- Gefei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Wenxiao Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Juan Mo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Boyang Cheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Shin-Ichiro Shoda
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, 6-6-11, Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| |
Collapse
|
28
|
Manufacturing Bacteriophages (Part 1 of 2): Cell Line Development, Upstream, and Downstream Considerations. Pharmaceuticals (Basel) 2021; 14:ph14090934. [PMID: 34577634 PMCID: PMC8471501 DOI: 10.3390/ph14090934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/15/2021] [Indexed: 01/21/2023] Open
Abstract
Within this first part of the two-part series on phage manufacturing, we will give an overview of the process leading to bacteriophages as a drug substance, before covering the formulation into a drug product in the second part. The principal goal is to provide the reader with a comprehensive framework of the challenges and opportunities that present themselves when developing manufacturing processes for bacteriophage-based products. We will examine cell line development for manufacture, upstream and downstream processes, while also covering the additional opportunities that engineered bacteriophages present.
Collapse
|
29
|
Abstract
The COVID-19 pandemic has motivated the rapid development of numerous vaccines that have proven effective against SARS-CoV-2. Several of these successful vaccines are based on the adenoviral vector platform. The mass manufacturing of these vaccines poses great challenges, especially in the context of a pandemic where extremely large quantities must be produced quickly at an affordable cost. In this work, two baseline processes for the production of a COVID-19 adenoviral vector vaccine, B1 and P1, were designed, simulated and economically evaluated with the aid of the software SuperPro Designer. B1 used a batch cell culture viral production step, with a viral titer of 5 × 1010 viral particles (VP)/mL in both stainless-steel and disposable equipment. P1 used a perfusion cell culture viral production step, with a viral titer of 1 × 1012 VP/mL in exclusively disposable equipment. Both processes were sized to produce 400 M/yr vaccine doses. P1 led to a smaller cost per dose than B1 ($0.15 vs. $0.23) and required a much smaller capital investment ($126 M vs. $299 M). The media and facility-dependent expenses were found to be the main contributors to the operating cost. The results indicate that adenoviral vector vaccines can be practically manufactured at large scale and low cost.
Collapse
|
30
|
Eckhardt D, Dieken H, Loewe D, Grein TA, Salzig D, Czermak P. Purification of oncolytic measles virus by cation-exchange chromatography using resin-based stationary phases. SEP SCI TECHNOL 2021. [DOI: 10.1080/01496395.2021.1955267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Dustin Eckhardt
- Department of cell culture technology, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Hauke Dieken
- Department of cell culture technology, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Daniel Loewe
- Department of cell culture technology, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
- Faculty of Biology and Chemistry, University of Giessen, Giessen Germany
| | - Tanja A. Grein
- Department of cell culture technology, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Denise Salzig
- Department of cell culture technology, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Peter Czermak
- Department of cell culture technology, Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
- Faculty of Biology and Chemistry, University of Giessen, Giessen Germany
| |
Collapse
|
31
|
Bissinger T, Wu Y, Marichal-Gallardo P, Riedel D, Liu X, Genzel Y, Tan WS, Reichl U. Towards integrated production of an influenza A vaccine candidate with MDCK suspension cells. Biotechnol Bioeng 2021; 118:3996-4013. [PMID: 34219217 DOI: 10.1002/bit.27876] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/01/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Seasonal influenza epidemics occur both in northern and southern hemispheres every year. Despite the differences in influenza virus surface antigens and virulence of seasonal subtypes, manufacturers are well-adapted to respond to this periodical vaccine demand. Due to decades of influenza virus research, the development of new influenza vaccines is relatively straight forward. In similarity with the ongoing coronavirus disease 2019 pandemic, vaccine manufacturing is a major bottleneck for a rapid supply of the billions of doses required worldwide. In particular, egg-based vaccine production would be difficult to schedule and shortages of other egg-based vaccines with high demands also have to be anticipated. Cell culture-based production systems enable the manufacturing of large amounts of vaccines within a short time frame and expand significantly our options to respond to pandemics and emerging viral diseases. In this study, we present an integrated process for the production of inactivated influenza A virus vaccines based on a Madin-Darby Canine Kidney (MDCK) suspension cell line cultivated in a chemically defined medium. Very high titers of 3.6 log10 (HAU/100 µl) were achieved using fast-growing MDCK cells at concentrations up to 9.5 × 106 cells/ml infected with influenza A/PR/8/34 H1N1 virus in 1 L stirred tank bioreactors. A combination of membrane-based steric-exclusion chromatography followed by pseudo-affinity chromatography with a sulfated cellulose membrane adsorber enabled full recovery for the virus capture step and up to 80% recovery for the virus polishing step. Purified virus particles showed a homogenous size distribution with a mean diameter of 80 nm. Based on a monovalent dose of 15 µg hemagglutinin (single-radial immunodiffusion assay), the level of total protein and host cell DNA was 58 µg and 10 ng, respectively. Furthermore, all process steps can be fully scaled up to industrial quantities for commercial manufacturing of either seasonal or pandemic influenza virus vaccines. Fast production of up to 300 vaccine doses per liter within 4-5 days makes this process competitive not only to other cell-based processes but to egg-based processes as well.
Collapse
Affiliation(s)
- Thomas Bissinger
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Yixiao Wu
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Pavel Marichal-Gallardo
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Dietmar Riedel
- Facility for Transmission Electron Microscopy, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Xuping Liu
- Shanghai BioEngine Sci-Tech Co., Shanghai, China
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Shanghai BioEngine Sci-Tech Co., Shanghai, China
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,Chair of Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
32
|
Zulkarnain NN, Anuar N, Abd Rahman N, Sheikh Abdullah SR, Alias MN, Yaacob M, Ma Z, Ding G. Cell-based influenza vaccine: current production, halal status assessment, and recommendations towards Islamic-compliant manufacturing. Hum Vaccin Immunother 2021; 17:2158-2168. [PMID: 33539195 DOI: 10.1080/21645515.2020.1865044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Influenza virus is a life-threatening pathogen that infects millions of people every year, with annual mortality in the hundreds of thousands. The scenario for controlling infection has worsened with increasing numbers of vaccine hesitancy cases reported worldwide due to objections on safety, religious and other grounds. Uses of haram (impermissible) and mashbooh (doubtful) ingredients in vaccine production has raised doubts among Muslim consumers and consequently stimulated serious vaccine hesitancy. To address this major problem, we have reviewed and recommended some alternatives appropriate for manufacturing cell-based influenza vaccine which comply with Islamic laws and consumers' needs. Intensive assessments of current influenza vaccine production in both scientific and Islamic views have led to the identification of four main ingredients deemed impermissible in novel sharia-compliant (approved by Islamic laws) vaccine manufacturing. Only some of these impermissible components could be replaced with halal (permissible) alternatives, while others remain impermissible due to unavailability and unsuitability.
Collapse
Affiliation(s)
- Nurul Nadiah Zulkarnain
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Nurina Anuar
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Norliza Abd Rahman
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Siti Rozaimah Sheikh Abdullah
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Muhammad Nazir Alias
- Centre for Contemporary Fiqh and Sharia Compliance, Faculty of Islamic Studies, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Mashitoh Yaacob
- Centre for Liberal Education, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia.,Institute of Islam Hadhari, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Zhongren Ma
- Biomedical Research Centre, Northwest Minzu University, Lanzhou, Gansu, China
| | - Gongtao Ding
- Biomedical Research Centre, Northwest Minzu University, Lanzhou, Gansu, China
| |
Collapse
|
33
|
B Carvalho S, Peixoto C, T Carrondo MJ, S Silva RJ. Downstream processing for influenza vaccines and candidates: An update. Biotechnol Bioeng 2021; 118:2845-2869. [PMID: 33913510 DOI: 10.1002/bit.27803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Seasonal and pandemic influenza outbreaks present severe health and economic burdens. To overcome limitations on influenza vaccines' availability and effectiveness, researchers chase universal vaccines providing broad, long-lasting protection against multiple influenza subtypes, and including pandemic ones. Novel influenza vaccine designs are under development, in clinical trials, or reaching the market, namely inactivated, or live-attenuated virus, virus-like particles, or recombinant antigens, searching for improved effectiveness; all these bring downstream processing (DSP) new challenges. Having to deal with new influenza strains, including pandemics, requires shorter development time, driving the development of faster bioprocesses. To cope with better upstream processes, new regulatory demands for quality and safety, and cost reduction requirements, new unit operations and integrated processes are increasing DSP efficiency for novel vaccine formats. This review covers recent advances in DSP strategies of different influenza vaccine formats. Focus is given to the improvements on relevant state-of-the-art unit operations, from harvest and clarification to purification steps, ending with sterile filtration and formulation. The development of more efficient unit operations to cope with biophysical properties of the new candidates is discussed: emphasis is given to the design of new stationary phases, 3D printing approaches, and continuous processing tools, such as continuous chromatography. The impact of the production platforms and vaccine designs on the downstream operations for the different influenza vaccine formats approved for this season are highlighted.
Collapse
Affiliation(s)
- Sofia B Carvalho
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cristina Peixoto
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Manuel J T Carrondo
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Ricardo J S Silva
- Animal Cell Technology Unit, iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
34
|
Mendes JP, Silva RJS, Berg M, Mathiasson L, Peixoto C, Alves PM, Carrondo MJT. Oncolytic virus purification with periodic counter-current chromatography. Biotechnol Bioeng 2021; 118:3522-3532. [PMID: 33818758 DOI: 10.1002/bit.27779] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 11/11/2022]
Abstract
Virus-based biologicals are one of the most promising biopharmaceuticals of the 21st century medicine and play a significant role in the development of innovative therapeutic, prophylactic, and clinical applications. Oncolytic virus manufacturing scale can range from 5 L in research and development up to 50 L for clinical studies and reach hundreds of liters for commercial scale. The inherent productivity and high integration potential of periodic counter-current chromatography (PCC) offer a transversal solution to decrease equipment footprint and the reduction of several non-value-added unit operations. We report on the design of an efficient PCC process applied to the intermediate purification of oncolytic adenovirus. The developed ion-exchange chromatographic purification method was carried out using a four-column setup for three different scenarios: (i) variation in the feedstock, (ii) potential use of a post-load washing step to improve virus recovery, and (iii) stability during extended operation. Obtained virus recoveries (57%-86%) and impurity reductions (>80% DNA, and >70% total protein) match or overcome batch purification. Regarding process stability and automation, our results show that not only the dynamic control strategy used is able to suppress perturbations in the sample inlet but also allows for unattended operation in the case of ion exchange capture.
Collapse
Affiliation(s)
- João P Mendes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ricardo J S Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | | | - Cristina Peixoto
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | |
Collapse
|
35
|
Shoaebargh S, Wright E, Csordas M, Medina MFC, Lichty B, Latulippe DR. Probing effects of additives on the filterability of oncolytic viruses via a microfiltration process. J Memb Sci 2021. [DOI: 10.1016/j.memsci.2020.118783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
36
|
Moreira AS, Cavaco DG, Faria TQ, Alves PM, Carrondo MJT, Peixoto C. Advances in Lentivirus Purification. Biotechnol J 2020; 16:e2000019. [PMID: 33089626 DOI: 10.1002/biot.202000019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022]
Abstract
Lentiviral vectors (LVs) have been increasingly used as a tool for gene and cell therapies since they can stably integrate the genome in dividing and nondividing cells. LV production and purification processes have evolved substantially over the last decades. However, the increasing demands for higher quantities with more restrictive purity requirements are stimulating the development of novel materials and strategies to supply the market with LV in a cost-effective manner. A detailed review of each downstream process unit operation is performed, limitations, strengths, and potential outcomes being covered. Currently, the majority of large-scale LV manufacturing processes are still based on adherent cell culture, although it is known that the industry is migrating fast to suspension cultures. Regarding the purification strategy, it consists of batch chromatography and membrane technology. Nevertheless, new solutions are being created to improve the current production schemes and expand its clinical use.
Collapse
Affiliation(s)
- Ana Sofia Moreira
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - David Guia Cavaco
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Tiago Q Faria
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Manuel J T Carrondo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal
| | - Cristina Peixoto
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal
| |
Collapse
|
37
|
Soldi M, Sergi Sergi L, Unali G, Kerzel T, Cuccovillo I, Capasso P, Annoni A, Biffi M, Rancoita PMV, Cantore A, Lombardo A, Naldini L, Squadrito ML, Kajaste-Rudnitski A. Laboratory-Scale Lentiviral Vector Production and Purification for Enhanced Ex Vivo and In Vivo Genetic Engineering. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:411-425. [PMID: 33294490 PMCID: PMC7683235 DOI: 10.1016/j.omtm.2020.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/13/2020] [Indexed: 12/31/2022]
Abstract
Lentiviral vectors (LVs) are increasingly employed in gene and cell therapy. Standard laboratory production of LVs is not easily scalable, and research-grade LVs often contain contaminants that can interfere with downstream applications. Moreover, purified LV production pipelines have been developed mainly for costly, large-scale, clinical-grade settings. Therefore, a standardized and cost-effective process is still needed to obtain efficient, reproducible, and properly executed experimental studies and preclinical development of ex vivo and in vivo gene therapies, as high infectivity and limited adverse reactions are important factors potentially influencing experimental outcomes also in preclinical settings. We describe here an optimized laboratory-scale workflow whereby an LV-containing supernatant is purified and concentrated by sequential chromatographic steps, obtaining biologically active LVs with an infectious titer and specific activity in the order of 109 transducing unit (TU)/mL and 5 × 104 TU/ng of HIV Gag p24, respectively. The purification workflow removes >99% of the starting plasmid, DNA, and protein impurities, resulting in higher gene transfer and editing efficiency in severe combined immunodeficiency (SCID)-repopulating hematopoietic stem and progenitor cells (HSPCs) ex vivo, as well as reduced activation of inflammatory responses ex vivo and in vivo as compared to TU-matched, laboratory-grade vectors. Our results highlight the value of accessible purified LV production for experimental studies and preclinical testing.
Collapse
Affiliation(s)
- Monica Soldi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Lucia Sergi Sergi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Giulia Unali
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Thomas Kerzel
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Paola Capasso
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Mauro Biffi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Paola Maria Vittoria Rancoita
- CUSSB-University Center for Statistics and the Biomedical Statistics, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, 20132 Milan, Italy
| | - Mario Leonardo Squadrito
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
38
|
Development of a downstream process for the production of an inactivated whole hepatitis C virus vaccine. Sci Rep 2020; 10:16261. [PMID: 33004836 PMCID: PMC7530675 DOI: 10.1038/s41598-020-72328-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
There is a large unmet need for a prophylactic hepatitis C virus (HCV) vaccine to control the ongoing epidemic with this deadly pathogen. Many antiviral vaccines employ whole viruses as antigens. For HCV, this approach became feasible following the development of infectious cell culture systems for virus production. However, the lack of efficient downstream processes (DSP) for HCV purification poses a roadblock for the development of a whole virus vaccine. Using cell culture-derived genotype 1a HCV we developed a scalable and efficient DSP train, employing commonly used clarification and ultrafiltration techniques, followed by two membrane-based chromatography steps. For virus capture, steric exclusion chromatography using cellulose membranes was established, resulting in a virtually complete virus recovery with > 99% protein and 84% DNA depletion. Virus polishing was achieved by sulphated cellulose membrane adsorbers with ~ 50% virus recovery and > 99% protein and 90% DNA depletion. Additional nuclease digestion resulted in 99% overall DNA depletion with final DNA concentrations of 2 ng/mL. Process results were comparable for cell culture-derived HCV of another major genotype (5a). This study provides proof-of-concept for establishment of an efficient and economically attractive DSP with potential application for production of an inactivated whole virus vaccine against HCV for human use.
Collapse
|
39
|
Lothert K, Dekevic G, Loewe D, Salzig D, Czermak P, Wolff MW. Upstream and Downstream Processes for Viral Nanoplexes as Vaccines. Methods Mol Biol 2020; 2183:217-248. [PMID: 32959247 DOI: 10.1007/978-1-0716-0795-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The increasing medical interest in viral nanoplexes, such as viruses or virus-like particles used for vaccines, gene therapy products, or oncolytic agents, raises the need for fast and efficient production processes. In general, these processes comprise upstream and downstream processing. For the upstream process, efficiency is mainly characterized by robustly achieving high titer yields, while reducing process times and costs with regard to the cell culture medium, the host cell selection, and the applied process conditions. The downstream part, on the other hand, should effectively remove process-related contaminants, such as host cells/cell debris as well as host cell DNA and proteins, while maintaining product stability and reducing product losses. This chapter outlines a combination of process steps to successfully produce virus particles in the controlled environment of a stirred tank bioreactor, combined with a platform-based purification approach using filtration-based clarification and steric exclusion chromatography. Additionally, suggestions for off-line analytics in terms of virus characterization and quantification as well as for contaminant estimation are provided.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology (IBPT), Technische Hochschule Mittelhessen (THM) - University of Applied Sciences, Giessen, Germany
| | - Gregor Dekevic
- Institute of Bioprocess Engineering and Pharmaceutical Technology (IBPT), Technische Hochschule Mittelhessen (THM) - University of Applied Sciences, Giessen, Germany
| | - Daniel Loewe
- Institute of Bioprocess Engineering and Pharmaceutical Technology (IBPT), Technische Hochschule Mittelhessen (THM) - University of Applied Sciences, Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology (IBPT), Technische Hochschule Mittelhessen (THM) - University of Applied Sciences, Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology (IBPT), Technische Hochschule Mittelhessen (THM) - University of Applied Sciences, Giessen, Germany.,Faculty of Biology and Chemistry, Justus-Liebig-University Giessen, Giessen, Germany.,Division Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology (IBPT), Technische Hochschule Mittelhessen (THM) - University of Applied Sciences, Giessen, Germany.
| |
Collapse
|
40
|
Production of Baculovirus and Stem Cells for Baculovirus-Mediated Gene Transfer into Human Mesenchymal Stem Cells. Methods Mol Biol 2020; 2183:367-390. [PMID: 32959254 DOI: 10.1007/978-1-0716-0795-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The discovery of the genome-editing tool CRISPR-Cas9 is revolutionizing the world of gene therapy and will extend the gene therapy product pipeline. While applying gene therapy products, the main difficulty is an efficient and effective transfer of the nucleic acids carrying the relevant information to their target destination, the nucleus of the cells. Baculoviruses have shown to be very suitable transport vehicles for this task due to, inter alia, their ability to transduce mammalian/human cells without being pathogenic. This property allows the usage of baculovirus-transduced cells as cell therapy products, thus, combining the advantages of gene and cell therapy. To make such pharmaceuticals available for patients, a successful production and purification is necessary. In this chapter, we describe the generation of a pseudotyped baculovirus vector, followed by downstream processing using depth and tangential-flow filtration. This vector is used subsequently to transduce human mesenchymal stem cells. The production of the cells and the subsequent transduction process are illustrated.
Collapse
|
41
|
Lothert K, Pagallies F, Eilts F, Sivanesapillai A, Hardt M, Moebus A, Feger T, Amann R, Wolff MW. A scalable downstream process for the purification of the cell culture-derived Orf virus for human or veterinary applications. J Biotechnol 2020; 323:221-230. [PMID: 32860824 DOI: 10.1016/j.jbiotec.2020.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/06/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022]
Abstract
The large demand for safe and efficient viral vector-based vaccines and gene therapies against both inherited and acquired diseases accelerates the development of viral vectors. One outstanding example, the Orf virus, has a wide range of applications, a superior efficacy and an excellent safety profile combined with a reduced pathogenicity compared to other viral vectors. However, besides these favorable attributes, an efficient and scalable downstream process still needs to be developed. Recently, we screened potential chromatographic stationary phases for Orf virus purification. Based on these previous accomplishments, we developed a complete downstream process for the cell culture-derived Orf virus. The described process comprises a membrane-based clarification step, a nuclease treatment, steric exclusion chromatography, and a secondary chromatographic purification step using Capto® Core 700 resin. The applicability of this process to a variety of diverse Orf virus vectors was shown, testing two different genotypes. These studies render the possibility to apply the developed downstream scheme for both genotypes, and lead to overall virus yields of about 64 %, with step recoveries of >70 % for the clarification, and >90 % for the chromatography train. Protein concentrations of the final product are below the detection limits, and the final DNA concentration of about 1 ng per 1E + 06 infective virus units resembles a total DNA depletion of 96-98 %.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Friederike Eilts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Arabi Sivanesapillai
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Martin Hardt
- Imaging Unit, Biomedical Research Centre Seltersberg, Justus Liebig University, Giessen, Germany
| | - Anna Moebus
- Imaging Unit, Biomedical Research Centre Seltersberg, Justus Liebig University, Giessen, Germany
| | - Thomas Feger
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany.
| |
Collapse
|
42
|
Lothert K, Pagallies F, Feger T, Amann R, Wolff MW. Selection of chromatographic methods for the purification of cell culture-derived Orf virus for its application as a vaccine or viral vector. J Biotechnol 2020; 323:62-72. [PMID: 32763261 PMCID: PMC7403136 DOI: 10.1016/j.jbiotec.2020.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/23/2020] [Accepted: 07/31/2020] [Indexed: 12/02/2022]
Abstract
Estimation of the isoelectric point and size of Vero cell-derived Orf virus. Limited dynamic binding capacity of tested Orf virus to sulfated cellulose. Purification of Orf virus by steric exclusion chromatography lead to 84 % recovery. Hydrophobic interaction chromatography suitable for Orf virus purification. Promising unit operations for a scalable DSP to produce Orf virus viral vectors.
In recent years, the Orf virus has become a promising tool for protective recombinant vaccines and oncolytic therapy. However, suitable methods for an Orf virus production, including up- and downstream, are very limited. The presented study focuses on downstream processing, describing the evaluation of different chromatographic unit operations. In this context, ion exchange-, pseudo-affinity- and steric exclusion chromatography were employed for the purification of the cell culture-derived Orf virus, aiming at a maximum in virus recovery and contaminant depletion. The most promising chromatographic methods for capturing the virus particles were the steric exclusion- or salt-tolerant anion exchange membrane chromatography, recovering 84 % and 86 % of the infectious virus. Combining the steric exclusion chromatography with a subsequent Capto™ Core 700 resin or hydrophobic interaction membrane chromatography as a secondary chromatographic step, overall virus recoveries of up to 76 % were achieved. Furthermore, a complete cellular protein removal and a host cell DNA depletion of up to 82 % was possible for the steric exclusion membranes and the Capto™ Core 700 combination. The study reveals a range of possible unit operations suited for the chromatographic purification of the cell culture-derived Orf virus, depending on the intended application, i.e. a human or veterinary use, and the required purity.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Thomas Feger
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany.
| |
Collapse
|
43
|
Pereira Aguilar P, Reiter K, Wetter V, Steppert P, Maresch D, Ling WL, Satzer P, Jungbauer A. Capture and purification of Human Immunodeficiency Virus-1 virus-like particles: Convective media vs porous beads. J Chromatogr A 2020; 1627:461378. [PMID: 32823092 DOI: 10.1016/j.chroma.2020.461378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 02/04/2023]
Abstract
Downstream processing (DSP) of large bionanoparticles is still a challenge. The present study aims to systematically compare some of the most commonly used DSP strategies for capture and purification of enveloped viruses and virus-like particles (eVLPs) by using the same staring material and analytical tools. As a model, Human Immunodeficiency Virus-1 (HIV-1) gag VLPs produced in CHO cells were used. Four different DSP strategies were tested. An anion-exchange monolith and a membrane adsorber, for direct capture and purification of eVLPs, and a polymer-grafted anion-exchange resin and a heparin-affinity resin for eVLP purification after a first flow-through step to remove small impurities. All tested strategies were suitable for capture and purification of eVLPs. The performance of the different strategies was evaluated regarding its binding capacity, ability to separate different particle populations and product purity. The highest binding capacity regarding total particles was obtained using the anion exchange membrane adsorber (5.3 × 1012 part/mL membrane), however this method did not allow the separation of different particle populations. Despite having a lower binding capacity (1.5 × 1011 part/mL column) and requiring a pre-processing step with flow-through chromatography, Heparin-affinity chromatography showed the best performance regarding separation of different particle populations, allowing not only the separation of HIV-1 gag VLPs from host cell derived bionanoparticles but also from chromatin. This work additionally shows the importance of thorough sample characterization combining several biochemical and biophysical methods in eVLP DSP.
Collapse
Affiliation(s)
- Patricia Pereira Aguilar
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Katrin Reiter
- Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Viktoria Wetter
- Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Petra Steppert
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Daniel Maresch
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Wai Li Ling
- Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Peter Satzer
- Austrian Centre of Industrial Biotechnology, Vienna, Austria.
| | - Alois Jungbauer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre of Industrial Biotechnology, Vienna, Austria
| |
Collapse
|
44
|
Martins DL, Sencar J, Hammerschmidt N, Flicker A, Kindermann J, Kreil TR, Jungbauer A. Truly continuous low pH viral inactivation for biopharmaceutical process integration. Biotechnol Bioeng 2020; 117:1406-1417. [PMID: 32017010 PMCID: PMC7187162 DOI: 10.1002/bit.27292] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/16/2019] [Accepted: 02/02/2020] [Indexed: 12/18/2022]
Abstract
Continuous virus inactivation (VI) has received little attention in the efforts to realize fully continuous biomanufacturing in the future. Implementation of continuous VI must assure a specific minimum incubation time, typically 60 min. To guarantee the minimum incubation time, we implemented a packed bed continuous viral inactivation reactor (CVIR) with narrow residence time distribution (RTD) for low pH incubation. We show that the RTD does not broaden significantly over a wide range of linear flow velocities-which highlights the flexibility and robustness of the design. Prolonged exposure to acidic pH has no impact on bed stability, assuring constant RTD throughout long term operation. The suitability of the packed bed CVIR for low pH inactivation is shown with two industry-standard model viruses, that is xenotropic murine leukemia virus and pseudorabies virus. Controls at neutral pH showed no system-induced VI. At low pH, significant VI is observed, even after only 15 min. Based on the low pH inactivation kinetics, the continuous process is equivalent to traditional batch operation. This study establishes a concept for continuous low pH inactivation and, together with previous reports, highlights the versatility of the packed bed reactor for continuous VI, regardless of the inactivation method.
Collapse
Affiliation(s)
- Duarte L. Martins
- Austria Centre for Industrial BiotechnologyViennaAustria
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Jure Sencar
- Austria Centre for Industrial BiotechnologyViennaAustria
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Nikolaus Hammerschmidt
- Austria Centre for Industrial BiotechnologyViennaAustria
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Andreas Flicker
- Department of VirologyGlobal Pathogen SafetyTakedaViennaAustria
| | | | - Thomas R. Kreil
- Department of VirologyGlobal Pathogen SafetyTakedaViennaAustria
| | - Alois Jungbauer
- Austria Centre for Industrial BiotechnologyViennaAustria
- Department of BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
45
|
Loewe D, Dieken H, Grein TA, Weidner T, Salzig D, Czermak P. Opportunities to debottleneck the downstream processing of the oncolytic measles virus. Crit Rev Biotechnol 2020; 40:247-264. [PMID: 31918573 DOI: 10.1080/07388551.2019.1709794] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oncolytic viruses (including measles virus) offer an alternative approach to reduce the high mortality rate of late-stage cancer. Several measles virus strains infect and lyse cancer cells efficiently, but the broad application of this therapeutic concept is hindered by the large number of infectious particles required (108-1012 TCID50 per dose). The manufacturing process must, therefore, achieve high titers of oncolytic measles virus (OMV) during upstream production and ensure that the virus product is not damaged during purification by applying appropriate downstream processing (DSP) unit operations. DSP is currently a production bottleneck because there are no specific platforms for OMV. Infectious OMV must be recovered as intact, enveloped particles, and host cell proteins and DNA must be reduced to acceptable levels to meet regulatory guidelines that were developed for virus-based vaccines and gene therapy vectors. Handling such high viral titers and process volumes is technologically challenging and expensive. This review considers the state of the art in OMV purification and looks at promising DSP technologies. We discuss here the purification of other enveloped viruses where such technologies could also be applied to OMV. The development of DSP technologies tailored for enveloped viruses is necessary to produce sufficient titers for virotherapy, which could offer hope to millions of patients suffering from incurable cancer.
Collapse
Affiliation(s)
- Daniel Loewe
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany.,Faculty of Biology and Chemistry, University of Giessen, Giessen, Germany
| | - Hauke Dieken
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Tanja A Grein
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Tobias Weidner
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany.,Faculty of Biology and Chemistry, University of Giessen, Giessen, Germany.,Project Group Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany
| |
Collapse
|
46
|
Silva RJS, Mendes JP, Carrondo MJT, Marques PM, Peixoto C. Continuous Chromatography Purification of Virus-Based Biopharmaceuticals: A Shortcut Design Method. Methods Mol Biol 2020; 2095:367-384. [PMID: 31858479 DOI: 10.1007/978-1-0716-0191-4_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Novel biopharmaceutical products, such as vaccines and viral vectors, play a significant role in the development of innovative therapeutic, prophylactic, and clinical applications. However, several challenges are posed when manufacturing these products. The diversity of cell lines and the different physical and chemical properties of these biologicals require the use of different production and processing technologies. Alternative purification strategies that can improve the purification yield, such as continuous chromatography, are regarded nowadays as enabling technologies to overcome some of the bottlenecks in biomanufacturing. This chapter offers a shortcut approach to implement a semi-continuous chromatography purification of hepatitis C virus-like particles produced in insect cells with recombinant baculovirus. Although the purification is based on ion exchange chromatography, the present methodology can be extended to other types of chromatography.
Collapse
Affiliation(s)
| | - João P Mendes
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | | | - Paula M Marques
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cristina Peixoto
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
47
|
Durous L, Rosa-Calatrava M, Petiot E. Advances in influenza virus-like particles bioprocesses. Expert Rev Vaccines 2019; 18:1285-1300. [DOI: 10.1080/14760584.2019.1704262] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Laurent Durous
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Emma Petiot
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| |
Collapse
|
48
|
Moleirinho MG, Silva RJS, Alves PM, Carrondo MJT, Peixoto C. Current challenges in biotherapeutic particles manufacturing. Expert Opin Biol Ther 2019; 20:451-465. [PMID: 31773998 DOI: 10.1080/14712598.2020.1693541] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: The development of novel complex biotherapeutics led to new challenges in biopharmaceutical industry. The potential of these particles has been demonstrated by the approval of several products, in the different fields of gene therapy, oncolytic therapy, and tumor vaccines. However, their manufacturing still presents challenges related to the high dosages and purity required.Areas covered: The main challenges that biopharmaceutical industry faces today and the most recent developments in the manufacturing of different biotherapeutic particles are reported here. Several unit operations and downstream trains to purify virus, virus-like particles and extracellular vesicles are described. Innovations on the different purification steps are also highlighted with an eye on the implementation of continuous and integrated processes.Expert opinion: Manufacturing platforms that consist of a low number of unit operations, with higher-yielding processes and reduced costs will be highly appreciated by the industry. The pipeline of complex therapeutic particles is expanding and there is a clear need for advanced tools and manufacturing capacity. The use of single-use technologies, as well as continuous integrated operations, are gaining ground in the biopharmaceutical industry and should be supported by more accurate and faster analytical methods.
Collapse
Affiliation(s)
- Mafalda G Moleirinho
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal.,ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Ricardo J S Silva
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal
| | - Paula M Alves
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal.,ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| | - Manuel J T Carrondo
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal
| | - Cristina Peixoto
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado, Oeiras, Portugal.,ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, Oeiras, Portugal
| |
Collapse
|
49
|
Use of sulfated cellulose membrane adsorbers for chromatographic purification of cell cultured-derived influenza A and B viruses. Sep Purif Technol 2019. [DOI: 10.1016/j.seppur.2019.05.101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
50
|
Lothert K, Sprick G, Beyer F, Lauria G, Czermak P, Wolff MW. Membrane-based steric exclusion chromatography for the purification of a recombinant baculovirus and its application for cell therapy. J Virol Methods 2019; 275:113756. [PMID: 31644888 DOI: 10.1016/j.jviromet.2019.113756] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/07/2019] [Accepted: 10/16/2019] [Indexed: 01/09/2023]
Abstract
The continuously increasing potential of stem cell treatments for various medical conditions has accelerated the need for fast and efficient purification techniques for individualized cell therapy applications. Genetic stem cell engineering is commonly done with viral vectors like the baculovirus. The baculovirus is a safe and efficient gene transfer tool, that has been used for the expression of recombinant proteins for many years. Its purification has been based mainly on ion exchange matrices. However, these techniques impair process robustness, if different genetically modified virus particles are applied. Here, we evaluated the membrane-based steric exclusion chromatography for the purification of insect cell culture-derived recombinant Autographa californica multicapsid nucleopolehydroviruses for an application in cell therapy. The method has already proven to be a powerful tool for the purification of Influenza A virus particles, using cellulose membranes. Aside from the aforementioned cellulose, we evaluated alternative stationary phases, such as glass fiber and polyamide membranes. The highest dynamic binding capacitiy was determined for cellulose with 5.08E + 07 pfu per cm² membrane. Critical process parameters were optimized, using a design of experiments (DoE) approach. The determined process conditions were verified by different production batches, obtaining a mean virus yield of 91% ± 6.5%. Impurity depletion was >99% and 85% for protein and dsDNA, without nuclease treatment. Due to the method's specificity, its application to other baculoviruses, with varying surface modifications, is conceivable without major process changes. The physiological buffer conditions enable a gentle handling of the virus particles without decreasing the transduction efficacy. The simple procedure with sufficient impurity removal enables the substitution of time-consuming ultra centrifugation steps and can serve as a first process unit operation to obtain higher purities.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany.
| | - Gundula Sprick
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany
| | - Felix Beyer
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany
| | - Guiliano Lauria
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany; Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Winchesterstr. 2, 35394, Giessen, Germany; Faculty of Biology and Chemistry, Justus-Liebig-University of Giessen, Ludwigstr. 23, 35390, Giessen, Germany.
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany.
| |
Collapse
|