1
|
Kishida N, Imai M, Ainai A, Asanuma H, Saito R, Fujisaki S, Shirakura M, Nakamura K, Kuwahara T, Takashita E, Tashiro M, Odagiri T, Watanabe S. Efficacy of an Inactivated Whole-Virus A/Victoria/361/2011 (IVR-165) (H3N2) Influenza Vaccine in Ferrets. Microbiol Immunol 2024; 68:427-437. [PMID: 39513563 PMCID: PMC11632575 DOI: 10.1111/1348-0421.13179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 10/02/2024] [Accepted: 10/20/2024] [Indexed: 11/15/2024]
Abstract
It has been reported that the high-growth reassortant (HGR) A(H3N2) influenza viruses used for split influenza vaccine (SV) production have some amino acid substitutions in hemagglutinin due to egg adaptation during virus propagation, causing antigenic differences between HGR and epidemic viruses. To clarify whether inactivated whole-virus vaccine (WV) derived from the A(H3N2) HGR virus possessing egg adaptation could induce cross-protective immune responses against epidemic A(H3N2) viruses, the efficacy of WV was compared with that of SV in a ferret model. When the ferrets immunized with WV or SV derived from HGR A/Victoria/361/2011 (IVR-165) virus were challenged with the homologous virus A/Victoria/361/2011 (IVR-165) or its original cell-propagated A/Victoria/361/2011 virus, respectively, WV successfully shortened the duration of virus shedding of both challenge viruses, whereas SV shortened only that of the homologous virus, A/Victoria/361/2011 (IVR-165). When WV-immunized ferrets were challenged with A/Fukushima/69/2015 virus, which is an epidemic virus antigenically different from the A/Victoria/361/2011 virus, WV could shorten the duration of shedding of this virus. In addition, we found that early induction of nasal IgG and IgA antibodies by vaccines helped shorten the virus-shedding period, although this was dependent on the degree of difference in antigenicity of the challenge virus. These results indicate that vaccination with WV, not with SV, would be a solution to avoid decreased vaccine effectiveness due to the antigenic change of HGR virus by egg adaptation during virus propagation.
Collapse
Affiliation(s)
- Noriko Kishida
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Masaki Imai
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Akira Ainai
- Department of PathologyNational Institute of Infectious DiseasesTokyoJapan
| | - Hideki Asanuma
- Laboratory of Vaccine Seed Virus DevelopmentResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Reiko Saito
- Department of Public HealthNiigata University School of MedicineNiigataJapan
| | - Seiichiro Fujisaki
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Masayuki Shirakura
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Kazuya Nakamura
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Tomoko Kuwahara
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Emi Takashita
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Masato Tashiro
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Takato Odagiri
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| | - Shinji Watanabe
- Laboratory of Influenza Virus SurveillanceResearch Center for Influenza and Respiratory Viruses, National Institute of Infectious DiseasesTokyoJapan
| |
Collapse
|
2
|
Harada Y, Takahashi H, Fujimoto T, Horikoshi F, Chida S, Tanaka K, Minari K, Tanimoto Y, Fujisaki S, Miura H, Nakauchi M, Shimasaki N, Suzuki Y, Arita T, Hamamoto I, Yamamoto N, Hasegawa H, Odagiri T, Tashiro M, Nobusawa E. Evaluation of a qualified MDCK cell line for virus isolation to develop cell-based influenza vaccine viruses with appropriate antigenicity. Vaccine 2024; 42:126242. [PMID: 39213922 DOI: 10.1016/j.vaccine.2024.126242] [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: 04/06/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
We established a qualified Madin-Darby canine kidney cell line (qMDCK-Cs) and investigated its suitability for source virus isolation to develop cell-based seasonal influenza vaccine viruses using vaccine manufacturer cells (Manuf-Cs). When inoculated with 81 influenza-positive clinical specimens, the initial virus isolation efficiency of qMDCK-Cs was exceeded 70%. Among the qMDCK-C isolates, 100% of the A/H1N1pdm09, B/Victoria and B/Yamagata strains and >70% of the A/H3N2 strains showed antigenicity equivalent to that of the contemporary vaccine or relevant viruses in haemagglutination inhibition (HI) or virus neutralization (VN) tests using ferret antisera. These qMDCK-C isolates were propagated in Manuf-Cs (MDCK and Vero cells) (Manuf-C viruses) to develop vaccine viruses. In reciprocal antigenicity tests, ferret antisera raised against corresponding reference viruses and Manuf-C viruses recognized 29 of 31 Manuf-C viruses and corresponding reference viruses, respectively at HI or VN titres more than half of the homologous virus titres, which is the antigenicity criterion for cell culture seasonal influenza vaccine viruses specified by the World Health Organization. Furthermore, ferret antisera against these Manuf-C viruses recognized ≥95% of the viruses circulating during the relevant influenza season with HI or VN titres greater than one-quarter of the homologous virus titres. No cell line-specific amino acid substitutions were observed in the resulting viruses. However, polymorphisms at positions 158/160 of H3HA, 148/151 of N2NA and 197/199 of B/Victoria HA were occasionally detected in the qMDCK-C and Manuf-C viruses but barely affected the viral antigenicity. These results indicated that qMDCK-Cs are suitable for isolating influenza viruses that can serve as a source of antigenically appropriate vaccine viruses. The use of the qMDCK-C isolates will eliminates the need for clinical sample collection, virus isolation, and antigenicity analysis every season, and is expected to contribute to the promotion of vaccine virus development using manufacturer cells.
Collapse
Affiliation(s)
- Yuichi Harada
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan; Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Hitoshi Takahashi
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Takao Fujimoto
- BIKEN CO., Ltd., 4-1-70, Seto-Cho, Kan-Onji, Kagawa 768-0065, Japan
| | | | - Shuhei Chida
- BIKEN CO., Ltd., 4-1-70, Seto-Cho, Kan-Onji, Kagawa 768-0065, Japan
| | - Kenji Tanaka
- Daiichi Sankyo Biotech Co., Ltd., 6-111 Arai, Kitamoto City, Saitama Prefecture 364-0026, Japan
| | - Kenji Minari
- Takeda Pharmaceutical Company Limited, Hikari Plant, 4720 Takeda, Mitsui, Hikari City, Yamaguchi 743-8502, Japan
| | - Yoshimi Tanimoto
- Takeda Pharmaceutical Company Limited, Hikari Plant, 4720 Takeda, Mitsui, Hikari City, Yamaguchi 743-8502, Japan
| | - Seiichiro Fujisaki
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Hideka Miura
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Mina Nakauchi
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Noriko Shimasaki
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan; Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Yasushi Suzuki
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Tomoko Arita
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Itsuki Hamamoto
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Norio Yamamoto
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Hideki Hasegawa
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Takato Odagiri
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Masato Tashiro
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Eri Nobusawa
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan.
| |
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
|
Chua BY, Sekiya T, Koutsakos M, Nomura N, Rowntree LC, Nguyen THO, McQuilten HA, Ohno M, Ohara Y, Nishimura T, Endo M, Itoh Y, Habel JR, Selva KJ, Wheatley AK, Wines BD, Hogarth PM, Kent SJ, Chung AW, Jackson DC, Brown LE, Shingai M, Kedzierska K, Kida H. Immunization with inactivated whole virus particle influenza virus vaccines improves the humoral response landscape in cynomolgus macaques. PLoS Pathog 2022; 18:e1010891. [PMID: 36206307 PMCID: PMC9581423 DOI: 10.1371/journal.ppat.1010891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/19/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Although antibody-inducing split virus vaccines (SV) are currently the most effective way to combat seasonal influenza, their efficacy can be modest, especially in immunologically-naïve individuals. We investigated immune responses towards inactivated whole influenza virus particle vaccine (WPV) formulations, predicated to be more immunogenic, in a non-human primate model, as an important step towards clinical testing in humans. Comprehensive analyses were used to capture 46 immune parameters to profile how WPV-induced responses differed to those elicited by antigenically-similar SV formulations. Naïve cynomolgus macaques vaccinated with either monovalent or quadrivalent WPV consistently induced stronger antibody responses and hemagglutination inhibition (HI) antibody titres against vaccine-matched viruses compared to SV formulations, while acute reactogenic effects were similar. Responses in WPV-primed animals were further increased by boosting with the same formulation, conversely to modest responses after priming and boosting with SV. 28-parameter multiplex bead array defined key antibody features and showed that while both WPV and SV induced elevated IgG responses against A/H1N1 nucleoprotein, only WPV increased IgG responses against A/H1N1 hemagglutinin (HA) and HA-Stem, and higher IgA responses to A/H1N1-HA after each vaccine dose. Antibodies to A/H1N1-HA and HA-Stem that could engage FcγR2a and FcγR3a were also present at higher levels after one dose of WPV compared to SV and remained elevated after the second dose. Furthermore, WPV-enhanced antibody responses were associated with higher frequencies of HA-specific B-cells and IFN-γ-producing CD4+ T-cell responses. Our data additionally demonstrate stronger boosting of HI titres by WPV following prior infection and support WPV administered as a priming dose irrespective of the follow up vaccine for the second dose. Our findings thus show that compared to SV vaccination, WPV-induced humoral responses are significantly increased in scope and magnitude, advocating WPV vaccination regimens for priming immunologically-naïve individuals and also in the event of a pandemic outbreak.
Collapse
Affiliation(s)
- Brendon Y. Chua
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Toshiki Sekiya
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Marios Koutsakos
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Naoki Nomura
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Thi H. O. Nguyen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hayley A. McQuilten
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Marumi Ohno
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | | | | | | | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Jennifer R. Habel
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Bruce D. Wines
- Immune Therapies Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
- Department of Pathology, The University of Melbourne, Parkville, Australia
| | - P. Mark Hogarth
- Immune Therapies Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
- Department of Pathology, The University of Melbourne, Parkville, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David C. Jackson
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Lorena E. Brown
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Masashi Shingai
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Katherine Kedzierska
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hiroshi Kida
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE) Hokkaido University, Sapporo, Japan
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| |
Collapse
|
5
|
Fei C, Gao J, Fei C, Ma L, Zhu W, He L, Wu Y, Song S, Li W, Zhou J, Liao G. A flow-through chromatography purification process for Vero cell-derived influenza virus (H7N9). J Virol Methods 2021; 301:114408. [PMID: 34896455 DOI: 10.1016/j.jviromet.2021.114408] [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: 10/23/2020] [Revised: 10/29/2021] [Accepted: 12/07/2021] [Indexed: 11/29/2022]
Abstract
Immunization is the most effective way to respond to an influenza epidemic. To produce Vero cell-derived influenza vaccines, a more efficient, stable and economical purification process is required. In this study, we purified the H7N9 influenza virus grown in Vero cells that were cultured in a serum-free medium by using a combination of anion exchange chromatography (AEC) and ligand-activated core chromatography (LCC), which avoids the virus capture step. After purification, 99.95 % host cell DNA (hcDNA) (final concentration: 28.69 pg/dose) and 98.87 % host cell protein (HCP) (final concentration: 28.28 ng/dose) were removed. The albumin content was 11.36 ng/dose. All these remnants met the current Chinese Pharmacopoeia and WHO requirements. The final virus recovery rate was 58.74 %, with the concentration of hemagglutinin recorded at 132.12 μg/mL. The flow-through chromatography purification process represents an alternative to the existing processes for cell-derived influenza viruses and might be suitable for the purification of other viruses as well.
Collapse
Affiliation(s)
- ChengRui Fei
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - JingXia Gao
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - ChengHua Fei
- Kunming Maternal and Child Health Hospital, 650031, China
| | - Lei Ma
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - WenYong Zhu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - LingYu He
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - YaNan Wu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - ShaoHui Song
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - WeiDong Li
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China
| | - Jian Zhou
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China.
| | - GuoYang Liao
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, 650118, China.
| |
Collapse
|
6
|
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: 14] [Impact Index Per Article: 3.5] [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
|
7
|
Scaled preparation of extracellular vesicles from conditioned media. Adv Drug Deliv Rev 2021; 177:113940. [PMID: 34419502 DOI: 10.1016/j.addr.2021.113940] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/13/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) especially of mesenchymal stem/stomal cells (MSCs) are increasingly considered as biotherapeutic agents for a variety of different diseases. For translating them effectively into the clinics, scalable production processes fulfilling good manufacturing practice (GMP) are needed. Like for other biotherapeutic agents, the manufacturing of EV products can be subdivided in the upstream and downstream processing and the subsequent quality control, each of them containing several unit operations. During upstream processing (USP), cells are isolated, stored (cell banking) and expanded; furthermore, EV-containing conditioned media are produced. During downstream processing (DSP), conditioned media (CM) are processed to obtain concentrated and purified EV products. CM are either stored until DSP or are directly processed. As first unit operation in DSP, clarification removes remaining cells, debris and other larger impurities. The key operations of each EV DSP is volume-reduction combined with purification of the concentrated EVs. Most of the EV preparation methods used in conventional research labs including differential centrifugation procedures are limited in their scalability. Consequently, it is a major challenge in the therapeutic EV field to identify appropriate EV concentration and purification methods allowing scale up. As EVs share several features with enveloped viruses, that are used for more than two decades in the clinics now, several principles can be adopted to EV manufacturing. Here, we introduce and discuss volume reducing and purification methods frequently used for viruses and analyze their value for the manufacturing of EV-based therapeutics.
Collapse
|
8
|
Dhamane S, Patil U, Smith M, Adhikari M, Nazem A, Conrad JC, Kourentzi K, Willson RC. Isocratic reporter-exclusion immunoassay using restricted-access adsorbents. Analyst 2021; 146:4835-4840. [PMID: 34198311 PMCID: PMC9798887 DOI: 10.1039/d1an00396h] [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: 12/31/2022]
Abstract
We introduce analyte-dependent exclusion of reporter reagents from restricted-access adsorbents as the basis of an isocratic reporter-exclusion immunoassay for viruses, proteins, and other analytes. Capto™ Core 700 and related resins possess a noninteracting size-selective outer layer surrounding a high-capacity nonspecific mixed-mode capture adsorbent core. In the absence of analyte, antibody-enzyme reporter conjugates can enter the adsorbent and be captured, and their signal is lost. In the presence of large or artificially-expanded analytes, reporter reagents bind to analyte species to form complexes large enough to be excluded from the adsorbent core, allowing their signal to be observed. This assay principle is demonstrated using M13 bacteriophage virus and human chorionic gonadotropin as model analytes. The simple isocratic detection approach described here allows a rapid implementation of immunoassay for detection of a wide range of analytes and uses inexpensive, generally-applicable, and stable column materials instead of costly analyte-specific immunoaffinity adsorbents.
Collapse
Affiliation(s)
- Sagar Dhamane
- Department of Biology and Biochemistry, University of Houston, Houston, TX USA
| | - Ujwal Patil
- Department of Biology and Biochemistry, University of Houston, Houston, TX USA
| | - Maxwell Smith
- Department of Chemical and Biomolecular Engineering, University of Houston, TX USA
| | - Meena Adhikari
- Department of Biology and Biochemistry, University of Houston, Houston, TX USA
| | - Ahmad Nazem
- Department of Biomedical Engineering, University of Houston, Houston, TX USA
| | - Jacinta C. Conrad
- Department of Chemical and Biomolecular Engineering, University of Houston, TX USA
| | - Katerina Kourentzi
- Department of Chemical and Biomolecular Engineering, University of Houston, TX USA
| | - Richard C. Willson
- Department of Biology and Biochemistry, University of Houston, Houston, TX USA,Department of Biomedical Engineering, University of Houston, Houston, TX USA,Department of Chemical and Biomolecular Engineering, University of Houston, TX USA,Escuela de Medicina y Ciencias de las Salud ITESM, Monterrey, Mexico,Corresponding author. (Richard C. Willson)
| |
Collapse
|
9
|
Do Minh A, Kamen AA. Critical Assessment of Purification and Analytical Technologies for Enveloped Viral Vector and Vaccine Processing and Their Current Limitations in Resolving Co-Expressed Extracellular Vesicles. Vaccines (Basel) 2021; 9:vaccines9080823. [PMID: 34451948 PMCID: PMC8402407 DOI: 10.3390/vaccines9080823] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Viral vectors and viral vaccines are invaluable tools in prevention and treatment of diseases. Many infectious diseases are controlled using vaccines designed from subunits or whole viral structures, whereas other genetic diseases and cancers are being treated by viruses used as vehicles for delivering genetic material in gene therapy or as therapeutic agents in virotherapy protocols. Viral vectors and vaccines are produced in different platforms, from traditional embryonated chicken eggs to more advanced cell cultures. All these expression systems, like most cells and cellular tissues, are known to spontaneously release extracellular vesicles (EVs). EVs share similar sizes, biophysical characteristics and even biogenesis pathways with enveloped viruses, which are currently used as key ingredients in a number of viral vectors and licensed vaccine products. Herein, we review distinctive features and similarities between EVs and enveloped viruses as we revisit the downstream processing steps and analytical technologies currently implemented to produce and document viral vector and vaccine products. Within a context of well-established viral vector and vaccine safety profiles, this review provides insights on the likely presence of EVs in the final formulation of enveloped virus products and discusses the potential to further resolve and document these components.
Collapse
|
10
|
Nucleic acid delivery with extracellular vesicles. Adv Drug Deliv Rev 2021; 173:89-111. [PMID: 33746014 DOI: 10.1016/j.addr.2021.03.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/16/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are membrane-enclosed particles, heterogeneous in size, shape, contents, biogenesis and structure. They are released by eukaryotic and prokaryotic cells and exert (patho-)physiological roles as mediators for transmitting molecular information from the producer (donor) to a recipient cell. This review focuses on the potential of EVs for delivering nucleic acids, as particularly problematic cargoes with regard to stability/protection and uptake efficacy. It highlights important properties of EVs for nucleic acid delivery and discusses their physiological and pathophysiological roles with regard to various cellular RNA species. It then describes the application of EVs for delivering a broad selection of nucleic acids/oligonucleotides, in particular giving a comprehensive overview of preclinical in vivo studies and the various strategies explored. In this context, different techniques for EV loading are discussed, as well as other important technical aspects related to EV preparation, characterization and in particular, the various approaches of artificial EV modification.
Collapse
|
11
|
Sekiya T, Ohno M, Nomura N, Handabile C, Shingai M, Jackson DC, Brown LE, Kida H. Selecting and Using the Appropriate Influenza Vaccine for Each Individual. Viruses 2021; 13:971. [PMID: 34073843 PMCID: PMC8225103 DOI: 10.3390/v13060971] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022] Open
Abstract
Despite seasonal influenza vaccines having been routinely used for many decades, influenza A virus continues to pose a global threat to humans, causing high morbidity and mortality each year. The effectiveness of the vaccine is largely dependent on how well matched the vaccine strains are with the circulating influenza virus strains. Furthermore, low vaccine efficacy in naïve populations such as young children, or in the elderly, who possess weakened immune systems, indicates that influenza vaccines need to be more personalized to provide broader community protection. Advances in both vaccine technologies and our understanding of influenza virus infection and immunity have led to the design of a variety of alternate vaccine strategies to extend population protection against influenza, some of which are now in use. In this review, we summarize the progress in the field of influenza vaccines, including the advantages and disadvantages of different strategies, and discuss future prospects. We also highlight some of the challenges to be faced in the ongoing effort to control influenza through vaccination.
Collapse
Affiliation(s)
- Toshiki Sekiya
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Marumi Ohno
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
| | - Naoki Nomura
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
| | - Chimuka Handabile
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
| | - Masashi Shingai
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
| | - David C. Jackson
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Lorena E. Brown
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Hiroshi Kida
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- Collaborating Research Center for the Control of Infectious Diseases, Nagasaki University, Nagasaki 852-8521, Japan
| |
Collapse
|
12
|
Dos Santos Morais R, El-Kirat-Chatel S, Burgain J, Simard B, Barrau S, Paris C, Borges F, Gaiani C. A Fast, Efficient and Easy to Implement Method to Purify Bacterial Pili From Lacticaseibacillus rhamnosus GG Based on Multimodal Chromatography. Front Microbiol 2020; 11:609880. [PMID: 33391233 PMCID: PMC7775309 DOI: 10.3389/fmicb.2020.609880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/24/2020] [Indexed: 01/02/2023] Open
Abstract
Pili are polymeric proteins located at the cell surface of bacteria. These filamentous proteins play a pivotal role in bacterial adhesion with the surrounding environment. They are found both in Gram-negative and Gram-positive bacteria but differ in their structural organization. Purifying these high molecular weight proteins is challenging and has certainly slowed down their characterization. Here, we propose a chromatography-based protocol, mainly relying on multimodal chromatography (core bead technology using Capto Core 700 resin), to purify sortase-dependent SpaCBA pili from the probiotic strain Lacticaseibacillus rhamnosus GG (LGG). Contrary to previously published methods, this purification protocol does not require specific antibodies nor complex laboratory equipment, including for the multimodal chromatography step, and provides high degree of protein purity. No other proteins were detectable by SDS-PAGE and the 260/280 nm ratio (∼0.6) of the UV spectrum confirmed the absence of any other co-purified macromolecules. One can obtain ∼50 μg of purified pili, starting from 1 L culture at OD600nm ≈ 1, in 2–3 working days. This simple protocol could be useful to numerous laboratories to purify pili from LGG easily. Therefore, the present work should boost specific studies dedicated to LGG SpaCBA pili and the characterization of the interactions occurring with their protein partners at the molecular level. Moreover, this straightforward purification process might be extended to the purification of sortase-dependant pili from other Gram-positive bacteria.
Collapse
Affiliation(s)
| | - Sofiane El-Kirat-Chatel
- Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564, CNRS-Université de Lorraine, Nancy, France
| | - Jennifer Burgain
- Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine, Nancy, France
| | - Blandine Simard
- Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine, Nancy, France
| | - Sarah Barrau
- Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine, Nancy, France
| | - Cédric Paris
- Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine, Nancy, France
| | - Frédéric Borges
- Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine, Nancy, France
| | - Claire Gaiani
- Laboratoire d'Ingénierie des Biomolécules, Université de Lorraine, Nancy, France.,Institut Universitaire de France, Parris, France
| |
Collapse
|
13
|
McNamara RP, Dittmer DP. Modern Techniques for the Isolation of Extracellular Vesicles and Viruses. J Neuroimmune Pharmacol 2020. [PMID: 31512168 DOI: 10.1007/s11481-%20019-09874-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Extracellular signaling is pivotal to maintain organismal homeostasis. A quickly emerging field of interest within extracellular signaling is the study of extracellular vesicles (EV), which act as messaging vehicles for nucleic acids, proteins, metabolites, lipids, etc. from donor cells to recipient cells. This transfer of biologically active material within a vesicular body is similar to the infection of a cell through a virus particle, which transfers genetic material from one cell to another to preserve an infection state, and viruses are known to modulate EV. Although considerable heterogeneity exists within EV and viruses, this review focuses on those that are small (< 200 nm in diameter) and of relatively low density (< 1.3 g/mL). A multitude of isolation methods for EV and virus particles exist. In this review, we present an update on methods for their isolation, purification, and phenotypic characterization. We hope that the information we provide will be of use to basic science and clinical investigators, as well as biotechnologists in this emerging field. Graphical Abstract.
Collapse
Affiliation(s)
- Ryan P McNamara
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
14
|
Junter GA, Lebrun L. Polysaccharide-based chromatographic adsorbents for virus purification and viral clearance. J Pharm Anal 2020; 10:291-312. [PMID: 32292625 PMCID: PMC7104128 DOI: 10.1016/j.jpha.2020.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/20/2022] Open
Abstract
Viruses still pose a significant threat to human and animal health worldwide. In the fight against viral infections, high-purity viral stocks are needed for manufacture of safer vaccines. It is also a priority to ensure the viral safety of biopharmaceuticals such as blood products. Chromatography techniques are widely implemented at both academic and industrial levels in the purification of viral particles, whole viruses and virus-like particles to remove viral contaminants from biopharmaceutical products. This paper focuses on polysaccharide adsorbents, particulate resins and membrane adsorbers, used in virus purification/removal chromatography processes. Different chromatographic modes are surveyed, with particular attention to ion exchange and affinity/pseudo-affinity adsorbents among which commercially available agarose-based resins (Sepharose®) and cellulose-based membrane adsorbers (Sartobind®) occupy a dominant position. Mainly built on the development of new ligands coupled to conventional agarose/cellulose matrices, the development perspectives of polysaccharide-based chromatography media in this antiviral area are stressed in the conclusive part.
Collapse
Affiliation(s)
- Guy-Alain Junter
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000, Rouen, France
| | - Laurent Lebrun
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000, Rouen, France
| |
Collapse
|
15
|
Sánchez-Trasviña C, Fuks P, Mushagasha C, Kimerer L, Mayolo-Deloisa K, Rito-Palomares M, Carta G. Structure and functional properties of Capto™ Core 700 core-shell particles. J Chromatogr A 2020; 1621:461079. [DOI: 10.1016/j.chroma.2020.461079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 01/13/2023]
|
16
|
Fabrication of macroporous microspheres with core-shell structure for negative chromatography purification of virus. J Chromatogr A 2020; 1610:460578. [DOI: 10.1016/j.chroma.2019.460578] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022]
|
17
|
McNamara RP, Dittmer DP. Modern Techniques for the Isolation of Extracellular Vesicles and Viruses. J Neuroimmune Pharmacol 2019; 15:459-472. [PMID: 31512168 DOI: 10.1007/s11481-019-09874-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
Extracellular signaling is pivotal to maintain organismal homeostasis. A quickly emerging field of interest within extracellular signaling is the study of extracellular vesicles (EV), which act as messaging vehicles for nucleic acids, proteins, metabolites, lipids, etc. from donor cells to recipient cells. This transfer of biologically active material within a vesicular body is similar to the infection of a cell through a virus particle, which transfers genetic material from one cell to another to preserve an infection state, and viruses are known to modulate EV. Although considerable heterogeneity exists within EV and viruses, this review focuses on those that are small (< 200 nm in diameter) and of relatively low density (< 1.3 g/mL). A multitude of isolation methods for EV and virus particles exist. In this review, we present an update on methods for their isolation, purification, and phenotypic characterization. We hope that the information we provide will be of use to basic science and clinical investigators, as well as biotechnologists in this emerging field. Graphical Abstract.
Collapse
Affiliation(s)
- Ryan P McNamara
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
18
|
Thorne B, Takeya R, Vitelli F, Swanson X. Gene Therapy. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:351-399. [PMID: 28289769 DOI: 10.1007/10_2016_53] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gene therapy refers to a rapidly growing field of medicine in which genes are introduced into the body to treat or prevent diseases. Although a variety of methods can be used to deliver the genetic materials into the target cells and tissues, modified viral vectors represent one of the more common delivery routes because of its transduction efficiency for therapeutic genes. Since the introduction of gene therapy concept in the 1970s, the field has advanced considerably with notable clinical successes being demonstrated in many clinical indications in which no standard treatment options are currently available. It is anticipated that the clinical success the field observed in recent years can drive requirements for more scalable, robust, cost effective, and regulatory-compliant manufacturing processes. This review provides a brief overview of the current manufacturing technologies for viral vectors production, drawing attention to the common upstream and downstream production process platform that is applicable across various classes of viral vectors and their unique manufacturing challenges as compared to other biologics. In addition, a case study of an industry-scale cGMP production of an AAV-based gene therapy product performed at 2,000 L-scale is presented. The experience and lessons learned from this largest viral gene therapy vector production run conducted to date as discussed and highlighted in this review should contribute to future development of commercial viable scalable processes for vial gene therapies.
Collapse
Affiliation(s)
- Barb Thorne
- Thorne Bio-Consulting LLC, Sammamish, WA, USA
| | | | | | | |
Collapse
|
19
|
Pato TP, Souza MC, Mattos DA, Caride E, Ferreira DF, Gaspar LP, Freire MS, Castilho LR. Purification of yellow fever virus produced in Vero cells for inactivated vaccine manufacture. Vaccine 2019; 37:3214-3220. [DOI: 10.1016/j.vaccine.2019.04.077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 11/16/2022]
|
20
|
McNamara RP, Caro-Vegas CP, Costantini LM, Landis JT, Griffith JD, Damania BA, Dittmer DP. Large-scale, cross-flow based isolation of highly pure and endocytosis-competent extracellular vesicles. J Extracell Vesicles 2018; 7:1541396. [PMID: 30533204 PMCID: PMC6282418 DOI: 10.1080/20013078.2018.1541396] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/02/2018] [Accepted: 10/12/2018] [Indexed: 12/15/2022] Open
Abstract
Isolation of extracellular vesicles (EVs) from cell culture supernatant or plasma can be accomplished in a variety of ways. Common measures to quantify relative success are: concentration of the EVs, purity from non-EVs associated protein, size homogeneity and functionality of the final product. Here, we present an industrial-scale workflow for isolating highly pure and functional EVs using cross-flow based filtration coupled with high-molecular weight Capto Core size exclusion. Through this combination, EVs loss is kept to a minimum. It outperforms other isolation procedures based on a number of biochemical and biophysical assays. Moreover, EVs isolated through this method can be further concentrated down or directly immunopurified to obtain discreet populations of EVs. From our results, we propose that cross-flow/Capto Core isolation is a robust method of purifying highly concentrated, homogenous, and functionally active EVs from industrial-scale input volumes with few contaminants relative to other methods.
Collapse
Affiliation(s)
- Ryan P. McNamara
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Carolina P. Caro-Vegas
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Lindsey M. Costantini
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Justin T. Landis
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Jack D. Griffith
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Blossom A. Damania
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Dirk P. Dittmer
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
21
|
Hindered diffusion of proteins in mixture adsorption on porous anion exchangers and impact on flow-through purification of large proteins. J Chromatogr A 2018; 1585:121-130. [PMID: 30503698 DOI: 10.1016/j.chroma.2018.11.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/15/2018] [Accepted: 11/22/2018] [Indexed: 01/21/2023]
Abstract
Complex adsorption kinetics behaviors of proteins in mixtures hampers chromatographic process development and complicates model-based prediction of separation. We investigated the adsorption characteristics of mixtures comprised of a larger protein (secretory immunoglobulins or thyroglobulin) and a smaller protein (serum albumin or green fluorescence protein) on the small-pore anion exchanger Q Sepharose FF. Confocal laser scanning microscopy measurements revealed that binding of the large protein was extremely slow and eventually stopped completely after the adsorption front penetrated just a few μm into the particle. Binding capacities after 24 h of incubation were nevertheless around 35 mg/mL of particle which is relatively high when considering that only a fraction of the particle was saturated, suggesting that locally-high bound protein concentrations are attained in a layer close to the particle surface. During mixture adsorption, the bound protein layer also significantly hindered diffusion of the smaller proteins into the particles resulting in about three times slower adsorption kinetics compared to single component adsorption. The combined effects of restricted diffusion and protein binding explain why flow-through purification of these mixtures with the small-pore resin Q Sepharose FF is effective under practical conditions. In this resin, diffusion of secretory immunoglobulins (or thyroglobulin) is restricted in the small pores so that despite their intrinsically greater affinity for the resin, much less binds compared to small proteins. Using the large-pore resin POROS 50 HQ results in faster transport, but also in more binding of secretory immunoglobulins (or thyroglobulin) compared to smaller protein impurities, preventing effective flow-through purification.
Collapse
|
22
|
Zaveckas M, Goda K, Ziogiene D, Gedvilaite A. Purification of recombinant trichodysplasia spinulosa–associated polyomavirus VP1-derived virus-like particles using chromatographic techniques. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1090:7-13. [DOI: 10.1016/j.jchromb.2018.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/27/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
|
23
|
Ng HW, Lee MFX, Chua GK, Gan BK, Tan WS, Ooi CW, Tang SY, Chan ES, Tey BT. Size-selective purification of hepatitis B virus-like particle in flow-through chromatography: Types of ion exchange adsorbent and grafted polymer architecture. J Sep Sci 2018; 41:2119-2129. [PMID: 29427396 DOI: 10.1002/jssc.201700823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 01/08/2018] [Accepted: 01/26/2018] [Indexed: 11/09/2022]
Abstract
Hepatitis B virus-like particles expressed in Escherichia coli were purified using anion exchange adsorbents grafted with polymer poly(oligo(ethylene glycol) methacrylate) in flow-through chromatography mode. The virus-like particles were selectively excluded, while the relatively smaller sized host cell proteins were absorbed. The exclusion of virus-like particles was governed by the accessibility of binding sites (the size of adsorbents and the charge of grafted dextran chains) as well as the architecture (branch-chain length) of the grafted polymer. The branch-chain length of grafted polymer was altered by changing the type of monomers used. The larger adsorbent (90 μm) had an approximately twofold increase in the flow-through recovery, as compared to the smaller adsorbent (30 μm). Generally, polymer-grafted adsorbents improved the exclusion of the virus-like particles. Overall, the middle branch-chain length polymer grafted on larger adsorbent showed optimal performance at 92% flow-through recovery with a purification factor of 1.53. A comparative study between the adsorbent with dextran grafts and the polymer-grafted adsorbent showed that a better exclusion of virus-like particles was achieved with the absorbent grafted with inert polymer. The grafted polymer was also shown to reduce strong interaction between binding sites and virus-like particles, which preserved the particles' structure.
Collapse
Affiliation(s)
- Hon Wei Ng
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Selangor, Malaysia
| | - Micky Fu Xiang Lee
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Selangor, Malaysia
| | - Gek Kee Chua
- Faculty of Chemical and Natural Resources Engineering, Universiti Malaysia Pahang, Pahang, Malaysia
| | - Bee Koon Gan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Selangor, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
| | - Chien Wei Ooi
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Selangor, Malaysia
| | - Siah Ying Tang
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Selangor, Malaysia
| | - Eng Seng Chan
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Selangor, Malaysia.,Advanced Engineering Platform, Monash University Malaysia, Selangor, Malaysia
| | - Beng Ti Tey
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Selangor, Malaysia.,Advanced Engineering Platform, Monash University Malaysia, Selangor, Malaysia
| |
Collapse
|
24
|
Chua BY, Sekiya T, Jackson DC. Opinion: Making Inactivated and Subunit-Based Vaccines Work. Viral Immunol 2018; 31:150-158. [PMID: 29369750 DOI: 10.1089/vim.2017.0146] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Empirically derived vaccines have in the past relied on the isolation and growth of disease-causing microorganisms that are then inactivated or attenuated before being administered. This is often done without prior knowledge of the mechanisms involved in conferring protective immunity. Recent advances in scientific technologies and in our knowledge of how protective immune responses are induced enable us to rationally design novel and safer vaccination strategies. Such advances have accelerated the development of inactivated whole-organism- and subunit-based vaccines. In this review, we discuss ideal attributes and criteria that need to be considered for the development of vaccines and some existing vaccine platforms. We focus on inactivated vaccines against influenza virus and ways by which vaccine efficacy can be improved with the use of adjuvants and Toll-like receptor-2 signaling.
Collapse
Affiliation(s)
- Brendon Y Chua
- 1 Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, Australia .,2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| | - Toshiki Sekiya
- 2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| | - David C Jackson
- 1 Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, Australia .,2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| |
Collapse
|
25
|
Corso G, Mäger I, Lee Y, Görgens A, Bultema J, Giebel B, Wood MJA, Nordin JZ, Andaloussi SE. Reproducible and scalable purification of extracellular vesicles using combined bind-elute and size exclusion chromatography. Sci Rep 2017; 7:11561. [PMID: 28912498 PMCID: PMC5599601 DOI: 10.1038/s41598-017-10646-x] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/11/2017] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) play a pivotal role in cell-to-cell communication and have been shown to take part in several physiological and pathological processes. EVs have traditionally been purified by ultracentrifugation (UC), however UC has limitations, including resulting in, operator-dependant yields, EV aggregation and altered EV morphology, and moreover is time consuming. Here we show that commercially available bind-elute size exclusion chromatography (BE-SEC) columns purify EVs with high yield (recovery ~ 80%) in a time-efficient manner compared to current methodologies. This technique is reproducible and scalable, and surface marker analysis by bead-based flow cytometry revealed highly similar expression signatures compared with UC-purified samples. Furthermore, uptake of eGFP labelled EVs in recipient cells was comparable between BE-SEC and UC samples. Hence, the BE-SEC based EV purification method represents an important methodological advance likely to facilitate robust and reproducible studies of EV biology and therapeutic application.
Collapse
Affiliation(s)
- Giulia Corso
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Institute of Technology, University of Tartu, Tartu, Estonia
| | - Yi Lee
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - André Görgens
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jarred Bultema
- Department of Chemistry and Biochemistry, University of Colorado, Colorado Springs, USA
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Evox Therapeutics, King Charles House, Park End Street, Oxford, United Kingdom
| | - Joel Z Nordin
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden. .,Evox Therapeutics, King Charles House, Park End Street, Oxford, United Kingdom.
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden. .,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom. .,Evox Therapeutics, King Charles House, Park End Street, Oxford, United Kingdom.
| |
Collapse
|
26
|
A fast and efficient purification platform for cell-based influenza viruses by flow-through chromatography. Vaccine 2017; 36:3146-3152. [PMID: 28342667 DOI: 10.1016/j.vaccine.2017.03.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 03/03/2017] [Accepted: 03/08/2017] [Indexed: 01/01/2023]
Abstract
Since newly emerging influenza viruses with pandemic potentials occurred in recent years, the demand for producing pandemic influenza vaccines for human use is high. For the development of a quick and efficient vaccine production, we proposed an efficient purification platform from the harvest to the purified bulk for the cell-based influenza vaccine production. This platform based on flow-through chromatography and filtration steps and the process only involves a few purification steps, including depth filtration, inactivation by formaldehyde, microfiltration, ultrafiltration, anion-exchange and ligand-core chromatography and sterile filtration. In addition, in the proposed chromatography steps, no virus capture steps were employed, and the purification results were not affected by the virus strain variation, host cells and culturing systems. The results from different virus strains which produced by Vero or MDCK cells in different culturing systems also obtained 33-46% HA recovery yields by this platform. The overall removal rates of the protein and DNA concentration in the purified bulk were over 96.1% and 99.7%, respectively. The low residual cellular DNA concentrations were obtained ranged from 30 to 130pg per human dose (15µg/dose). All influenza H5N1 purified bulks met the regulatory requirements for human vaccine use.
Collapse
|
27
|
James KT, Cooney B, Agopsowicz K, Trevors MA, Mohamed A, Stoltz D, Hitt M, Shmulevitz M. Novel High-throughput Approach for Purification of Infectious Virions. Sci Rep 2016; 6:36826. [PMID: 27827454 PMCID: PMC5101806 DOI: 10.1038/srep36826] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/21/2016] [Indexed: 01/02/2023] Open
Abstract
Viruses are extensively studied as pathogens and exploited as molecular tools and therapeutic agents. Existing methods to purify viruses such as gradient ultracentrifugation or chromatography have limitations, for example demand for technical expertise or specialized equipment, high time consumption, and restricted capacity. Our laboratory explores mutations in oncolytic reovirus that could improve oncolytic activity, and makes routine use of numerous virus variants, genome reassortants, and reverse engineered mutants. Our research pace was limited by the lack of high-throughput virus purification methods that efficiently remove confounding cellular contaminants such as cytokines and proteases. To overcome this shortcoming, we evaluated a commercially available resin (Capto Core 700) that captures molecules smaller than 700 kDa. Capto. Core 700 chromatography produced virion purity and infectivity indistinguishable from CsCl density gradient ultracentrifugation as determined by electron microscopy, gel electrophoresis analysis and plaque titration. Capto Core 700 resin was then effectively adapted to a rapid in-slurry pull-out approach for high-throughput purification of reovirus and adenovirus. The in-slurry purification approach offered substantially increased virus purity over crude cell lysates, media, or high-spin preparations and would be especially useful for high-throughput virus screening applications where density gradient ultracentrifugation is not feasible.
Collapse
Affiliation(s)
- Kevin T James
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Brad Cooney
- GE Healthcare, LifeSciences, Marlborough, MA, US
| | - Kate Agopsowicz
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Mary Ann Trevors
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Adil Mohamed
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Don Stoltz
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mary Hitt
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Maya Shmulevitz
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
28
|
Mundle ST, Kishko M, Groppo R, DiNapoli J, Hamberger J, McNeil B, Kleanthous H, Parrington M, Zhang L, Anderson SF. Core bead chromatography for preparation of highly pure, infectious respiratory syncytial virus in the negative purification mode. Vaccine 2016; 34:3690-6. [PMID: 27238375 DOI: 10.1016/j.vaccine.2016.04.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 12/01/2022]
Abstract
Respiratory syncytial virus (RSV) is an important human pathogen, and is the most frequent viral cause of severe respiratory disease in infants. In addition, it is increasingly being recognized as an important cause of respiratory disease in the elderly and immunocompromised. Although a passive prophylactic treatment does exist for high-risk neonates and children, the overall disease burden warrants the development of a safe and effective prophylactic vaccine for use in otherwise healthy newborns and children. RSV is known to be an extremely labile virus, prone to aggregation and loss of infectious titer during virus handling and preparation procedures. To date infective RSV virions have been prepared by methods which are not readily scalable, such as density gradient ultracentrifugation. In this study we describe a scalable, chromatography-based purification procedure for preparation of highly pure, infectious RSV. The purification scheme is based on core bead technology and hollow fiber tangential flow filtration (TFF) and results in a ∼60% recovery of infectious virus titer. This method can be used to prepare highly purified wild type or live-attenuated vaccine strain viruses with titers as high as 1×10(8) plaque forming units per mL. A live-attenuated RSV vaccine prepared by this method was found to be immunogenic and protective in vivo, and its purity was 50-200-fold greater with respect to host cell dsDNA and Vero host cell proteins, than the raw feed stream. The results presented here can be considered a starting point for downstream process development of a live-attenuated vaccine approach for prevention of disease by RSV.
Collapse
Affiliation(s)
- Sophia T Mundle
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA.
| | - Michael Kishko
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Rachel Groppo
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Joshua DiNapoli
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - John Hamberger
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Bryan McNeil
- Sanofi Pasteur, ARD North America, 1755 Steeles Avenue West, Toronto, ON M2R 3T4, Canada
| | - Harry Kleanthous
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Mark Parrington
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Linong Zhang
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Stephen F Anderson
- Sanofi Pasteur, Discovery North America, 38 Sidney Street, Cambridge, MA 02139, USA
| |
Collapse
|
29
|
Lee MFX, Chan ES, Tan WS, Tam KC, Tey BT. Negative chromatography of hepatitis B virus-like particle: Comparative study of different adsorbent designs. J Chromatogr A 2016; 1445:1-9. [DOI: 10.1016/j.chroma.2016.03.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/09/2016] [Accepted: 03/22/2016] [Indexed: 02/06/2023]
|
30
|
Cunha B, Silva RJS, Aguiar T, Serra M, Daicic J, Maloisel JL, Clachan J, Åkerblom A, Carrondo MJT, Peixoto C, Alves PM. Improving washing strategies of human mesenchymal stem cells using negative mode expanded bed chromatography. J Chromatogr A 2015; 1429:292-303. [PMID: 26739915 DOI: 10.1016/j.chroma.2015.12.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/16/2015] [Accepted: 12/17/2015] [Indexed: 12/15/2022]
Abstract
The use of human mesenchymal stem cells (hMSC) in clinical applications has been increasing over the last decade. However, to be applied in a clinical setting hMSC need to comply with specific requirements in terms of identity, potency and purity. This study reports the improvement of established tangential flow filtration (TFF)-based washing strategies, further increasing hMSC purity, using negative mode expanded bed adsorption (EBA) chromatography with a new multimodal prototype matrix based on core-shell bead technology. The matrix was characterized and a stable, expanded bed could be obtained using standard equipment adapted from what is used for conventional packed bed chromatography processes. The effect of different expansion rates on cell recovery yield and protein removal capacity was assessed. The best trade-off between cell recovery (89%) and protein clearance (67%) was achieved using an intermediate expansion bed rate (1.4). Furthermore, we also showed that EBA chromatography can be efficiently integrated on the already established process for the downstream processing (DSP) of hMSC, where it improved the washing efficiency more than 10-fold, recovering approximately 70% of cells after global processing. This strategy showed not to impact cell viability (>95%), neither hMSC's characteristics in terms of morphology, immunophenotype, proliferation, adhesion capacity and multipotent differentiation potential.
Collapse
Affiliation(s)
- Bárbara Cunha
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Ricardo J S Silva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Tiago Aguiar
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Margarida Serra
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - John Daicic
- GE Healthcare Bio-Sciences AB, Björkgatan 30, SE-751 84 Uppsala, Sweden
| | - Jean-Luc Maloisel
- GE Healthcare Bio-Sciences AB, Björkgatan 30, SE-751 84 Uppsala, Sweden
| | - John Clachan
- GE Healthcare Bio-Sciences AB, Björkgatan 30, SE-751 84 Uppsala, Sweden
| | - Anna Åkerblom
- GE Healthcare Bio-Sciences AB, Björkgatan 30, SE-751 84 Uppsala, Sweden
| | - Manuel J T Carrondo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Monte da Caparica, Portugal
| | - Cristina Peixoto
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
| | - Paula M Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| |
Collapse
|
31
|
Nestola P, Peixoto C, Silva RRJS, Alves PM, Mota JPB, Carrondo MJT. Improved virus purification processes for vaccines and gene therapy. Biotechnol Bioeng 2015; 112:843-57. [PMID: 25677990 DOI: 10.1002/bit.25545] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/05/2015] [Accepted: 01/13/2015] [Indexed: 01/10/2023]
Abstract
The downstream processing of virus particles for vaccination or gene therapy is becoming a critical bottleneck as upstream titers keep improving. Moreover, the growing pressure to develop cost-efficient processes has brought forward new downstream trains. This review aims at analyzing the state-of-the-art in viral downstream purification processes, encompassing the classical unit operations and their recent developments. Emphasis is given to novel strategies for process intensification, such as continuous or semi-continuous systems based on multicolumn technology, opening up process efficiency. Process understanding in the light of the pharmaceutical quality by design (QbD) initiative is also discussed. Finally, an outlook of the upcoming breakthrough technologies is presented.
Collapse
Affiliation(s)
- Piergiuseppe Nestola
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
| | | | | | | | | | | |
Collapse
|