Original Article

Evaluating the viral clearance ability of continuous monoclonal antibody purification steps, in order to inactivate and/or remove four model viruses

Abstract

Background and Objectives: Viral clearance studies are an essential part of a manufacturer's plan to ensure the safety of an injectable biologic product. In this way, viral safety is a critical quality attribute for biologics such as monoclonal antibodies (Mabs). Evaluation of virus purification by downstream processes is a key component of risk mitigation. In this study, the capability of continuous monoclonal antibody purification steps was evaluated in the process of instant monoclonal antibody purification in different stages of purification, and the amount of reduction or inactivation of each step was determined.
Materials and Methods: Four enveloped and non-enveloped viral models VSV, Reovirus, EMCV, and HSV1 were used for spiking in selected samples in the designated tests, to have a comprehensive examination of the ability to clear the virus such as the type of genetic material, chemical resistance, and particle size. A TCID50 and qPCR methods were used to measure viral reduction. Two cell lines, Vero (African green monkey kidney) and L929 (Mouse fibroblast) were used for 4 model viruses propagation. The steps that were evaluated included 4 steps monoclonal antibody purification; cation exchange chromatography, acidic pH treatment, affinity chromatography, and nanofiltration.
Results: The nano-filter stage showed the highest viral reduction and cation exchange chromatography showed the lowest reduction. The cumulative decrease using TCID50 is equal to 19.27 [log10] for all steps and for the qPCR method is equal to 12.47 [log10] in three steps of nano-filter, affinity chromatography, and ion exchange chromatography.
Conclusion: The overall average reduction coefficient for all four model viruses is significantly high, which indicates the high capacity of the monoclonal antibody production process in inactivating and removing viruses leads to reducing the load of all four model viruses.

1. Bethencourt V. Virus stalls genzyme plant. Nat Biotechnol 2009; 27: 681.
2. Victoria JG, Wang C, Jones MS, Jaing C, McLoughlin K, Gardner S, et al. Viral nucleic acids in live-attenuated vaccines: detection of minority variants and an adventitious virus. J Virol 2010; 84: 6033-6040.
3. Berting A, Farcet MR, Kreil TR. Virus susceptibility of Chinese hamster ovary (CHO) cells and detection of viral contaminations by adventitious agent testing. Biotechnol Bioeng 2010; 106: 598-607.
4. Kerr A, Nims R. Adventitious viruses detected in biopharmaceutical bulk harvest samples over a 10-year period. PDA J Pharm Sci Technol 2010; 64: 481-485.
5. Nims RW. Detection of adventitious viruses in biologicals - a rare occurrence. Dev Biol (Basel) 2006; 123: 153-164.
6. Anthony SJ, Darpel KE, Belaganahalli MN, Maan N, Nomikou K, Sutton G, et al. RNA segment 9 exists as a duplex concatemer in an Australian strain of epizootic haemorrhagic disease virus (EHDV): Genetic analysis and evidence for the presence of concatemers as a normal feature of orbivirus replication. Virology 2011; 420: 164-171.
7. Rubino MJ. Experiences with HEK293: a human cell line. PDA J Pharm Sci Technol 2010; 64: 392-395.
8. Barone PW, Wiebe ME, Leung JC, Hussein ITM, Keumurian FJ, Bouressa J, et al. Viral contamination in biologic manufacture and implications for emerging therapies. Nat Biotechnol 2020; 38: 563-572.
9. Chiang AWT, Li S, Kellman BP, Chattopadhyay G, Zhang Y, Kuo CC, et al. Combating viral contaminants in CHO cells by engineering innate immunity. Sci Rep 2019; 9: 8827.
10. Moody M, Alves W, Varghese J, Khan F. Mouse minute virus (MMV) contamination - a case study: detection, root cause determination, and corrective actions. PDA J Pharm Sci Technol 2011; 65: 580-588.
11. Qiu Y, Jones N, Busch M, Pan P, Keegan J, Zhou W, et al. Identification and quantitation of vesivirus 2117 particles in bioreactor fluids from infected Chinese hamster ovary cell cultures. Biotechnol Bioeng 2013; 110: 1342-1353.
12. Jayapal KP, Wlaschin KF, Hu WS, Yap MGS. Recombinant protein therapeutics from CHO cells - 20 years and counting. Chem Eng Prog 2007; 103: 40-47.
13. Li L, Wang L. Multiple Myeloma: What do we do about immunodeficiency? J Cancer 2019; 10: 1675-1684.
14. Roingeard P, Raynal P-I, Eymieux S, Blanchard E. Virus detection by transmission electron microscopy: Still useful for diagnosis and a plus for biosafety. Rev Med Virol 2019; 29(1): e2019.
15. Lopez MA Jr, Mackler RM, Yoder KE. Removal of nuclease contamination during purification of recombinant prototype foamy virus integrase. J Virol Methods 2016; 235: 134-138.
16. Lopez MA Jr, Mackler RM, Altman MP, Yoder KE. Detection and removal of nuclease contamination during purification of recombinant prototype foamy virus integrase. J Vis Exp 2017; (130): 56605.
17. Paul W Barone, Stephen Avgerinos, Rob Ballard, Audrey Brussel, Philip Clark, Chris Dowd, et al. Biopharmaceutical industry approaches to facility segregation for viral safety: an effort from the consortium on adventitious agent contamination in biomanufacturing. PDA J Pharm Sci Technol 2019;73:191-203.
18. Johnson SA, Chen S, Bolton G, Chen Q, Lute S, Fisher J, Brorson K. Virus filtration: A review of current and future practices in bioprocessing. Biotechnol Bioeng 2022; 119: 743-761.
19. Villafañe L, Forrellad MA, López MG, Garbaccio S, Garro C, Rocha RV, et al. Production of Mycobacterium bovis antigens included in recombinant occlusion bodies of Baculovirus. J Mol Microbiol Biotechnol 2019; 29: 83-90.
20. Burnouf T, Radosevich M. Nanofiltration of plasma-derived biopharmaceutical products. Haemophilia 2003; 9: 24-37.
21. PDA. Virus filtration. Technical report No. 41. PDA J Pharm Sci Technol 2005; 59(2 Suppl TR41): 8-42.
22. Aranha H, Forbes S. Viral clearance strategies for biopharmaceutical safety: part II: a multifaceted approach to process validation. Pharm Techn North Am 2001; 25: 26-31.
23. Chen Q, Chen D. Viral clearance of traditional unit operations: virus-retentive filtration. PDA J Pharm Sci Technol 2015; 69: 142-153.
24. Hastie E, Cataldi M, Marriott I, Grdzelishvili VZ. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res 2013; 176: 16-32.
25. Danthi P, Holm GH, Stehle T, Dermody TS. Reovirus receptors, cell entry, and proapoptotic signaling. Adv Exp Med Biol 2013; 790: 42-47.
26. Smith JS, Robinson NJ. Age-specific prevalence of infection with herpes simplex virus types 2 and 1: a global review. J Infect Dis 2002; 186, Suppl 1: S3-28.
27. Carocci M, Bakkali-Kassimi L. The encephalomyocarditis virus. Virulence 2012; 3: 351-367.
28. Lei C, Yang J, Hu J, Sun X. On the calculation of TCID50 for quantitation of virus infectivity. Virol Sin 2021; 36: 141-144.
29. https://www.ema.europa.eu/en/ich-guideline-q5ar2-viral-safety-evaluation-biotechnology-products-derived-cell-lines-human-animal. Note for guidance on quality of biotechnological products: viral safety evaluation of biotechnological products derived from cell lines of human or animal origin. Access Date:August,2023.
30. Hole K, Velazques-Salinas L, Clavijo A. Improvement and optimization of a multiplex real-time reverse transcription polymerase chain reaction assay for the detection and typing of Vesicular stomatitis virus. J Vet Diagn Invest 2010; 22: 428-433.
31. https://www.ema.europa.eu/en/documents/scientific-guideline/note-guidance-virus-validation-studies-design-contribution-interpretation-studies-validating_en.pdf. CPMC, Note for guidance on virus validation studies: the design, contribution and interpretation of studies validating the inactivation and removal of viruses, Access Date:August,2023.
32. Aranha H. Viral clearance strategies for biopharmaceutical safety: part I: general considerations. BioPharm 2001; 14: 28-35.
33. Goussen C, Goldstein L, Brèque C, You B, Boyer S, Bataille D, et al. Viral clearance capacity by continuous Protein A chromatography step using Sequential MultiColumn Chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1145: 122056.
34. Asper M, Hanrieder T, Quellmalz A, Mihranyan A. Removal of xenotropic murine leukemia virus by nano cellulose based filter paper. Biologicals 2015; 43: 452-456.
Files
IssueVol 15 No 5 (2023) QRcode
SectionOriginal Article(s)
DOI https://doi.org/10.18502/ijm.v15i5.13877
Keywords
Viral inactivation; Virus removal; Cation exchange chromatography; Viral clearance

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
How to Cite
1.
Rasouli-Nejad Mousavi SM, Hosseini SM, Ansari S. Evaluating the viral clearance ability of continuous monoclonal antibody purification steps, in order to inactivate and/or remove four model viruses. Iran J Microbiol. 2023;15(5):711-722.