Root Cause and Prevention of Monoclonal Antibody Disulfide Bonds Reduction During Biopharmaceutical Manufacturing Process

ZHANG Xin-di,FAN Chang-wei,SONG Xiao-qing,XU Cui-yun,HUANG Feng-jie

China Biotechnology ›› 2022, Vol. 42 ›› Issue (6) : 66-75.

PDF(1593 KB)
PDF(1593 KB)
China Biotechnology ›› 2022, Vol. 42 ›› Issue (6) : 66-75. DOI: 10.13523/j.cb.2201016

Root Cause and Prevention of Monoclonal Antibody Disulfide Bonds Reduction During Biopharmaceutical Manufacturing Process

Author information +
History +

Abstract

Monoclonal antibody disulfide bonds reduction has been a common issue in biopharmaceutical process, which could produce low-molecular weight fragments, affect product quality, and lead to decreased protein purity and stability. Moreover, it could also affect the safety and effectiveness of drugs. Antibody disulfide bonds reduction is a reversible redox reaction caused by intracellular thioredoxin system and glutathione system, and is related to the specific production process. In recent years, with the development of antibody drugs and mammalian cell culture scale, disulfide bonds reduction is observed more frequently. To solve this problem, scientists have been constantly developing mitigation strategies to ensure the product quality. In this paper, the antibody disulfide bonds structure, cause of disulfide bonds reduction and influencing factors in manufacturing process were summarized. It is focused on the prevention methods in the manufacturing process. Additionally, several feasible process analysis techniques are listed so as to provide reference for the further development of monoclonal antibody drugs in manufacturing.

Key words

Antibody / Disulfide bond / Oxidation and reduction / Process development / Process analytical technology

Cite this article

Download Citations
Xin-di ZHANG, Chang-wei FAN, Xiao-qing SONG, et al. Root Cause and Prevention of Monoclonal Antibody Disulfide Bonds Reduction During Biopharmaceutical Manufacturing Process[J]. China Biotechnology, 2022, 42(6): 66-75 https://doi.org/10.13523/j.cb.2201016

References

[1]
Kaplon H, Reichert J M. Antibodies to watch in 2021. MAbs, 2021, 13(1): 1860476.
[2]
Goydel R S, Rader C. Antibody-based cancer therapy. Oncogene, 2021, 40(21): 3655-3664.
[3]
Mullard A. FDA approves 100th monoclonal antibody product. Nature Reviews Drug Discovery, 2021, 20(7): 491-495.
[4]
Carrara S C, Ulitzka M, Grzeschik J, et al. From cell line development to the formulated drug product: the art of manufacturing therapeutic monoclonal antibodies. International Journal of Pharmaceutics, 2021, 594: 120164.
[5]
Lakbub J C, Shipman J T, Desaire H. Recent mass spectrometry-based techniques and considerations for disulfide bond characterization in proteins. Analytical and Bioanalytical Chemistry, 2018, 410(10): 2467-2484.
Disulfide bonds are important structural moieties of proteins: they ensure proper folding, provide stability, and ensure proper function. With the increasing use of proteins for biotherapeutics, particularly monoclonal antibodies, which are highly disulfide bonded, it is now important to confirm the correct disulfide bond connectivity and to verify the presence, or absence, of disulfide bond variants in the protein therapeutics. These studies help to ensure safety and efficacy. Hence, disulfide bonds are among the critical quality attributes of proteins that have to be monitored closely during the development of biotherapeutics. However, disulfide bond analysis is challenging because of the complexity of the biomolecules. Mass spectrometry (MS) has been the go-to analytical tool for the characterization of such complex biomolecules, and several methods have been reported to meet the challenging task of mapping disulfide bonds in proteins. In this review, we describe the relevant, recent MS-based techniques and provide important considerations needed for efficient disulfide bond analysis in proteins. The review focuses on methods for proper sample preparation, fragmentation techniques for disulfide bond analysis, recent disulfide bond mapping methods based on the fragmentation techniques, and automated algorithms designed for rapid analysis of disulfide bonds from liquid chromatography-MS/MS data. Researchers involved in method development for protein characterization can use the information herein to facilitate development of new MS-based methods for protein disulfide bond analysis. In addition, individuals characterizing biotherapeutics, especially by disulfide bond mapping in antibodies, can use this review to choose the best strategies for disulfide bond assignment of their biologic products. Graphical Abstract This review, describing characterization methods for disulfide bonds in proteins, focuses on three critical components: sample preparation, mass spectrometry data, and software tools.
[6]
Ren T W, Tan Z J, Ehamparanathan V, et al. Antibody disulfide bond reduction and recovery during biopharmaceutical process development- a review. Biotechnology and Bioengineering, 2021, 118(8): 2829-2844.
[7]
Wang W, Singh S, Zeng D L, et al. Antibody structure, instability, and formulation. Journal of Pharmaceutical Sciences, 2007, 96(1): 1-26.
The number of therapeutic monoclonal antibody in development has increased tremendously over the last several years and this trend continues. At present there are more than 23 approved antibodies on the US market and an estimated 200 or more are in development. Although antibodies share certain structural similarities, development of commercially viable antibody pharmaceuticals has not been straightforward because of their unique and somewhat unpredictable solution behavior. This article reviews the structure and function of antibodies and the mechanisms of physical and chemical instabilities. Various aspects of formulation development have been examined to identify the critical attributes for the stabilization of antibodies.(c) 2006 Wiley-Liss, Inc. and the American Pharmacists Association.
[8]
Liu H C, May K. Disulfide bond structures of IgG molecules: structural variations, chemical modifications and possible impacts to stability and biological function. MAbs, 2012, 4(1): 17-23.
[9]
Wang X L, Kumar S, Singh S K. Disulfide scrambling in IgG2 monoclonal antibodies: insights from molecular dynamics simulations. Pharmaceutical Research, 2011, 28(12): 3128-3144.
[10]
Moritz B, Stracke J O. Assessment of disulfide and hinge modifications in monoclonal antibodies. Electrophoresis, 2017, 38(6): 769-785.
During the last years there was a substantial increase in the use of antibodies and related proteins as therapeutics. The emphasis of the pharmaceutical industry is on IgG1, IgG2, and IgG4 antibodies, which are therefore in the focus of this article. In order to ensure appropriate quality control of such biopharmaceuticals, deep understanding of their chemical degradation pathways and the resulting impact on potency, pharmacokinetics, and safety is required. Criticality of modifications may be specific for individual antibodies and has to be assessed for each molecule. However, some modifications of conserved structure elements occur in all or at least most IgGs. In these cases, criticality assessment may be applicable to related molecules or molecule formats. The relatively low dissociation energy of disulfide bonds and the high flexibility of the hinge region frequently lead to modifications and cleavages. Therefore, the hinge region and disulfide bonds require specific consideration during quality assessment of mAbs. In this review, available literature knowledge on underlying chemical reaction pathways of modifications, analytical methods for quantification and criticality are discussed. The hinge region is prone to cleavage and is involved in pathways that lead to thioether bond formation, cysteine racemization, and iso-Asp (Asp, aspartic acid) formation. Disulfide or sulfhydryl groups were found to be prone to reductive cleavage, trisulfide formation, cysteinylation, glutathionylation, disulfide bridging to further light chains, and disulfide scrambling. With regard to potency, disulfide cleavage, hinge cleavage, disulfide bridging to further light chains, and cysteinylation were found to influence antigen binding and fragment crystallizable (Fc) effector functionalities. Renal clearance of small fragments may be faster, whereas clearance of larger fragments appears to depend on their neonatal Fc receptor (FcRn) functionality, which in turn may be impeded by disulfide bond cleavage. Certain modifications such as disulfide induced aggregation and heterodimers from different antibodies are generally regarded critical with respect to safety. However, the detection of some modifications in endogenous antibodies isolated from human blood and the possibility of in vivo repair mechanisms may reduce some safety concerns.© 2016 The Authors. Electrophoresis published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.
[11]
Hutterer K M, Hong R W, Lull J, et al. Monoclonal antibody disulfide reduction during manufacturing: Untangling process effects from product effects. MAbs, 2013, 5(4): 608-613.
[12]
Wang T, Liu Y D, Cai B, et al. Investigation of antibody disulfide reduction and re-oxidation and impact to biological activities. Journal of Pharmaceutical and Biomedical Analysis, 2015, 102: 519-528.
[13]
Dionne B, Mishra N, Butler M. A low redox potential affects monoclonal antibody assembly and glycosylation in cell culture. Journal of Biotechnology, 2017, 246: 71-80.
[14]
Manteca A, Alonso-Caballero Á, Fertin M, et al. The influence of disulfide bonds on the mechanical stability of proteins is context dependent. Journal of Biological Chemistry, 2017, 292(32): 13374-13380.
Disulfide bonds play a crucial role in proteins, modulating their stability and constraining their conformational dynamics. A particularly important case is that of proteins that need to withstand forces arising from their normal biological function and that are often disulfide bonded. However, the influence of disulfides on the overall mechanical stability of proteins is poorly understood. Here, we used single-molecule force spectroscopy (smFS) to study the role of disulfide bonds in different mechanical proteins in terms of their unfolding forces. For this purpose, we chose the pilus protein FimG from Gram-negative bacteria and a disulfide-bonded variant of the I91 human cardiac titin polyprotein. Our results show that disulfide bonds can alter the mechanical stability of proteins in different ways depending on the properties of the system. Specifically, disulfide-bonded FimG undergoes a 30% increase in its mechanical stability compared with its reduced counterpart, whereas the unfolding force of I91 domains experiences a decrease of 15% relative to the WT form. Using a coarse-grained simulation model, we rationalized that the increase in mechanical stability of FimG is due to a shift in the mechanical unfolding pathway. The simple topology-based explanation suggests a neutral effect in the case of titin. In summary, our results indicate that disulfide bonds in proteins act in a context-dependent manner rather than simply as mechanical lockers, underscoring the importance of considering disulfide bonds both computationally and experimentally when studying the mechanical properties of proteins.© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.
[15]
Sakurai K, Nakahata R, Lee Y H, et al. Effects of a reduced disulfide bond on aggregation properties of the human IgG1 CH3 domain. Biochimica et Biophysica Acta, 2015, 1854(10): 1526-1535.
[16]
Derfus G E, Dizon-Maspat J, Broddrick J T, et al. Red colored IgG 4 caused by vitamin B12 from cell culture media combined with disulfide reduction at harvest. MAbs, 2014, 6(3): 679-688.
While many antibody therapeutics are formulated at low concentration (~10-20 mg/mL) for intravenous administration, high concentration (> 100 mg/mL) formulations may be required for subcutaneous delivery in certain clinical indications. For such high concentration formulations, product color is more apparent due to the higher molecular density across a given path-length. Color is therefore a product quality attribute that must be well-understood and controlled, to demonstrate process consistency and enable clinical trial blinding. Upon concentration of an IgG4 product at the 2000 L manufacturing scale, variability in product color, ranging from yellow to red, was observed. A small-scale experimental model was developed to assess the effect of processing conditions (medium composition and harvest conditions) on final bulk drug substance (BDS) color. The model was used to demonstrate that, for two distinct IgG4 products, red coloration occurred only in the presence of disulfide reduction-mediated antibody dissociation. The red color-causing component was identified as vitamin B 12, in the hydroxocobalamin form, and the extent of red color was correlated with the cobalt (vitamin B 12) concentration in the final pools. The intensity of redness in the final BDS was modulated by changing the concentration of vitamin B 12 in the cell culture media.
[17]
Du C, Martin R, Huang Y P, et al. Vitamin B12 association with mAbs: mechanism and potential mitigation strategies. Biotechnology and Bioengineering, 2018, 115(4): 900-909.
[18]
Chung W K, Russell B, Yang Y H, et al. Effects of antibody disulfide bond reduction on purification process performance and final drug substance stability. Biotechnology and Bioengineering, 2017, 114(6): 1264-1274.
Antibody disulfide bond reduction during monoclonal antibody (mAb) production is a phenomenon that has been attributed to the reducing enzymes from CHO cells acting on the mAb during the harvest process. However, the impact of antibody reduction on the downstream purification process has not been studied. During the production of an IgG mAb, antibody reduction was observed in the harvested cell culture fluid (HCCF), resulting in high fragment levels. In addition, aggregate levels increased during the low pH treatment step in the purification process. A correlation between the level of free thiol in the HCCF (as a result of antibody reduction) and aggregation during the low pH step was established, wherein higher levels of free thiol in the starting sample resulted in increased levels of aggregates during low pH treatment. The elevated levels of free thiol were not reduced over the course of purification, resulting in carry-over of high free thiol content into the formulated drug substance. When the drug substance with high free thiols was monitored for product degradation at room temperature and 2-8°C, faster rates of aggregation were observed compared to the drug substance generated from HCCF that was purified immediately after harvest. Further, when antibody reduction mitigations (e.g., chilling, aeration, and addition of cystine) were applied, HCCF could be held for an extended period of time while providing the same product quality/stability as material that had been purified immediately after harvest. Biotechnol. Bioeng. 2017;114: 1264-1274. © 2017 The Authors. Biotechnology and Bioengineering Published by Wiley Periodicals Inc.© 2017 The Authors. Biotechnology and Bioengineering Published by Wiley Periodicals Inc.
[19]
Gurjar S A, Wheeler J X, Wadhwa M, et al. The impact of thioredoxin reduction of allosteric disulfide bonds on the therapeutic potential of monoclonal antibodies. Journal of Biological Chemistry, 2019, 294(51): 19616-19634.
[20]
Li F, Vijayasankaran N, Shen A, et al. Cell culture processes for monoclonal antibody production. MAbs, 2010, 2(5): 466-479.
[21]
Cherkaoui S, Bettinger T, Hauwel M, et al. Tracking of antibody reduction fragments by capillary gel electrophoresis during the coupling to microparticles surface. Journal of Pharmaceutical and Biomedical Analysis, 2010, 53(2): 172-178.
Due to their high specificity and efficiency, antibodies are ideal ligands for target-specific ultrasound contrast agents. The present study focuses on the chemical stability of antibodies during functionalisation with sulfosuccinimidyl-pyridyldithiopropionamidohexanoate (SPDP), a heterobifunctional linker, which exposes free thiol groups upon treatment with a reducing agent. Thiolated antibodies can then react with thiol-reactive group, such as maleimide present on the microbubble surface to form stable covalent complexes. The immunoglobulin structure relies on several intra- and inter-chain disulfide bridges which might be affected by reducing agents. A capillary electrophoresis-sodium dodecyl sulfate (CE-SDS) method with UV detection was applied to address the effect of the functionalisation process on the structural integrity of the antibodies and revealed that antibody disulfide bonds are prone to reduction as function of the reducing agents. Depending on the coupling conditions, various IgG fragments were identified reflecting different combinations between the light and heavy chains. Furthermore, two commonly used reducing agents, namely triscarboxyethylphosphine (TCEP) and 1,4-dithiothreitol (DTT) were compared under various preparation conditions. Results showed that reduction conditions based on DTT as a reducing agent under acidic pH were more appropriate to preserve intra- and inter-disulfide bridges of SPDP-modified antibodies.
[22]
Dada O O, Rao R, Jones N, et al. Comparison of SEC and CE-SDS methods for monitoring hinge fragmentation in IgG1 monoclonal antibodies. Journal of Pharmaceutical and Biomedical Analysis, 2017, 145: 91-97.
[23]
Guan X Y, Zhang L, Wypych J. Direct mass spectrometric characterization of disulfide linkages. MAbs, 2018, 10(4): 572-582.
[24]
Deslignière E, Botzanowski T, Diemer H, et al. High-resolution IMS-MS to assign additional disulfide bridge pairing in complementarity-determining regions of an IgG 4 monoclonal antibody. Journal of the American Society for Mass Spectrometry, 2021, 32(10): 2505-2512.
Monoclonal antibodies (mAbs) have taken on an increasing importance for the treatment of various diseases, including cancers and immunological disorders. Disulfide bonds play a pivotal role in therapeutic antibody structure and activity relationships. Disulfide connectivity and cysteine-related variants are considered as critical quality attributes that must be monitored during mAb manufacturing and storage, as non-native disulfide bridges and aggregates might be responsible for loss of biological function and immunogenicity. The presence of cysteine residues in the complementarity-determining regions (CDRs) is rare in human antibodies but may be critical for the antigen-binding or deleterious for therapeutic antibody development. Consequently, in-depth characterization of their disulfide network is a prerequisite for mAb developability assessment. Mass spectrometry (MS) techniques represent powerful tools for accurate identification of disulfide connectivity. We report here on the MS-based characterization of an IgG4 comprising two additional cysteine residues in the CDR of its light chain. Classical bottom-up approaches after trypsin digestion first allowed identification of a dipeptide containing two disulfide bridges. To further investigate the conformational heterogeneity of the disulfide-bridged dipeptide, we performed ion mobility spectrometry-mass spectrometry (IMS-MS) experiments. Our results highlight benefits of high resolution IMS-MS to tackle the conformational landscape of disulfide peptides generated after trypsin digestion of a humanized IgG4 mAb under development. By comparing arrival time distributions of the mAb-collected and synthetic peptides, cyclic IMS afforded unambiguous assessment of disulfide bonds. In addition to classical peptide mapping, qualitative high-resolution IMS-MS can be of great interest to identify disulfide bonds within therapeutic mAbs.
[25]
Liu H C, Gaza-Bulseco G, Chumsae C. Analysis of reduced monoclonal antibodies using size exclusion chromatography coupled with mass spectrometry. Journal of the American Society for Mass Spectrometry, 2009, 20(12): 2258-2264.
[26]
Mullan B, Dravis B, Lim A, et al. Disulphide bond reduction of a therapeutic monoclonal antibody during cell culture manufacturing operations. BMC Proceedings, 2011, 5(Suppl 8): P110.
[27]
Trivedi M V, Laurence J S, Siahaan T J. The role of thiols and disulfides on protein stability. Current Protein & Peptide Science, 2009, 10(6): 614-625.
[28]
Du C, Huang Y P, Borwankar A, et al. Using hydrogen peroxide to prevent antibody disulfide bond reduction during manufacturing process. MAbs, 2018, 10(3): 500-510.
[29]
Park S Y, Egan S, Cura A J, et al. Untargeted proteomics reveals upregulation of stress response pathways during CHO-based monoclonal antibody manufacturing process leading to disulfide bond reduction. MAbs, 2021, 13(1): 1963094.
[30]
Xie W L, Ma W J, Liu P, et al. Overview of thioredoxin system and targeted therapies for acute leukemia. Mitochondrion, 2019, 47: 38-46.
[31]
Lillig C H, Berndt C, Holmgren A. Glutaredoxin systems. Biochimica et Biophysica Acta (BBA) - General Subjects, 2008, 1780(11): 1304-1317.
[32]
Handlogten M W, Zhu M, Ahuja S. Glutathione and thioredoxin systems contribute to recombinant monoclonal antibody interchain disulfide bond reduction during bioprocessing. Biotechnology and Bioengineering, 2017, 114(7): 1469-1477.
Antibody interchain disulfide bond reduction during biopharmaceutical manufacturing has received increased attention since it was first reported in 2010. Antibody reduction leads to loss of product and reduced product stability. It is therefore critical to understand the underlying mechanisms of reduction. To date, the thioredoxin system has been reported as the sole contributor to antibody reduction during bioprocessing. In this work, we show that the glutathione system, in addition to the thioredoxin system, is involved in reducing antibody molecules and the contributions of the two systems can vary depending upon the cell culture process. The roles of the glutathione and thioredoxin systems were evaluated for three molecules with different IgG subclass where reduction was observed during manufacturing: mAb A, mAb B, and mAb C representing an IgG, IgG, and IgG respectively. The expression of enzymes for both the thioredoxin and glutathione systems were confirmed in all three cell lines. Inhibitors were evaluated using purified mammalian reductases to evaluate their specificity. The optimized experimental conditions enabled both the determination of reductase activity contributed from as well as the amount of antibody reduced by each enzymatic system. Our results demonstrate that the underlying enzymatic mechanisms are different depending upon the cell culture process; one of the two systems may be the dominant mechanism, or both enzymatic systems may be involved. Specifically, the glutathione system was found to be the major contributor to mAb A reduction while the thioredoxin system was the major contributor to mAb C reduction. Intriguingly, mAb B experienced significant reduction from both enzymatic systems. In summary, we have demonstrated that in addition to the thioredoxin pathway, the glutathione system is a second major pathway contributing to antibody reduction and this knowledge can be leveraged to develop more specific antibody reduction mitigation strategies targeted at the dominant reduction mechanism. Biotechnol. Bioeng. 2017;114: 1469-1477. © 2017 Wiley Periodicals, Inc.© 2017 Wiley Periodicals, Inc.
[33]
Kao Y H, Hewitt D P, Trexler-Schmidt M, et al. Mechanism of antibody reduction in cell culture production processes. Biotechnology and Bioengineering, 2010, 107(4): 622-632.
[34]
Cura A J, Xu X K, Egan S, et al. Metabolic understanding of disulfide reduction during monoclonal antibody production. Applied Microbiology and Biotechnology, 2020, 104(22): 9655-9669.
[35]
Chakrabarti S, Barrow C J, Kanwar R K, et al. Studies to prevent degradation of recombinant fc-fusion protein expressed in mammalian cell line and protein characterization. International Journal of Molecular Sciences, 2016, 17(6): 913.
[36]
Li F, Hashimura Y, Pendleton R, et al. A systematic approach for scale-down model development and characterization of commercial cell culture processes. Biotechnology Progress, 2006, 22(3): 696-703.
[37]
Trexler-Schmidt M, Sargis S, Chiu J, et al. Identification and prevention of antibody disulfide bond reduction during cell culture manufacturing. Biotechnology and Bioengineering, 2010, 106(3): 452-461.
In the biopharmaceutical industry, therapeutic monoclonal antibodies are primarily produced in mammalian cell culture systems. During the scale-up of a monoclonal antibody production process, we observed excessive mechanical cell shear as well as significant reduction of the antibody's interchain disulfide bonds during harvest operations. This antibody reduction event was catastrophic as the product failed to meet the drug substance specifications and the bulk product was lost. Subsequent laboratory studies have demonstrated that cells subjected to mechanical shear release cellular enzymes that contribute to this antibody reduction phenomenon (manuscript submitted; Kao et al., 2009). Several methods to prevent this antibody reduction event were developed using a lab-scale model to reproduce the lysis and reduction events. These methods included modifications to the cell culture media with chemicals (e.g., cupric sulfate (CuSO(4))), pre- and post-harvest chemical additions to the cell culture fluid (CCF) (e.g., CuSO(4), EDTA, L-cystine), as well as lowering the pH and air sparging of the harvested CCF (HCCF). These methods were evaluated for their effectiveness in preventing disulfide bond reduction and their impact to product quality. Effective prevention methods, which yielded acceptable product quality were evaluated for their potential to be implemented at manufacturing-scale. The work described here identifies numerous effective reduction prevention measures from lab-scale studies; several of these methods were then successfully translated into manufacturing processes.2010 Wiley Periodicals, Inc.
[38]
O’Mara B, Gao Z H, Kuruganti M, et al. Impact of depth filtration on disulfide bond reduction during downstream processing of monoclonal antibodies from CHO cell cultures. Biotechnology and Bioengineering, 2019, 116(7): 1669-1683.
[39]
Handlogten M W, Zhu M, Ahuja S. Intracellular response of CHO cells to oxidative stress and its influence on metabolism and antibody production. Biochemical Engineering Journal, 2018, 133: 12-20.
[40]
Xie P P, Niu H J, Chen X N, et al. Elucidating the effects of pH shift on IgG 1 monoclonal antibody acidic charge variant levels in Chinese hamster ovary cell cultures. Applied Microbiology and Biotechnology, 2016, 100(24): 10343-10353.
[41]
Le Basle Y, Chennell P, Tokhadze N, et al. Physicochemical stability of monoclonal antibodies: a review. Journal of Pharmaceutical Sciences, 2020, 109(1): 169-190.
[42]
Liu H C, Nowak C, Shao M, et al. Impact of cell culture on recombinant monoclonal antibody product heterogeneity. Biotechnology Progress, 2016, 32(5): 1103-1112.
[43]
Koterba K L, Borgschulte T, Laird M W. Thioredoxin 1 is responsible for antibody disulfide reduction in CHO cell culture. Journal of Biotechnology, 2012, 157(1): 261-267.
[44]
Brühlmann D, Jordan M, Hemberger J, et al. Tailoring recombinant protein quality by rational media design. Biotechnology Progress, 2015, 31(3): 615-629.
Clinical efficacy and safety of recombinant proteins are closely associated with their structural characteristics. The major quality attributes comprise glycosylation, charge variants (oxidation, deamidation, and C- & N-terminal modifications), aggregates, low-molecular-weight species (LMW), and misincorporation of amino acids in the protein backbone. Cell culture media design has a great potential to modulate these quality attributes due to the vital role of medium in mammalian cell culture. The purpose of this review is to provide an overview of the way both classical cell culture medium components and novel supplements affect the quality attributes of recombinant therapeutic proteins expressed in mammalian hosts, allowing rational and high-throughput optimization of mammalian cell culture media. A selection of specific and/or potent inhibitors and activators of oligosaccharide processing as well as components affecting multiple quality attributes are presented. Extensive research efforts in this field show the feasibility of quality engineering through media design, allowing to significantly modulate the protein function.© 2015 American Institute of Chemical Engineers.
[45]
Chaderjian W B, Chin E T, Harris R J, et al. Effect of copper sulfate on performance of a serum-free CHO cell culture process and the level of free thiol in the recombinant antibody expressed. Biotechnology Progress, 2005, 21(2): 550-553.
A recombinant Chinese hamster ovary (CHO) cell line was used to express a humanized antibody. Product quality analysis of this humanized antibody showed the presence of free thiol, due to unpaired cysteine residues in the Fab region. Decreased potency of this thiol Fab made it critical to minimize the levels of free thiol. In an effort to do this, we evaluated the effect of copper sulfate addition to the cell culture production medium. As a component of the production medium, copper sulfate can act as an oxidizing agent, thereby facilitating disulfide bond formation. Four concentrations of copper sulfate were added at the beginning of 2-L benchtop production cultures of the recombinant CHO cell line: 0, 5, 50, and 100 microM. We found that these copper sulfate additions had no effect on cell growth or antibody production. However, a slight dose-dependent depression in culture viability was observed. Analysis of the purified antibody showed that either the 50 or 100 microM copper sulfate additions reduced the level of free thiol by more than 10-fold.
[46]
Franey H, Brych S R, Kolvenbach C G, et al. Increased aggregation propensity of IgG 2 subclass over IgG1: role of conformational changes and covalent character in isolated aggregates. Protein Science, 2010, 19(9): 1601-1615.
[47]
Handlogten M W, Wang J H, Ahuja S. Online control of cell culture redox potential prevents antibody interchain disulfide bond reduction. Biotechnology and Bioengineering, 2020, 117(5): 1329-1336.
The phenomenon of monoclonal antibody (mAb) interchain disulfide bond reduction during manufacturing processes continues to be a focus of the biotechnology industry due to the potential for loss of product, increased complexity of purification processes, and reduced stability of the drug product. We hypothesized that antibody reduction can be mitigated by controlling the cell culture redox potential and subsequently established a threshold redox potential above which the mAb remained intact and below which there were significant and highly variable amounts of reduced mAb. Using this knowledge, we developed three control schemes to prevent mAb reduction in the bioreactor by controlling the cell culture redox potential via an online redox probe. These control methodologies functioned by increasing the concentration of dissolved oxygen (DO), copper (II) (Cu), or both DO and Cu to maintain the redox potential above the threshold value. Using these methods, we were able to demonstrate successful control of antibody reduction. Importantly, the redox control strategies did not significantly impact the cell growth, viability, mAb production, or product quality attributes including aggregates, C-terminal lysine, high mannose, deamidation, and glycation. Our results demonstrate that controlling the cell culture redox potential is a simple and effective method to prevent mAb reduction.© 2020 Wiley Periodicals, Inc.
[48]
Ishikawa T, Ito T, Endo R, et al. Influence of pH on heat-induced aggregation and degradation of therapeutic monoclonal antibodies. Biological & Pharmaceutical Bulletin, 2010, 33(8): 1413-1417.
[49]
Zhang W J, Liu X P, Tang H P, et al. Investigation into the impact of tyrosine on the product formation and quality attributes of mAbs in rCHO cell cultures. Applied Microbiology and Biotechnology, 2020, 104(16): 6953-6966.
[50]
Hutchinson N, Bingham N, Murrell N, et al. Shear stress analysis of mammalian cell suspensions for prediction of industrial centrifugation and its verification. Biotechnology and Bioengineering, 2006, 95(3): 483-491.
This article describes the use of ultra scale-down studies requiring milliliter quantities of process material to study the clarification of mammalian cell culture broths using industrial-scale continuous centrifuges during the manufacture of a monoclonal antibody for therapeutic use. Samples were pretreated in a small high-speed rotating-disc device in order to mimic the effect on the cells of shear stresses in the feed zone of the industrial scale centrifuges. The use of this feed mimic was shown to predict a reduction of the clarification efficiency by significantly reducing the particle size distribution of the mammalian cells. The combined use of the rotating-disc device and a laboratory-scale test tube centrifuge successfully predicted the separation characteristics of industrial-scale, disc stack centrifuges operating with different feed zones. A 70% reduction in flow rate in the industrial-scale centrifuge was shown to arise from shear effects. A predicted 2.5-fold increase in throughput for the same clarification performance, achieved by the change to a centrifuge using a feed zone designed to give gentler acceleration of the bioprocess fluid, was also verified at large-scale.
[51]
Mamathambika B S, Bardwell J C. Disulfide-linked protein folding pathways. Annual Review of Cell and Developmental Biology, 2008, 24: 211-235.
Determining the mechanism by which proteins attain their native structure is an important but difficult problem in basic biology. The study of protein folding is difficult because it involves the identification and characterization of folding intermediates that are only very transiently present. Disulfide bond formation is thermodynamically linked to protein folding. The availability of thiol trapping reagents and the relatively slow kinetics of disulfide bond formation have facilitated the isolation, purification, and characterization of disulfide-linked folding intermediates. As a result, the folding pathways of several disulfide-rich proteins are among the best known of any protein. This review discusses disulfide bond formation and its relationship to protein folding in vitro and in vivo.
[52]
Tang P F, Tan Z J, Ehamparanathan V, et al. Optimization and kinetic modeling of interchain disulfide bond reoxidation of monoclonal antibodies in bioprocesses. MAbs, 2020, 12(1): 1829336.
[53]
Tan Z J, Ehamparanathan V, Ren T W, et al. On-column disulfide bond formation of monoclonal antibodies during protein A chromatography eliminates low molecular weight species and rescues reduced antibodies. MAbs, 2020, 12(1): 1829333.
[54]
Wasalathanthri D P, Shah R, Ding J L, et al. Process analytics 4.0: a paradigm shift in rapid analytics for biologics development. Biotechnology Progress, 2021, 37(4): e3177.
Analytical testing of product quality attributes and process parameters during the biologics development (Process analytics) has been challenging due to the rapid growth of biomolecules with complex modalities to support unmet therapeutic needs. Thus, the expansion of process analytics tool box for rapid analytics with the deployment of cutting edge technologies and cyber physical systems is a necessity. We introduce the term, Process Analytics 4.0; which entails not only technology aspects such as process analytical technology (PAT), assay automation, and high-throughput analytics, but also cyber physical systems that enable data management, visualization, augmented reality and internet of things (IoT) infrastructure for real time analytics in process development environment. This review is exclusively focused on dissecting high-level features of PAT, automation, and data management with some insights into the business aspects of implementing during process analytical testing in biologics process development. Significant technological and business advantages can be gained with the implementation of digitalization, automation and real time testing. A systematic development and employment of PAT in process development workflows enable real time analytics for better process understanding, agility and sustainability. Robotics and liquid handling workstations allow rapid assay and sample preparation automation to facilitate high-throughput testing of attributes and molecular properties which are otherwise challenging to monitor with PAT tools due to technological and business constraints. Cyber-physical systems for data management, visualization and repository must be established as part of Process Analytics 4.0 framework. Furthermore, we review some of the challenges in implementing these technologies based on our expertise in process analytics for biopharmaceutical drug substance development.© 2021 American Institute of Chemical Engineers.
[55]
Wasalathanthri D P, Rehmann M S, Song Y L, et al. Technology outlook for real-time quality attribute and process parameter monitoring in biopharmaceutical development-a review. Biotechnology and Bioengineering, 2020, 117(10): 3182-3198.
Real-time monitoring of bioprocesses by the integration of analytics at critical unit operations is one of the paramount necessities for quality by design manufacturing and real-time release (RTR) of biopharmaceuticals. A well-defined process analytical technology (PAT) roadmap enables the monitoring of critical process parameters and quality attributes at appropriate unit operations to develop an analytical paradigm that is capable of providing real-time data. We believe a comprehensive PAT roadmap should entail not only integration of analytical tools into the bioprocess but also should address automated-data piping, analysis, aggregation, visualization, and smart utility of data for advanced-data analytics such as machine and deep learning for holistic process understanding. In this review, we discuss a broad spectrum of PAT technologies spanning from vibrational spectroscopy, multivariate data analysis, multiattribute chromatography, mass spectrometry, sensors, and automated-sampling technologies. We also provide insights, based on our experience in clinical and commercial manufacturing, into data automation, data visualization, and smart utility of data for advanced-analytics in PAT. This review is catered for a broad audience, including those new to the field to those well versed in applying these technologies. The article is also intended to give some insight into the strategies we have undertaken to implement PAT tools in biologics process development with the vision of realizing RTR testing in biomanufacturing and to meet regulatory expectations.© 2020 Wiley Periodicals LLC.
[56]
Chemmalil L, Prabhakar T, Kuang J E, et al. Online/at-line measurement, analysis and control of product titer and critical product quality attributes (CQAs) during process development. Biotechnology and Bioengineering, 2020, 117(12): 3757-3765.
[57]
Sinharoy P, McFarland K S, Majewska N I, et al. Redox as a bioprocess parameter: analytical redox quantification in biological therapeutic production. Current Opinion in Biotechnology, 2021, 71: 49-54.
Engineered Chinese hamster ovary (CHO) cells are the most widely utilized cell line for protein-based therapeutics production at industrial scales. Process development strategies which improve production capacity and quality are often implemented without an understanding of underlying intracellular changes. Intracellular redox conditions drive reactions in pathways critical to biologics production, including bioenergetic and biosynthetic pathways, necessitating methods to quantify redox-related changes. Advances in methods for analytical redox quantification presented here, including bioreactor probes, redox-targeted proteomics, genetically encoded redox-sensitive fluorescent proteins, and biochemical assays, are creating new opportunities to characterize the effects of redox in biologics production. Implementing these methods will lead to enhanced media formulations, improved bioprocess strategies, and new cell line engineering targets and ultimately develop redox into an optimizable bioprocess parameter to improve the yield and quality of these lifesaving medicines.Copyright © 2021 Elsevier Ltd. All rights reserved.
[58]
Meneses-Acosta A, Gómez A, Ramírez O T. Control of redox potential in hybridoma cultures: effects on MAb production, metabolism, and apoptosis. Journal of Industrial Microbiology and Biotechnology, 2012, 39(8): 1189-1198.
Culture redox potential (CRP) has proven to be a valuable monitoring tool in several areas of biotechnology; however, it has been scarcely used in animal cell culture. In this work, a proportional feedback control was employed, for the first time, to maintain the CRP at different constant values in hybridoma batch cultures for production of a monoclonal antibody (MAb). Reducing and oxidant conditions, in the range of -130 and +70 mV, were maintained in 1-l bioreactors through automatic control of the inlet gas composition. Cultures at constant DOT, in the range of 3 and 300 %, were used for comparison. The effect of constant CRP on cell concentration, MAb production, metabolism of glucose, glutamine, thiols, oxygen consumption, and programmed cell death, was evaluated. Reducing conditions resulted in the highest viable cell and MAb concentrations and thiols production, whereas specific glucose and glutamine consumption rates remained at the lowest values. In such conditions, programmed cell death, particularly apoptosis, occurred only after nutrient exhaustion. The optimum specific MAb production rate occurred at intermediate CRP levels. Oxidant conditions resulted in a detrimental effect in all culture parameters, increasing the specific glucose, glutamine, and oxygen consumption rates and inducing the apoptotic process, which was detected as early as 24 h even when glutamine and glucose were present at non-limiting concentrations. In most cases, such results were similar to those obtained in control cultures at constant DOT.
[59]
Switzar L, Nicolardi S, Rutten J W, et al. In-depth characterization of protein disulfide bonds by online liquid chromatography-electrochemistry-mass spectrometry. Journal of the American Society for Mass Spectrometry, 2016, 27(1): 50-58.
Disulfide bonds are an important class of protein post-translational modifications, yet this structurally crucial modification type is commonly overlooked in mass spectrometry (MS)-based proteomics approaches. Recently, the benefits of online electrochemistry-assisted reduction of protein S-S bonds prior to MS analysis were exemplified by successful characterization of disulfide bonds in peptides and small proteins. In the current study, we have combined liquid chromatography (LC) with electrochemistry (EC) and mass analysis by Fourier transform ion cyclotron resonance (FTICR) MS in an online LC-EC-MS platform to characterize protein disulfide bonds in a bottom-up proteomics workflow. A key advantage of a LC-based strategy is the use of the retention time in identifying both intra- and interpeptide disulfide bonds. This is demonstrated by performing two sequential analyses of a certain protein digest, once without and once with electrochemical reduction. In this way, the "parent" disulfide-linked peptide detected in the first run has a retention time-based correlation with the EC-reduced peptides detected in the second run, thus simplifying disulfide bond mapping. Using this platform, both inter- and intra-disulfide-linked peptides were characterized in two different proteins, ß-lactoglobulin and ribonuclease B. In order to prevent disulfide reshuffling during the digestion process, proteins were digested at a relatively low pH, using (a combination of) the high specificity proteases trypsin and Glu-C. With this approach, disulfide bonds in ß-lactoglobulin and ribonuclease B were comprehensively identified and localized, showing that online LC-EC-MS is a useful tool for the characterization of protein disulfide bonds.
[60]
Wu S L, Jiang H T, Lu Q Z, et al. Mass spectrometric determination of disulfide linkages in recombinant therapeutic proteins using online LC-MS with electron-transfer dissociation. Analytical Chemistry, 2009, 81(1): 112-122.
[61]
Abu-Absi N R, Kenty B M, Cuellar M E, et al. Real time monitoring of multiple parameters in mammalian cell culture bioreactors using an in-line Raman spectroscopy probe. Biotechnology and Bioengineering, 2011, 108(5): 1215-1221.
[62]
Matthews T E, Berry B N, Smelko J, et al. Closed loop control of lactate concentration in mammalian cell culture by Raman spectroscopy leads to improved cell density, viability, and biopharmaceutical protein production. Biotechnology and Bioengineering, 2016, 113(11): 2416-2424.
Accumulation of lactate in mammalian cell culture often negatively impacts culture performance, impeding production of therapeutic proteins. Many efforts have been made to limit the accumulation of lactate in cell culture. Here, we describe a closed loop control scheme based on online spectroscopic measurements of glucose and lactate concentrations. A Raman spectroscopy probe was used to monitor a fed-batch mammalian cell culture and predict glucose and lactate concentrations via multivariate calibration using partial least squares regression (PLS). The PLS models had a root mean squared error of prediction (RMSEP) of 0.27 g/L for glucose and 0.20 g/L for lactate. All glucose feeding was controlled by the Raman PLS model predictions. Glucose was automatically fed when lactate levels were beneath a setpoint (either 4.0 or 2.5 g/L) and glucose was below its own setpoint (0.5 g/L). This control scheme was successful in maintaining lactate levels at an arbitrary setpoint throughout the culture, as compared to the eventual accumulate of lactate to 8.0 g/L in the historical process. Automated control of lactate by restricted glucose feeding led to improvements in culture duration, viability, productivity, and robustness. Culture duration was extended from 11 to 13 days, and harvest titer increased 85% over the historical process. Biotechnol. Bioeng. 2016;113: 2416-2424. © 2016 Wiley Periodicals, Inc.© 2016 Wiley Periodicals, Inc.
[63]
Liu Z M, Zhang Z J, Qin Y J, et al. The application of Raman spectroscopy for monitoring product quality attributes in perfusion cell culture. Biochemical Engineering Journal, 2021, 173: 108064.
PDF(1593 KB)

Accesses

Citation

Detail

Sections
Recommended

/