1
|
Wang X, Wang J, Han Y, Jiang X, Cao S, Xu D, Xiong T, Guo X, Wang C, Guo S, Song H, Dong T, Zhang L, An Z, Liu J, Han J, Wu H. Utilizing a hydrophobic primary container surface to reduce the formation of subvisible particles in monoclonal antibody solution caused by fluid shear. Eur J Pharm Biopharm 2024; 204:114502. [PMID: 39293723 DOI: 10.1016/j.ejpb.2024.114502] [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: 03/27/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
The exposure of protein molecules to interfaces may cause protein aggregation and particle formation in protein formulations, especially hydrophobic interfaces, which may promote protein aggregation in solution. In this study, we found that modification of the surface properties by application of a hydrophobic Octadecyltrichlorosilane (OTS) could reduce the generation of protein aggregates and particles in protein solution induced by fluid shear. A stable protein adsorption layer was formed at the hydrophobic interface through the strong hydrophobic interaction between the protein and hydrophobic surface, which could prevent the aggregated protein from falling off into the bulk solution to form subvisible particles and insoluble protein aggregates. In addition, human complement enzyme linked immunosorbent assay results showed that the particles that were generated in the OTS-coated container did not activate human complement which indicated the OTS-coated container could be used as primary containers for certain types of monoclonal antibody formulation.
Collapse
Affiliation(s)
- Xinyue Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Junjie Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Yang Han
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Xingchun Jiang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Sixian Cao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Dongze Xu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Tiancheng Xiong
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Xiang Guo
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Cui Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China
| | - Sha Guo
- National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | | | - Ting Dong
- Qilu Pharmaceutical, Shandong 250104, China
| | - Le Zhang
- Qilu Pharmaceutical, Shandong 250104, China
| | | | - Jun Liu
- Qilu Pharmaceutical, Shandong 250104, China
| | - Jing Han
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Liaoning 110016, China.
| | - Hao Wu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning 110016, China.
| |
Collapse
|
2
|
São Pedro MN, Eppink MHM, Ottens M. Application of a fluorescent dye-based microfluidic sensor for real-time detection of mAb aggregates. Biotechnol Prog 2024; 40:e3355. [PMID: 37161718 DOI: 10.1002/btpr.3355] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/11/2023]
Abstract
The lack of process analytical technologies able to provide real-time information and process control over a biopharmaceutical process has long impaired the transition to continuous biomanufacturing. For the monoclonal antibody (mAb) production, aggregate formation is a major critical quality attribute (CQA) with several known process parameters (i.e., protein concentration and agitation) influencing this phenomenon. The development of a real-time tool to monitor aggregate formation is then crucial to gain control and achieve a continuous processing. Due to an inherent short operation time, miniaturized biosensors placed after each step can be a powerful solution. In this work, the development of a fluorescent dye-based microfluidic sensor for fast at-line PAT is described, using fluorescent dyes to examine possible mAb size differences. A zigzag microchannel, which provides 90% of mixing efficiency under 30 s, coupled to an UV-Vis detector, and using four FDs, was studied and validated. With different generated mAb aggregation samples, the FDs Bis-ANS and CCVJ were able to robustly detect from, at least, 2.5% to 10% of aggregation. The proposed FD-based micromixer is then ultimately implemented and validated in a lab-scale purification system, demonstrating the potential of a miniaturized biosensor to speed up CQAs measurement in a continuous process.
Collapse
Affiliation(s)
- Mariana N São Pedro
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands
| | - Michel H M Eppink
- Byondis B.V., Nijmegen, The Netherlands
- Bioprocessing Engineering, Wageningen University, Wageningen, The Netherlands
| | - Marcel Ottens
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
3
|
Lee JH, Kim JW, Lee HE, Song JY, Cho AH, Hwang JH, Heo K, Lee S. A dual-targeting approach using a human bispecific antibody against the receptor-binding domain of the Middle East Respiratory Syndrome Coronavirus. Virus Res 2024; 345:199383. [PMID: 38697296 PMCID: PMC11074968 DOI: 10.1016/j.virusres.2024.199383] [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: 01/17/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
The emergence of the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) has posed a significant global health concern due to its severe respiratory illness and high fatality rate. Currently, despite the potential for resurgence, there are no specific treatments for MERS-CoV, and only supportive care is available. Our study aimed to address this therapeutic gap by developing a potent neutralizing bispecific antibody (bsAb) against MERS-CoV. Initially, we isolated four human monoclonal antibodies (mAbs) that specifically target the MERS-CoV receptor-binding domain (RBD) using phage display technology and an established human antibody library. Among these four selected mAbs, our intensive in vitro functional analyses showed that the MERS-CoV RBD-specific mAb K111.3 exhibited the most potent neutralizing activity against MERS-CoV pseudoviral infection and the molecular interaction between MERS-CoV RBD and human dipeptidyl peptidase 4. Consequently, we engineered a novel bsAb, K207.C, by utilizing K111.3 as the IgG base and fusing it with the single-chain variable fragment of its non-competing pair, K111.1. This engineered bsAb showed significantly enhanced neutralization potential against MERS-CoV compared to its parental mAb. These findings suggest that K207.C may serve as a potential candidate for effective MERS-CoV neutralization, further highlighting the promise of the bsAb dual-targeting approach in MERS-CoV neutralization.
Collapse
Affiliation(s)
- Ji Hyun Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Woong Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Hee Eon Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Jin Young Song
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ah Hyun Cho
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Jae Hyeon Hwang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea; Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea.
| |
Collapse
|
4
|
Watanabe H, Hayashida N, Sato M, Honda S. Biosensing-based quality control monitoring of the higher-order structures of therapeutic antibody domains. Anal Chim Acta 2024; 1303:342439. [PMID: 38609254 DOI: 10.1016/j.aca.2024.342439] [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: 11/06/2023] [Revised: 01/18/2024] [Accepted: 02/28/2024] [Indexed: 04/14/2024]
Abstract
Advanced biopharmaceutical manufacturing requires novel process analytical technologies for the rapid and sensitive assessment of the higher-order structures of therapeutic proteins. However, conventional physicochemical analyses of denatured proteins have limitations in terms of sensitivity, throughput, analytical resolution, and real-time monitoring capacity. Although probe-based sensing can overcome these limitations, typical non-specific probes lack analytical resolution and provide little to no information regarding which parts of the protein structure have been collapsed. To meet these analytical demands, we generated biosensing probes derived from artificial proteins that could specifically recognize the higher-order structural changes in antibodies at the protein domain level. Biopanning of phage-displayed protein libraries generated artificial proteins that bound to a denatured antibody domain, but not its natively folded structure, with nanomolar affinity. The protein probes not only recognized the higher-order structural changes in intact IgGs but also distinguished between the denatured antibody domains. These domain-specific probes were used to generate response contour plots to visualize the antibody denaturation caused by various process parameters, such as pH, temperature, and holding time for acid elution and virus inactivation. These protein probes can be combined with established analytical techniques, such as surface plasmon resonance for real-time monitoring or plate-based assays for high-throughput analysis, to aid in the development of new analytical technologies for the process optimization and monitoring of antibody manufacturing.
Collapse
Affiliation(s)
- Hideki Watanabe
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Naoko Hayashida
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan
| | - Megumi Sato
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Shinya Honda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan.
| |
Collapse
|
5
|
Kim JW, Lee JH, Kim HJ, Heo K, Lee Y, Jang HJ, Lee HY, Park JW, Cho YB, Shin HG, Yang HR, Lee HE, Song JY, Lee S. Empowering SARS-CoV-2 variant neutralization with a bifunctional antibody engineered with tandem heptad repeat 2 peptides. J Med Virol 2024; 96:e29506. [PMID: 38445718 DOI: 10.1002/jmv.29506] [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: 11/17/2023] [Revised: 01/28/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024]
Abstract
With the global pandemic and the continuous mutations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the need for effective and broadly neutralizing treatments has become increasingly urgent. This study introduces a novel strategy that targets two aspects simultaneously, using bifunctional antibodies to inhibit both the attachment of SARS-CoV-2 to host cell membranes and viral fusion. We developed pioneering IgG4-(HR2)4 bifunctional antibodies by creating immunoglobulin G4-based and phage display-derived human monoclonal antibodies (mAbs) that specifically bind to the SARS-CoV-2 receptor-binding domain, engineered with four heptad repeat 2 (HR2) peptides. Our in vitro experiments demonstrate the superior neutralization efficacy of these engineered antibodies against various SARS-CoV-2 variants, ranging from original SARS-CoV-2 strain to the recently emerged Omicron variants, as well as SARS-CoV, outperforming the parental mAb. Notably, intravenous monotherapy with the bifunctional antibody neutralizes a SARS-CoV-2 variant in a murine model without causing significant toxicity. In summary, this study unveils the significant potential of HR2 peptide-driven bifunctional antibodies as a potent and versatile strategy for mitigating SARS-CoV-2 infections. This approach offers a promising avenue for rapid development and management in the face of the continuously evolving SARS-CoV-2 variants, holding substantial promise for pandemic control.
Collapse
Affiliation(s)
- Ji Woong Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ji Hyun Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Hyun Jung Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
- Department of Chemistry, Kookmin University, Seoul, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul, Republic of Korea
| | - Yoonwoo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Hui Jeong Jang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | - Yea Bin Cho
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Hee Eon Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Jin Young Song
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul, Republic of Korea
- Department of Chemistry, Kookmin University, Seoul, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Lambiase G, Klottrup-Rees K, Lovelady C, Ali S, Shepherd S, Muroni M, Lindo V, James DC, Dickman MJ. An automated, low volume, and high-throughput analytical platform for aggregate quantitation from cell culture media. J Chromatogr A 2023; 1691:463809. [PMID: 36731329 DOI: 10.1016/j.chroma.2023.463809] [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: 10/07/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
High throughput screening methods have driven a paradigm shift in biopharmaceutical development by reducing the costs of good manufactured (COGM) and accelerate the launch to market of novel drug products. Scale-down cell culture systems such as shaken 24- and 96-deep-well plates (DWPs) are used for initial screening of hundreds of recombinant mammalian clonal cell lines to quickly and efficiently select the best producing strains expressing product quality attributes that fit to industry platform. A common modification monitored from early-stage product development is protein aggregation due to its impact on safety and efficacy. This study aims to integrate high-throughput analysis of aggregation-prone therapeutic proteins with 96-deep well plate screening to rank clones based on the aggregation levels of the expressed proteins. Here we present an automated, small-scale analytical platform workflow combining the purification and subsequent aggregation analysis of protein biopharmaceuticals expressed in 96-DWP cell cultures. Product purification was achieved by small-scale solid-phase extraction using dual flow chromatography (DFC) automated on a robotic liquid handler for the parallel processing of up to 96 samples at a time. At-line coupling of size-exclusion chromatography (SEC) using a 2.1 mm ID column enabled the detection of aggregates with sub-2 µg sensitivity and a 3.5 min run time. The entire workflow was designed as an application to aggregation-prone mAbs and "mAb-like" next generation biopharmaceuticals, such as bispecific antibodies (BsAbs). Application of the high-throughput analytical workflow to a shake plate overgrow (SPOG) screen, enabled the screening of 384 different clonal cell lines in 32 h, requiring < 2 μg of protein per sample. Aggregation levels expressed by the clones varied between 9 and 76%. This high-throughput analytical workflow allowed for the early elimination of clonal cell lines with high aggregation, demonstrating the advantage of integrating analytical testing for critical quality attributes (CQAs) earlier in product development to drive better decision making.
Collapse
Affiliation(s)
- Giulia Lambiase
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St., Sheffield, UK; Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Kerensa Klottrup-Rees
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, AstraZeneca, Cambridge, UK
| | - Clare Lovelady
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, AstraZeneca, Cambridge, UK
| | - Salma Ali
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, AstraZeneca, Cambridge, UK
| | - Samuel Shepherd
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Maurizio Muroni
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Vivian Lindo
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK.
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St., Sheffield, UK.
| | - Mark J Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St., Sheffield, UK.
| |
Collapse
|
7
|
São Pedro MN, Santos MS, Eppink MHM, Ottens M. Design of a microfluidic mixer channel: First steps into creating a fluorescent dye-based biosensor for mAb aggregate detection. Biotechnol J 2023; 18:e2200332. [PMID: 36330557 DOI: 10.1002/biot.202200332] [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: 06/27/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/06/2022]
Abstract
A major challenge in the transition to continuous biomanufacturing is the lack of process analytical technology (PAT) tools which are able to collect real-time information on the process and elicit a response to facilitate control. One of the critical quality attributes (CQAs) of interest during monoclonal antibodies production is aggregate formation. The development of a real-time PAT tool to monitor aggregate formation is then crucial to have immediate feedback and process control. Miniaturized sensors placed after each unit operation can be a powerful solution to speed up an analytical measurement due to their characteristic short reaction time. In this work, a micromixer structure capable of mixing two streams is presented, to be employed in the detection of mAb aggregates using fluorescent dyes. Computational fluid dynamics (CFD) simulations were used to compare the mixing performance of a series of the proposed designs. A final design of a zigzag microchannel with 45° angle was reached and this structure was subsequently fabricated and experimentally validated with colour dyes and, later, with a FITC-IgG molecule. The designed zigzag micromixer presents a mixing index of around 90%, obtained in less than 30 seconds. Therefore, a micromixer channel capable of a fast and efficient mixing is hereby demonstrated, to be used as a real-time PAT tool for a fluorescence based detection of protein aggregation.
Collapse
Affiliation(s)
- Mariana N São Pedro
- Department of Biotechnology, Delft University of Technology, Delft, the Netherlands
| | - Mafalda S Santos
- Department of Biotechnology, Delft University of Technology, Delft, the Netherlands
| | - Michel H M Eppink
- Byondis B.V., Nijmegen, the Netherlands.,Bioprocessing Engineering, Wageningen University, Wageningen, the Netherlands
| | - Marcel Ottens
- Department of Biotechnology, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
8
|
Senga Y, Doi M, Onitsuka M, Honda S. Live-cell imaging to analyze intracellular aggregation of recombinant IgG in CHO cells. Cell Chem Biol 2021; 29:120-132.e4. [PMID: 34739851 DOI: 10.1016/j.chembiol.2021.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/05/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022]
Abstract
Recombinant immunoglobulin G (IgG) aggregates are formed during their production. However, the process underlying intracellular/extracellular aggregation in cell culture conditions is not well understood, and no effective method exists to assess IgG aggregates. Here, we establish an approach to detect intracellular aggregates using AF.2A1, a small artificial protein that binds to non-native IgG conformers and aggregates. Fluorescent-labeled AF.2A1 is prepared via conjugation and transfected into antibody-producing Chinese hamster ovary (CHO) cells. Micrographic images show intracellular IgG aggregates in CHO cells. The relative amount of intracellular aggregates (versus total intracellular IgG) differed depending on the type of additives used during cell culture. Interestingly, the relative amount of intracellular aggregates moderately correlates with that of in vitro extracellular IgG aggregates, suggesting they are secreted. This method will allow the investigation of antibody aggregation in cells, and may guide the production of therapeutic antibodies with high yield/quality.
Collapse
Affiliation(s)
- Yukako Senga
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Motomichi Doi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Masayoshi Onitsuka
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8513, Japan
| | - Shinya Honda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| |
Collapse
|
9
|
Akhunzada Z, Wu Y, Haby T, Jayawickrama D, McGeorge G, La Colla M, Bernstein J, Semones M, Abraham A. Analysis of biopharmaceutical formulations by Time Domain Nuclear Magnetic Resonance (TD-NMR) spectroscopy: A potential method for detection of counterfeit biologic pharmaceuticals. J Pharm Sci 2021; 110:2765-2770. [PMID: 33745914 DOI: 10.1016/j.xphs.2021.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 10/21/2022]
Abstract
1H Time-Domain Nuclear Magnetic Resonance (TD-NMR) is used to characterize solutions of antibodies that simulate biologic pharmaceutical formulations. The results from these measurements are compared with those from solutions in which the concentration or identity of the antibody has been altered. TD-NMR is shown to be very sensitive to differences in the amount of antibody in solution, with the ability to detect variations in as low as 2 mg/mL. It is therefore capable, by comparison with data from known formulations, of determining whether a particular sample is likely to be of an authentic biologic formulation. This method expands on the previous use of HPLC, UV/VIS, Near-IR and High-Resolution NMR to detect adulterated pharmaceutical materials. While the sensitivity of the method is high, it is a fingerprinting methodology, illustrating differences but not elucidating their origin. The extracted relaxation times reflect the combined effect of all solutes (antibody, buffer components, etc.) on the solvent (water).
Collapse
Affiliation(s)
- Zahir Akhunzada
- Drug Product Development, Bristol-Myers Squibb via PPD Inc., New Brunswick, NJ 08903, United States.
| | - Yongmei Wu
- Drug Product Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Thomas Haby
- Drug Product Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Dimuthu Jayawickrama
- Drug Product Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Gary McGeorge
- Drug Product Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | | | - Jeffrey Bernstein
- WaveGuide Corporation, Cambridge, Massachusetts 02140, United States
| | - Marcus Semones
- WaveGuide Corporation, Cambridge, Massachusetts 02140, United States
| | - Anuji Abraham
- Drug Product Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States.
| |
Collapse
|
10
|
Handl A, López-Lorente ÁI, Handrick R, Mizaikoff B, Hesse F. Infrared attenuated total reflection and 2D fluorescence spectroscopy for the discrimination of differently aggregated monoclonal antibodies. Analyst 2019; 144:6334-6341. [PMID: 31553337 DOI: 10.1039/c9an00424f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antibody aggregates may occur as undesirable by-products during the manufacturing process of biopharmaceutical proteins since parameters such as pH, temperature, ionic strength, protein concentration, oxygen, and shear forces can lead to aggregate formation. These aggregates have to be detected, quantified and removed cost extensively, since they may reduce the safety and efficacy of the product. Protein aggregates can range from small soluble dimers up to large visible agglomerates. Differently aggregated antibody samples were characterized for their soluble and insoluble aggregate concentration by size exclusion chromatography and fluorescence microscopy, respectively. The samples exhibited a high diversity of protein aggregates, which varied in amount, size and shape. For secondary structure characterization, infrared attenuated total reflection (IR-ATR) and two-dimensional fluorescence (2D-FL) spectroscopy were applied. Using direct spectroscopy, only marginal differences of various antibody aggregates were evident. However, using appropriate chemometric strategies, the evaluation of IR-ATR and 2D-FL spectra yielded the discrimination of differently aggregated antibody samples with yet unprecedented precision.
Collapse
Affiliation(s)
- Alina Handl
- Biberach University, Institute of Applied Biotechnology, Hubertus-Liebrecht-Str. 35, 88400 Biberach, Germany.
| | | | | | | | | |
Collapse
|
11
|
Wu H, Randolph TW. Rapid Quantification of Protein Particles in High-Concentration Antibody Formulations. J Pharm Sci 2019; 108:1110-1116. [DOI: 10.1016/j.xphs.2018.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/21/2018] [Accepted: 10/11/2018] [Indexed: 11/16/2022]
|
12
|
Guerra A, von Stosch M, Glassey J. Toward biotherapeutic product real-time quality monitoring. Crit Rev Biotechnol 2019; 39:289-305. [DOI: 10.1080/07388551.2018.1524362] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- André Guerra
- School of Chemical Engineering and Advanced Materials, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Moritz von Stosch
- School of Chemical Engineering and Advanced Materials, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jarka Glassey
- School of Chemical Engineering and Advanced Materials, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
13
|
de Marco A, Ferrer-Miralles N, Garcia-Fruitós E, Mitraki A, Peternel S, Rinas U, Trujillo-Roldán MA, Valdez-Cruz NA, Vázquez E, Villaverde A. Bacterial inclusion bodies are industrially exploitable amyloids. FEMS Microbiol Rev 2019; 43:53-72. [PMID: 30357330 DOI: 10.1093/femsre/fuy038] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022] Open
Abstract
Understanding the structure, functionalities and biology of functional amyloids is an issue of emerging interest. Inclusion bodies, namely protein clusters formed in recombinant bacteria during protein production processes, have emerged as unanticipated, highly tunable models for the scrutiny of the physiology and architecture of functional amyloids. Based on an amyloidal skeleton combined with varying amounts of native or native-like protein forms, bacterial inclusion bodies exhibit an unusual arrangement that confers mechanical stability, biological activity and conditional protein release, being thus exploitable as versatile biomaterials. The applicability of inclusion bodies in biotechnology as enriched sources of protein and reusable catalysts, and in biomedicine as biocompatible topographies, nanopills or mimetics of endocrine secretory granules has been largely validated. Beyond these uses, the dissection of how recombinant bacteria manage the aggregation of functional protein species into structures of highly variable complexity offers insights about unsuspected connections between protein quality (conformational status compatible with functionality) and cell physiology.
Collapse
Affiliation(s)
- Ario de Marco
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska Cesta 13, 5000 Nova Gorica, Slovenia
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina (IBB), Carrer de la Vall Moronta s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,Departament de Genètica i de Microbiologia, Carrer de la Vall Moronta s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Carrer de la Vall Moronta s/n, 08193 Cerdanyola del Vallès, Spain
| | - Elena Garcia-Fruitós
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Torre Marimon, 08140 Caldes de Montbui, Barcelona, Spain
| | - Anna Mitraki
- Department of Materials Science and Technology, University of Crete, Vassilika Vouton, 70013 Heraklion, Crete, Greece.,Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), N. Plastira 100, Vassilika Vouton, 70013 Heraklion, Crete, Greece
| | | | - Ursula Rinas
- Leibniz University of Hannover, Technical Chemistry and Life Science, 30167 Hannover, Germany.,Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Mauricio A Trujillo-Roldán
- Programa de Investigación de Producción de Biomoléculas, Unidad de Bioprocesos, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Norma A Valdez-Cruz
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina (IBB), Carrer de la Vall Moronta s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,Departament de Genètica i de Microbiologia, Carrer de la Vall Moronta s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Carrer de la Vall Moronta s/n, 08193 Cerdanyola del Vallès, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina (IBB), Carrer de la Vall Moronta s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,Departament de Genètica i de Microbiologia, Carrer de la Vall Moronta s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Carrer de la Vall Moronta s/n, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
14
|
Wang B, Albanetti T, Miro-Quesada G, Flack L, Li L, Klover J, Burson K, Evans K, Ivory W, Bowen M, Schoner R, Hawley-Nelson P. High-throughput screening of antibody-expressing CHO clones using an automated shaken deep-well system. Biotechnol Prog 2018; 34:1460-1471. [PMID: 30298994 PMCID: PMC6587815 DOI: 10.1002/btpr.2721] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/30/2018] [Accepted: 09/20/2018] [Indexed: 12/15/2022]
Abstract
Biopharmaceutical protein manufacturing requires the highest producing cell lines to satisfy current multiple grams per liter requirements. Screening more clones increases the probability of identifying the high producers within the pool of available transfectant candidate cell lines. For the predominant industry mammalian host cell line, Chinese hamster ovary (CHO), traditional static‐batch culture screening does not correlate with the suspension fed‐batch culture used in manufacturing, and thus has little predictive utility. Small scale fed‐batch screens in suspension culture correlate better with bioreactor processes but a limited number of clones can be screened manually. Scaled‐down systems, such as shaken deep well plates, combined with automated liquid handling, offer a way for a limited number of scientists to screen many clones. A statistical analysis determined that 384 is the optimal number of clones to screen, with a 99% probability that six clones in the 95th percentile for productivity are included in the screen. To screen 384 clones efficiently by the predictive method of suspension fed‐batch, the authors developed a shaken deep‐well plate culturing platform, with an automated liquid handling system integrating cell counting and protein titering instruments. Critical factors allowing deep‐well suspension culture to correlate with shake flask culture were agitation speed and culture volume. Using our automated system, one scientist can screen five times more clones than by manual fed‐batch shake‐flask or shaken culture tube screens and can identify cell lines for some therapeutic protein projects with production levels greater than 6 g/L. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1460–1471, 2018
Collapse
Affiliation(s)
- Benjamin Wang
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Thomas Albanetti
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | | | - Layla Flack
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Lina Li
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Judith Klover
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Kerri Burson
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Krista Evans
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - William Ivory
- Analytical Biochemistry, MedImmune, Gaithersburg, Maryland, 20878
| | - Michael Bowen
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Ronald Schoner
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| | - Pamela Hawley-Nelson
- Cell Culture and Fermentation Sciences, MedImmune, Gaithersburg, Maryland, 20878
| |
Collapse
|
15
|
Wang W, Roberts CJ. Protein aggregation – Mechanisms, detection, and control. Int J Pharm 2018; 550:251-268. [DOI: 10.1016/j.ijpharm.2018.08.043] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
|
16
|
Debottlenecking protein secretion and reducing protein aggregation in the cellular host. Curr Opin Biotechnol 2018; 53:151-157. [DOI: 10.1016/j.copbio.2018.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 01/05/2023]
|
17
|
Oshinbolu S, Shah R, Finka G, Molloy M, Uden M, Bracewell DG. Evaluation of fluorescent dyes to measure protein aggregation within mammalian cell culture supernatants. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2018; 93:909-917. [PMID: 29540956 PMCID: PMC5838538 DOI: 10.1002/jctb.5519] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 06/28/2023]
Abstract
BACKGROUND A current challenge in bioprocessing is the ability to analyse critical quality attributes such as aggregation without prior purification. This study evaluated the use of fluorescent dyes (Bis-ANS, SYPRO Orange, Thioflavin T and ProteoStat) to characterise mAb aggregates in Chinese hamster ovary clarified cultures. RESULTS The null and mAb culture supernatants showed an increase in fluorescence intensity over the duration of the culture. The null cultures on day 14 saw a rapid increase in fluorescence intensity; day 10 to day 14, Bis-ANS and Thioflavin T had average increases of 21% and 48%, respectively, whereas ProteoStat and SYPRO Orange showed an average increase of 60%. Higher fluorescence intensity on day 14 with the null cultures, also correlated with loss of viability. CONCLUSION Fluorescent dyes are not a specific indicator of mAb aggregation, but rather an indicator of overall protein aggregation or high molecular weight species. SYPRO Orange was more sensitive at detecting very large molecular weight species and ProteoStat seemed better suited to smaller aggregates. Although the assay cannot be used to measure mAb aggregates in cell culture, it could be used to aid cell line selection in maximising viabilities and minimising the amount of aggregates. © 2017 The Authors. Journal of Chemical Technology & Biotechnology published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Sheun Oshinbolu
- Dept of Biochemical EngineeringUniversity College LondonLondonUK
- GlaxoSmithKlineStevenageHertsUK
| | | | | | | | | | | |
Collapse
|
18
|
Kim TK, Park CS, Jang J, Kim MR, Na HJ, Lee K, Kim HJ, Heo K, Yoo BC, Kim YM, Lee JW, Kim SJ, Kim ES, Kim DY, Cha K, Lee TG, Lee S. Inhibition of VEGF-dependent angiogenesis and tumor angiogenesis by an optimized antibody targeting CLEC14a. Mol Oncol 2018; 12:356-372. [PMID: 29316206 PMCID: PMC5830631 DOI: 10.1002/1878-0261.12169] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/12/2022] Open
Abstract
The C‐type lectin‐like domain of CLEC14a (CLEC14a‐C‐type lectin‐like domain [CTLD]) is a key domain that mediates endothelial cell–cell contacts in angiogenesis. However, the role of CLEC14a‐CTLD in pathological angiogenesis has not yet been clearly elucidated. In this study, through complementarity‐determining region grafting, consecutive deglycosylation, and functional isolation, we generated a novel anti‐angiogenic human monoclonal antibody that specifically targets CLEC14a‐CTLD and that shows improved stability and homogeneity relative to the parental antibody. We found that this antibody directly inhibits CLEC14a‐CTLD‐mediated endothelial cell–cell contact and simultaneously downregulates expression of CLEC14a on the surface of endothelial cells. Using various in vitro and in vivo functional assays, we demonstrated that this antibody effectively suppresses vascular endothelial growth factor (VEGF)‐dependent angiogenesis and tumor angiogenesis of SNU182 human hepatocellular carcinoma, CFPAC‐1 human pancreatic cancer, and U87 human glioma cells. Furthermore, we also found that this antibody significantly inhibits tumor angiogenesis of HCT116 and bevacizumab‐adapted HCT116 human colorectal cancer cells. These findings suggest that antibody targeting of CLEC14a‐CTLD has the potential to suppress VEGF‐dependent angiogenesis and tumor angiogenesis and that CLEC14a‐CTLD may be a novel anti‐angiogenic target for VEGF‐dependent angiogenesis and tumor angiogenesis.
Collapse
Affiliation(s)
- Taek-Keun Kim
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Chang Sik Park
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Jihye Jang
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Mi Ra Kim
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Hee-Jun Na
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Kangseung Lee
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Hyun Jung Kim
- Research Institute, National Cancer Center, Goyang, South Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang, South Korea
| | - Byong Chul Yoo
- Research Institute, National Cancer Center, Goyang, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Je-Wook Lee
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Su Jin Kim
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Eun Sung Kim
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Dae Young Kim
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Kiweon Cha
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Tae Gyu Lee
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Sukmook Lee
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| |
Collapse
|
19
|
Paul AJ, Handrick R, Ebert S, Hesse F. Identification of process conditions influencing protein aggregation in Chinese hamster ovary cell culture. Biotechnol Bioeng 2018; 115:1173-1185. [DOI: 10.1002/bit.26534] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/16/2017] [Accepted: 12/20/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Albert J. Paul
- Institute of Applied Biotechnology; Biberach University of Applied Sciences; Biberach Germany
| | - René Handrick
- Institute of Applied Biotechnology; Biberach University of Applied Sciences; Biberach Germany
| | - Sybille Ebert
- Institute of Applied Biotechnology; Biberach University of Applied Sciences; Biberach Germany
| | - Friedemann Hesse
- Institute of Applied Biotechnology; Biberach University of Applied Sciences; Biberach Germany
| |
Collapse
|
20
|
Estimating Extrinsic Dyes for Fluorometric Online Monitoring of Antibody Aggregation in CHO Fed-Batch Cultivations. Bioengineering (Basel) 2017; 4:bioengineering4030065. [PMID: 28952544 PMCID: PMC5615311 DOI: 10.3390/bioengineering4030065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 07/15/2017] [Accepted: 07/17/2017] [Indexed: 12/13/2022] Open
Abstract
Multi-wavelength fluorescence spectroscopy was evaluated in this work as tool for real-time monitoring of antibody aggregation in CHO fed-batch cultivations via partial least square (PLS) modeling. Therefore, we used the extrinsic fluorescence dyes 1-anilinonaphthalene-8-sulfonate (ANS), 4,4′-bis-1-anilinonaphthalene-8-sulfonate (Bis-ANS), or Thioflavin T (ThT) as medium additives. This is a new application area, since these dyes are commonly used for aggregate detection during formulation development. We determined the half maximum inhibitory concentrations of ANS (203 ± 11 µmol·L−1), Bis-ANS (5 ± 0.5 µmol·L−1), and ThT (3 ± 0.2 µmol·L−1), and selected suitable concentrations for this application. The results showed that the emission signals of non-covalent dye antibody aggregate interaction superimposed the fluorescence signals originating from feed medium and cell culture. The fluorescence datasets were subsequently used to build PLS models, and the dye-related elevated fluorescence signals dominated the model calibration. The soft sensors based on ANS and Bis-ANS signals showed high predictability with a low error of prediction (1.7 and 2.3 mg·mL−1 aggregates). In general, the combination of extrinsic dye and used concentration influenced the predictability. Furthermore, the ThT soft sensor indicated that the intrinsic fluorescence of the culture might be sufficient to predict antibody aggregation online.
Collapse
|
21
|
High-throughput analysis of sub-visible mAb aggregate particles using automated fluorescence microscopy imaging. Anal Bioanal Chem 2017; 409:4149-4156. [DOI: 10.1007/s00216-017-0362-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 04/10/2017] [Indexed: 12/29/2022]
|
22
|
Watanabe H, Yageta S, Imamura H, Honda S. Biosensing Probe for Quality Control Monitoring of the Structural Integrity of Therapeutic Antibodies. Anal Chem 2016; 88:10095-10101. [PMID: 27700033 DOI: 10.1021/acs.analchem.6b02526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ideal quality control of therapeutic antibodies involves analytical techniques with high-sensitivity, high-resolution, and high-throughput performance. Few technologies that assess the physicochemical heterogeneity of antibodies, however, meet all the required demands. We developed a biosensing method for the quality control of therapeutic antibodies based on an artificial protein, AF.2A1, which discriminates between the native and the non-native three-dimensional structures of immunoglobulin G (IgG). AF.2A1 specifically recognized non-native IgG spiked into a solution of native IgG, thereby making it possible to detect contamination by a small amount of non-native IgG, which is difficult using conventional size-based separation or spectroscopic techniques. Using AF.2A1 as an analytical probe, we determined the concentration of non-native IgG formed under various pH conditions. The probe was also applicable to accelerated tests of the long-term stability of a therapeutic antibody, allowing monitoring of the formation of non-native IgG at elevated temperatures and extended periods of storage. AF.2A1, a proteinous probe, can be combined with established methods or devices to achieve high-throughput assays and provides the potential for probe-based biosensing for the quality control of therapeutic antibodies.
Collapse
Affiliation(s)
- Hideki Watanabe
- Biomedical Research Institute, the National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Seiki Yageta
- Biomedical Research Institute, the National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, the University of Tokyo , 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Hiroshi Imamura
- Biomedical Research Institute, the National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Shinya Honda
- Biomedical Research Institute, the National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, the University of Tokyo , 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| |
Collapse
|
23
|
Purdie JL, Kowle RL, Langland AL, Patel CN, Ouyang A, Olson DJ. Cell culture media impact on drug product solution stability. Biotechnol Prog 2016; 32:998-1008. [DOI: 10.1002/btpr.2289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/20/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Jennifer L. Purdie
- Bioproduct Research and Development, Lilly Research Laboratories, Eli Lilly and Company; Indianapolis IN 46285
| | - Ronald L. Kowle
- Bioproduct Research and Development, Lilly Research Laboratories, Eli Lilly and Company; Indianapolis IN 46285
| | - Amie L. Langland
- Bioproduct Research and Development, Lilly Research Laboratories, Eli Lilly and Company; Indianapolis IN 46285
| | - Chetan N. Patel
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company; Indianapolis IN 46285
| | - Anli Ouyang
- Bioproduct Research and Development, Lilly Research Laboratories, Eli Lilly and Company; Indianapolis IN 46285
| | - Donald J. Olson
- Bioproduct Research and Development, Lilly Research Laboratories, Eli Lilly and Company; Indianapolis IN 46285
| |
Collapse
|
24
|
Patel KA, Sethi R, Dhara AR, Roy I. Challenges with osmolytes as inhibitors of protein aggregation: Can nucleic acid aptamers provide an answer? Int J Biol Macromol 2016; 100:75-88. [PMID: 27156694 DOI: 10.1016/j.ijbiomac.2016.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 02/07/2023]
Abstract
Protein aggregation follows some common motifs. Whether in the formation of inclusion bodies in heterologous overexpression systems or inclusions in protein conformational diseases, or aggregation during storage or transport of protein formulations, aggregates form cross beta-sheet structures and stain with amyloidophilic dyes like Thioflavin T and Congo Red, irrespective of the concerned protein. Traditionally, osmolytes are used to stabilize proteins against stress conditions. They are employed right from protein expression, through production and purification, to formulation and administration. As osmolytes interact with the solvent, the differential effect of the stress condition on the solvent mostly determines the effect of the osmolyte on protein stability. Nucleic acid aptamers, on the other hand, are highly specific for their targets. When selected against monomeric, natively folded proteins, they bind to them with very high affinity. This binding inhibits the unfolding of the protein and/or monomer-monomer interaction which are the initial common steps of protein aggregation. Thus, by changing the approach to a protein-centric model, aptamers are able to function as universal stabilizers of proteins. The review discusses cases where osmolytes were unable to provide stabilization to proteins against different stress conditions, a gap which the aptamers seem to be able to fill.
Collapse
Affiliation(s)
- Kinjal A Patel
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Ratnika Sethi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Anita R Dhara
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India.
| |
Collapse
|