251
|
Jia H, Guo Y, Song X, Shao C, Wu J, Ma J, Shi M, Miao Y, Li R, Wang D, Tian Z, Xiao W. Elimination of N-glycosylation by site mutation further prolongs the half-life of IFN-α/Fc fusion proteins expressed in Pichia pastoris. Microb Cell Fact 2016; 15:209. [PMID: 27927205 PMCID: PMC5142404 DOI: 10.1186/s12934-016-0601-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 11/23/2016] [Indexed: 12/16/2022] Open
Abstract
Background Interferon (IFN)-α has been commonly used as an antiviral drug worldwide; however, its short half-life in circulation due to its low molecular weight and sensitivity to proteases impacts its efficacy and patient compliance. Results In this study, we present an IgG1 Fc fusion strategy to improve the circulation half-life of IFN-α. Three different forms of IgG1 Fc fragments, including the wild type, aglycosylated homodimer and aglycosylated single chain, were each fused with IFN-α and designated as IFN-α/Fc-WT, IFN-α/Fc-MD, and IFN-α/Fc-SC, respectively. The recombinant proteins were expressed in Pichia pastoris and tested using antiviral and pharmacokinetic assays in comparison with the commercial pegylated-IFN-α (PEG-IFN-α). The in vitro study demonstrated that IFN-α/Fc-SC has the highest antiviral activity, while IFN-α/Fc-WT and IFN-α/Fc-MD exhibited antiviral activities comparable to that of PEG-IFN-α. The in vivo pharmacokinetic assay showed that both IFN-α/Fc-WT and IFN-α/Fc-MD have a longer half-life than PEG-IFN-α in SD rats, but IFN-α/Fc-SC has the shortest half-life among them. Importantly, the circulating half-life of 68.3 h for IFN-α/Fc-MD was significantly longer than those of 38.2 h for IFN-α/Fc-WT and 22.2 h for PEG-IFN-α. Conclusions The results demonstrate that the elimination of N-glycosylation by mutation of putative N-glycosylation site further prolongs the half-life of the IFN-α/Fc fusion protein and could present an alternative strategy for extending the half-life of low-molecular-weight proteins expressed by P. pastoris for in vivo studies as well as for future clinical applications. Electronic supplementary material The online version of this article (doi:10.1186/s12934-016-0601-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hao Jia
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China
| | - Yugang Guo
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China. .,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China. .,Anhui Engineering Research Center of Recombinant Protein Pharmaceutical Biotechnology, Institute of Advanced Technology, University of Science and Technology of China, Hefei, China.
| | - Xiaoping Song
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Department of Pharmacy, Anhui Medical College, Hefei, China
| | - Changsheng Shao
- Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China.,Anhui Engineering Research Center of Recombinant Protein Pharmaceutical Biotechnology, Institute of Advanced Technology, University of Science and Technology of China, Hefei, China
| | - Jing Wu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China.,Anhui Engineering Research Center of Recombinant Protein Pharmaceutical Biotechnology, Institute of Advanced Technology, University of Science and Technology of China, Hefei, China
| | - Jiajia Ma
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China.,Anhui Engineering Research Center of Recombinant Protein Pharmaceutical Biotechnology, Institute of Advanced Technology, University of Science and Technology of China, Hefei, China
| | - Mingyang Shi
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China
| | - Yuhui Miao
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China
| | - Rui Li
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China
| | - Dong Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China.,Anhui Engineering Research Center of Recombinant Protein Pharmaceutical Biotechnology, Institute of Advanced Technology, University of Science and Technology of China, Hefei, China
| | - Weihua Xiao
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, China. .,Hefei National Laboratory for Physical Sciences at the Microscale, Engineering Technology Research Center of Biotechnology Drugs, Anhui Province, University of Science and Technology of China, Hefei, China. .,Anhui Engineering Research Center of Recombinant Protein Pharmaceutical Biotechnology, Institute of Advanced Technology, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
252
|
Hayes JM, Wormald MR, Rudd PM, Davey GP. Fc gamma receptors: glycobiology and therapeutic prospects. J Inflamm Res 2016; 9:209-219. [PMID: 27895507 PMCID: PMC5118039 DOI: 10.2147/jir.s121233] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Therapeutic antibodies hold great promise for the treatment of cancer and autoimmune diseases, and developments in antibody–drug conjugates and bispecific antibodies continue to enhance treatment options for patients. Immunoglobulin (Ig) G antibodies are proteins with complex modifications, which have a significant impact on their function. The most important of these modifications is glycosylation, the addition of conserved glycans to the antibody Fc region, which is critical for its interaction with the immune system and induction of effector activities such as antibody-dependent cell cytotoxicity, complement activation and phagocytosis. Communication of IgG antibodies with the immune system is controlled and mediated by Fc gamma receptors (FcγRs), membrane-bound proteins, which relay the information sensed and gathered by antibodies to the immune system. These receptors are also glycoproteins and provide a link between the innate and adaptive immune systems. Recent information suggests that this receptor glycan modification is also important for the interaction with antibodies and downstream immune response. In this study, the current knowledge on FcγR glycosylation is discussed, and some insight into its role and influence on the interaction properties with IgG, particularly in the context of biotherapeutics, is provided. For the purpose of this study, other Fc receptors such as FcαR, FcεR or FcRn are not discussed extensively, as IgG-based antibodies are currently the only therapeutic antibody-based products on the market. In addition, FcγRs as therapeutics and therapeutic targets are discussed, and insight into and comment on the therapeutic aspects of receptor glycosylation are provided.
Collapse
Affiliation(s)
- Jerrard M Hayes
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Mark R Wormald
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, UK
| | - Pauline M Rudd
- NIBRT Glycoscience Group, National Institute for Bioprocessing, Research and Training, Dublin, Ireland
| | - Gavin P Davey
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| |
Collapse
|
253
|
Tsui P, Higazi DR, Wu Y, Dunmore R, Solier E, Kasali T, Bond NJ, Huntington C, Carruthers A, Hood J, Borrok MJ, Barnes A, Rickert K, Phipps S, Shirinian L, Zhu J, Bowen MA, Dall'Acqua W, Murray LA. The TGF-β inhibitory activity of antibody 37E1B5 depends on its H-CDR2 glycan. MAbs 2016; 9:104-113. [PMID: 27834568 DOI: 10.1080/19420862.2016.1255390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Excessive transforming growth factor (TGF)-β is associated with pro-fibrotic responses in lung disease, yet it also plays essential roles in tissue homeostasis and autoimmunity. Therefore, selective inhibition of excessive and aberrant integrin-mediated TGF-β activation via targeting the α-v family of integrins is being pursued as a therapeutic strategy for chronic lung diseases, to mitigate any potential safety concerns with global TGF-β inhibition. In this work, we reveal a novel mechanism of inhibiting TGF-β activation utilized by an αvβ8 targeting antibody, 37E1B5. This antibody blocks TGF-β activation while not inhibiting cell adhesion. We show that an N-linked complex-type Fab glycan in H-CDR2 of 37E1B5 is directly involved in the inhibition of latent TGF-β activation. Removal of the Fab N-glycosylation site by single amino acid substitution, or removal of N-linked glycans by enzymatic digestion, drastically reduced the antibody's ability to inhibit latency-associated peptide (LAP) and αvβ8 association, and TGF-β activation in an αvβ8-mediated TGF-β signaling reporter assay. Our results indicate a non-competitive, allosteric inhibition of 37E1B5 on αvβ8-mediated TGF-β activation. This unique, H-CDR2 glycan-mediated mechanism may account for the potent but tolerable TGF-b activation inhibition and lack of an effect on cellular adhesion by the antibody.
Collapse
Affiliation(s)
- Ping Tsui
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Daniel R Higazi
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | - Yanli Wu
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Rebecca Dunmore
- c Respiratory, Inflammation and Autoimmunity, MedImmune Ltd , Cambridge , UK
| | - Emilie Solier
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | - Toyin Kasali
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | - Nicholas J Bond
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | | | - Alan Carruthers
- c Respiratory, Inflammation and Autoimmunity, MedImmune Ltd , Cambridge , UK
| | - John Hood
- e Translational Sciences, Medimmune Ltd ., Cambridge , UK
| | - M Jack Borrok
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Arnita Barnes
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Keith Rickert
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Sandrina Phipps
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Lena Shirinian
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Jie Zhu
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Michael A Bowen
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - William Dall'Acqua
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Lynne A Murray
- c Respiratory, Inflammation and Autoimmunity, MedImmune Ltd , Cambridge , UK
| |
Collapse
|
254
|
Kizhedath A, Wilkinson S, Glassey J. Applicability of predictive toxicology methods for monoclonal antibody therapeutics: status Quo and scope. Arch Toxicol 2016; 91:1595-1612. [PMID: 27766364 PMCID: PMC5364268 DOI: 10.1007/s00204-016-1876-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/12/2016] [Indexed: 12/31/2022]
Abstract
Biopharmaceuticals, monoclonal antibody (mAb)-based therapeutics in particular, have positively impacted millions of lives. MAbs and related therapeutics are highly desirable from a biopharmaceutical perspective as they are highly target specific and well tolerated within the human system. Nevertheless, several mAbs have been discontinued or withdrawn based either on their inability to demonstrate efficacy and/or due to adverse effects. Approved monoclonal antibodies and derived therapeutics have been associated with adverse effects such as immunogenicity, cytokine release syndrome, progressive multifocal leukoencephalopathy, intravascular haemolysis, cardiac arrhythmias, abnormal liver function, gastrointestinal perforation, bronchospasm, intraocular inflammation, urticaria, nephritis, neuropathy, birth defects, fever and cough to name a few. The advances made in this field are also impeded by a lack of progress in bioprocess development strategies as well as increasing costs owing to attrition, wherein the lack of efficacy and safety accounts for nearly 60 % of all factors contributing to attrition. This reiterates the need for smarter preclinical development using quality by design-based approaches encompassing carefully designed predictive models during early stages of drug development. Different in vitro and in silico methods are extensively used for predicting biological activity as well as toxicity during small molecule drug development; however, their full potential has not been utilized for biological drug development. The scope of in vitro and in silico tools in early developmental stages of monoclonal antibody-based therapeutics production and how it contributes to lower attrition rates leading to faster development of potential drug candidates has been evaluated. The applicability of computational toxicology approaches in this context as well as the pitfalls and promises of extending such techniques to biopharmaceutical development has been highlighted.
Collapse
Affiliation(s)
- Arathi Kizhedath
- Chemical Engineering and Advanced Materials, Newcastle University, Newcastle upon Tyne, NE17RU, UK. .,Medical Toxicology Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4AA, UK.
| | - Simon Wilkinson
- Medical Toxicology Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4AA, UK
| | - Jarka Glassey
- Chemical Engineering and Advanced Materials, Newcastle University, Newcastle upon Tyne, NE17RU, UK
| |
Collapse
|
255
|
Mascarenhas JX, Korokhov N, Burger L, Kassim A, Tuter J, Miller D, Borgschulte T, George HJ, Chang A, Pintel DJ, Onions D, Kayser KJ. Genetic engineering of CHO cells for viral resistance to minute virus of mice. Biotechnol Bioeng 2016; 114:576-588. [PMID: 27642072 DOI: 10.1002/bit.26186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
Contamination by the parvovirus minute virus of mice (MVM) remains a challenge in Chinese hamster ovary (CHO) biopharmaceutical production processes. Although infrequent, infection of a bioreactor can be catastrophic for a manufacturer, can impact patient drug supply and safety, and can have regulatory implications. We evaluated engineering a CHO parental cell line (CHOZN® GS-/- ) to create a new host cell line that is resistant to MVM infection by modifying the major receptors used by the virus to enter cells. Attachment to a cell surface receptor is a key first step in the infection cycle for many viruses. While the exact functional receptor for MVM binding to CHO cell surface is unknown, sialic acid on the cell surface has been implicated. In this work, we used the zinc finger nuclease gene editing technology to validate the role of sialic acid on the cell surface in the binding and internalization of the MVM virus. Our approach was to systematically mutate genes involved in cell surface sialylation and then challenge each cell line for their ability to resist viral entry and propagation. To test the importance of sialylation, the following genes were knocked out: the CMP-sialic acid transporter, solute carrier family 35A1 (Slc35a1), the core 1-β-1,3-galactosyltransferase-1 specific chaperone (Cosmc), and mannosyl (α-1,3-)-glycoprotein β-1,2-N-acetylglucosaminyltransferase (Mgat1) as well as members of the sialyltransferase family. Slc35a1 is responsible for transporting sialic acid into the Golgi. Knocking out function of this gene in a cell results in asialylated glycan structures, thus eliminating the ability of MVM to bind to and enter the cell. The complete absence of sialic acid on the Slc35a1 knockout cell line led to complete resistance to MVM infection. The Cosmc and Mgat1 knockouts also show significant inhibition of infection likely due to their effect on decreasing cell surface sialic acid. Previously in vitro glycan analysis has been used to elucidate the precise sialic acid structures required for MVM binding and internalization. In this work, we performed the sequential knockout of various sialyltransferases that add terminal sialic acid to glycans with different linkage specificities. Cell lines with modifications of the various genes included in this study resulted in varying effects on MVM infection expanding on the knowledge of MVM receptors. MVM resistant host cell lines were also tested for the production of model recombinant proteins. Our data demonstrate that resistance against the MVM virus can be incorporated into CHO production cell lines, adding another level of defense against the devastating financial consequences of MVM infection without compromising recombinant protein yield or quality. Biotechnol. Bioeng. 2017;114: 576-588. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Lisa Burger
- University of Missouri School of Medicine, Columbia, Missouri
| | - Ademola Kassim
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Jason Tuter
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Daniel Miller
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Trissa Borgschulte
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Henry J George
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Audrey Chang
- Bioreliance, MilliporeSigma, Rockville, Maryland
| | - David J Pintel
- University of Missouri School of Medicine, Columbia, Missouri
| | - David Onions
- Bioreliance, MilliporeSigma, Rockville, Maryland
| | - Kevin J Kayser
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| |
Collapse
|
256
|
Chiang AW, Li S, Spahn PN, Richelle A, Kuo CC, Samoudi M, Lewis NE. Modulating carbohydrate-protein interactions through glycoengineering of monoclonal antibodies to impact cancer physiology. Curr Opin Struct Biol 2016; 40:104-111. [PMID: 27639240 DOI: 10.1016/j.sbi.2016.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/08/2016] [Accepted: 08/29/2016] [Indexed: 01/05/2023]
Abstract
Diverse glycans on proteins impact cell and organism physiology, along with drug activity. Since many protein-based biotherapeutics are glycosylated and these glycans have biological activity, there is a desire to engineer glycosylation for recombinant protein-based biotherapeutics. Engineered glycosylation can impact the recombinant protein efficacy and also influence many cell pathways by first changing glycan-protein interactions and consequently modulating disease physiologies. However, its complexity is enormous. Recent advances in glycoengineering now make it easier to modulate protein-glycan interactions. Here, we discuss how engineered glycans contribute to therapeutic monoclonal antibodies (mAbs) in the treatment of cancers, how these glycoengineered therapeutic mAbs affect the transformed phenotypes and downstream cell pathways. Furthermore, we suggest how systems biology can help in the next generation mAb glycoengineering process by aiding in data analysis and guiding engineering efforts to tailor mAb glycan and ultimately drug efficacy, safety and affordability.
Collapse
Affiliation(s)
- Austin Wt Chiang
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Shangzhong Li
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, CA, USA
| | - Philipp N Spahn
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Anne Richelle
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Chih-Chung Kuo
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, CA, USA
| | - Mojtaba Samoudi
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA.
| |
Collapse
|
257
|
Comprehensive N-Glycan Profiling of Avian Immunoglobulin Y. PLoS One 2016; 11:e0159859. [PMID: 27459092 PMCID: PMC4961449 DOI: 10.1371/journal.pone.0159859] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 07/08/2016] [Indexed: 01/30/2023] Open
Abstract
Recent exploitation of the avian immune system has highlighted its suitability for the generation of high-quality, high-affinity antibodies to a wide range of antigens for a number of therapeutic and biotechnological applications. The glycosylation profile of potential immunoglobulin therapeutics is species specific and is heavily influenced by the cell-line/culture conditions used for production. Hence, knowledge of the carbohydrate moieties present on immunoglobulins is essential as certain glycan structures can adversely impact their physicochemical and biological properties. This study describes the detailed N-glycan profile of IgY polyclonal antibodies from the serum of leghorn chickens using a fully quantitative high-throughput N-glycan analysis approach, based on ultra-performance liquid chromatography (UPLC) separation of released glycans. Structural assignments revealed serum IgY to contain complex bi-, tri- and tetra-antennary glycans with or without core fucose and bisects, hybrid and high mannose glycans. High sialic acid content was also observed, with the presence of rare sialic acid structures, likely polysialic acids. It is concluded that IgY is heavily decorated with complex glycans; however, no known non-human or immunogenic glycans were identified. Thus, IgY is a potentially promising candidate for immunoglobulin-based therapies for the treatment of various infectious diseases.
Collapse
|
258
|
Abstract
Antibody drug conjugates (ADCs) have emerged as a viable option in targeted delivery of highly potent cytotoxic drugs in treatment of solid tumors. At the time of writing, only two ADCs have received regulatory approval with >40 others in clinical development. The first generation ADCs suffered from a lack of specificity in amino acid site-conjugations, yielding statistically heterogeneous stoichiometric ratios of drug molecules per antibody molecule. For the second generation ADCs, however, site-specific amino acid conjugation using enzymatic ligation, introduction of unnatural amino acids, and site-specific protein engineering hold promise to alleviate some of the current technical limitations. The rapid progress in technology platforms and antibody engineering has introduced novel linkers, site-specific conjugation chemistry, and new payload candidates that could possibly be exploited in the context of ADCs. A search using the Clinical Trial Database registry ( www.clinicaltrials.gov ), using the keyword 'antibody drug conjugate', yielded ~270 hits. The main focus of this article is to present a brief overview of the recent developments and current challenges related to ADC development.
Collapse
|
259
|
Moura-da-Silva AM, Almeida MT, Portes-Junior JA, Nicolau CA, Gomes-Neto F, Valente RH. Processing of Snake Venom Metalloproteinases: Generation of Toxin Diversity and Enzyme Inactivation. Toxins (Basel) 2016; 8:toxins8060183. [PMID: 27294958 PMCID: PMC4926149 DOI: 10.3390/toxins8060183] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 12/28/2022] Open
Abstract
Snake venom metalloproteinases (SVMPs) are abundant in the venoms of vipers and rattlesnakes, playing important roles for the snake adaptation to different environments, and are related to most of the pathological effects of these venoms in human victims. The effectiveness of SVMPs is greatly due to their functional diversity, targeting important physiological proteins or receptors in different tissues and in the coagulation system. Functional diversity is often related to the genetic diversification of the snake venom. In this review, we discuss some published evidence that posit that processing and post-translational modifications are great contributors for the generation of functional diversity and for maintaining latency or inactivation of enzymes belonging to this relevant family of venom toxins.
Collapse
Affiliation(s)
- Ana M Moura-da-Silva
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo CEP 05503-900, Brazil.
| | - Michelle T Almeida
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo CEP 05503-900, Brazil.
| | - José A Portes-Junior
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo CEP 05503-900, Brazil.
| | - Carolina A Nicolau
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Rio de Janeiro CEP 21040-360, Brazil.
| | - Francisco Gomes-Neto
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Rio de Janeiro CEP 21040-360, Brazil.
| | - Richard H Valente
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Rio de Janeiro CEP 21040-360, Brazil.
| |
Collapse
|
260
|
Cho IH, Lee N, Song D, Jung SY, Bou-Assaf G, Sosic Z, Zhang W, Lyubarskaya Y. Evaluation of the structural, physicochemical, and biological characteristics of SB4, a biosimilar of etanercept. MAbs 2016; 8:1136-55. [PMID: 27246928 PMCID: PMC4968139 DOI: 10.1080/19420862.2016.1193659] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A biosimilar is a biological medicinal product that is comparable to a reference medicinal product in terms of quality, safety, and efficacy. SB4 was developed as a biosimilar to Enbrel® (etanercept) and was approved as Benepali®, the first biosimilar of etanercept licensed in the European Union (EU). The quality assessment of SB4 was performed in accordance with the ICH comparability guideline and the biosimilar guidelines of the European Medicines Agency and Food and Drug Administration. Extensive structural, physicochemical, and biological testing was performed with state-of-the-art technologies during a side-by-side comparison of the products. Similarity of critical quality attributes (CQAs) was evaluated on the basis of tolerance intervals established from quality data obtained from more than 60 lots of EU-sourced and US-sourced etanercept. Additional quality assessment was focused on a detailed investigation of immunogenicity-related quality attributes, including hydrophobic variants, high-molecular-weight (HMW) species, N-glycolylneuraminic acid (NGNA), and α-1,3-galactose. This comprehensive characterization study demonstrated that SB4 is highly similar to the reference product, Enbrel®, in structural, physicochemical, and biological quality attributes. In addition, the levels of potential immunogenicity-related quality attributes of SB4 such as hydrophobic variants, HMW aggregates, and α-1,3-galactose were less than those of the reference product.
Collapse
Affiliation(s)
- Ick Hyun Cho
- a Quality Evaluation Team , Samsung Bioepis , Incheon , South Korea
| | - Nayoung Lee
- a Quality Evaluation Team , Samsung Bioepis , Incheon , South Korea
| | - Dami Song
- a Quality Evaluation Team , Samsung Bioepis , Incheon , South Korea
| | - Seong Young Jung
- a Quality Evaluation Team , Samsung Bioepis , Incheon , South Korea
| | | | | | | | | |
Collapse
|
261
|
Yang X, Kim SM, Ruzanski R, Chen Y, Moses S, Ling WL, Li X, Wang SC, Li H, Ambrogelly A, Richardson D, Shameem M. Ultrafast and high-throughput N-glycan analysis for monoclonal antibodies. MAbs 2016; 8:706-17. [PMID: 27082290 DOI: 10.1080/19420862.2016.1156828] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Glycosylation is a critical attribute for development and manufacturing of therapeutic monoclonal antibodies (mAbs) in the pharmaceutical industry. Conventional antibody glycan analysis is usually achieved by the 2-aminobenzamide (2-AB) hydrophilic interaction liquid chromatography (HILIC) method following the release of glycans. Although this method produces satisfactory results, it has limited use for screening a large number of samples because it requires expensive reagents and takes several hours or even days for the sample preparation. A simple and rapid glycan analysis method was not available. To overcome these constraints, we developed and compared 2 ultrafast methods for antibody glycan analysis (UMAG) that involve the rapid generation and purification of glycopeptides in either organic solvent or aqueous buffer followed by label-free quantification using matrix-assisted laser desorption/ionization-time of flight mass spectrometry. Both methods quickly yield N-glycan profiles of test antibodies similar to those obtained by the 2-AB HILIC-HPLC method. In addition, the UMAG method performed in aqueous buffer has a shorter assay time of less than 15 min, and enables high throughput analysis in 96-well PCR plates with minimal sample handling. This method, the fastest, and simplest as reported thus far, has been evaluated for glycoprofiling of mAbs expressed under various cell culture conditions, as well as for the evaluation of antibody culture clones and various production batches. Importantly the method sensitively captured changes in glycoprofiles detected by traditional 2-AB HILIC-HPLC or HILIC-UPLC. The simplicity, high speed, and low cost of this method may facilitate basic research and process development for novel mAbs and biosimilar products.
Collapse
Affiliation(s)
- Xiaoyu Yang
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Sunnie Myung Kim
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Richard Ruzanski
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Yuetian Chen
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Sarath Moses
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Wai Lam Ling
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Xiaojuan Li
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Shao-Chun Wang
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Huijuan Li
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | | | - Daisy Richardson
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| | - Mohammed Shameem
- a Bioprocess Development, Merck Research Laboratories, Merck & Co., Inc. , Kenilworth , USA
| |
Collapse
|
262
|
Jefferis R. Posttranslational Modifications and the Immunogenicity of Biotherapeutics. J Immunol Res 2016; 2016:5358272. [PMID: 27191002 PMCID: PMC4848426 DOI: 10.1155/2016/5358272] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/20/2016] [Indexed: 12/23/2022] Open
Abstract
Whilst the amino acid sequence of a protein is determined by its gene sequence, the final structure and function are determined by posttranslational modifications (PTMs), including quality control (QC) in the endoplasmic reticulum (ER) and during passage through the Golgi apparatus. These processes are species and cell specific and challenge the biopharmaceutical industry when developing a production platform for the generation of recombinant biologic therapeutics. Proteins and glycoproteins are also subject to chemical modifications (CMs) both in vivo and in vitro. The individual is naturally tolerant to molecular forms of self-molecules but nonself variants can provoke an immune response with the generation of anti-drug antibodies (ADA); aggregated forms can exhibit enhanced immunogenicity and QC procedures are developed to avoid or remove them. Monoclonal antibody therapeutics (mAbs) are a special case because their purpose is to bind the target, with the formation of immune complexes (ICs), a particular form of aggregate. Such ICs may be removed by phagocytic cells that have antigen presenting capacity. These considerations may frustrate the possibility of ameliorating the immunogenicity of mAbs by rigorous exclusion of aggregates from drug product. Alternate strategies for inducing immunosuppression or tolerance are discussed.
Collapse
Affiliation(s)
- Roy Jefferis
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
263
|
Parsons TB, Struwe WB, Gault J, Yamamoto K, Taylor TA, Raj R, Wals K, Mohammed S, Robinson CV, Benesch JLP, Davis BG. Optimal Synthetic Glycosylation of a Therapeutic Antibody. ACTA ACUST UNITED AC 2016; 128:2407-2413. [PMID: 27546920 PMCID: PMC4974915 DOI: 10.1002/ange.201508723] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/02/2015] [Indexed: 11/25/2022]
Abstract
Glycosylation patterns in antibodies critically determine biological and physical properties but their precise control is a significant challenge in biology and biotechnology. We describe herein the optimization of an endoglycosidase‐catalyzed glycosylation of the best‐selling biotherapeutic Herceptin, an anti‐HER2 antibody. Precise MS analysis of the intact four‐chain Ab heteromultimer reveals nonspecific, non‐enzymatic reactions (glycation), which are not detected under standard denaturing conditions. This competing reaction, which has hitherto been underestimated as a source of side products, can now be minimized. Optimization allowed access to the purest natural form of Herceptin to date (≥90 %). Moreover, through the use of a small library of sugars containing non‐natural functional groups, Ab variants containing defined numbers of selectively addressable chemical tags (reaction handles at Sia C1) in specific positions (for attachment of cargo molecules or “glycorandomization”) were readily generated.
Collapse
Affiliation(s)
- Thomas B Parsons
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Weston B Struwe
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Joseph Gault
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Keisuke Yamamoto
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Thomas A Taylor
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Ritu Raj
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Kim Wals
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK; Department of Pharmacology University of Oxford Oxford OX1 3QT UK
| | - Shabaz Mohammed
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Carol V Robinson
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Justin L P Benesch
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Benjamin G Davis
- Department of Chemistry University of Oxford Chemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
264
|
Parsons TB, Struwe WB, Gault J, Yamamoto K, Taylor TA, Raj R, Wals K, Mohammed S, Robinson CV, Benesch JLP, Davis BG. Optimal Synthetic Glycosylation of a Therapeutic Antibody. Angew Chem Int Ed Engl 2016; 55:2361-7. [PMID: 26756880 PMCID: PMC4973692 DOI: 10.1002/anie.201508723] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/02/2015] [Indexed: 12/11/2022]
Abstract
Glycosylation patterns in antibodies critically determine biological and physical properties but their precise control is a significant challenge in biology and biotechnology. We describe herein the optimization of an endoglycosidase-catalyzed glycosylation of the best-selling biotherapeutic Herceptin, an anti-HER2 antibody. Precise MS analysis of the intact four-chain Ab heteromultimer reveals nonspecific, non-enzymatic reactions (glycation), which are not detected under standard denaturing conditions. This competing reaction, which has hitherto been underestimated as a source of side products, can now be minimized. Optimization allowed access to the purest natural form of Herceptin to date (≥90 %). Moreover, through the use of a small library of sugars containing non-natural functional groups, Ab variants containing defined numbers of selectively addressable chemical tags (reaction handles at Sia C1) in specific positions (for attachment of cargo molecules or "glycorandomization") were readily generated.
Collapse
Affiliation(s)
- Thomas B Parsons
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Weston B Struwe
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Joseph Gault
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Keisuke Yamamoto
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Thomas A Taylor
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Ritu Raj
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Kim Wals
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.,Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Shabaz Mohammed
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Carol V Robinson
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| | - Justin L P Benesch
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Benjamin G Davis
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK.
| |
Collapse
|
265
|
More AS, Toprani VM, Okbazghi SZ, Kim JH, Joshi SB, Middaugh CR, Tolbert TJ, Volkin DB. Correlating the Impact of Well-Defined Oligosaccharide Structures on Physical Stability Profiles of IgG1-Fc Glycoforms. J Pharm Sci 2015; 105:588-601. [PMID: 26869421 DOI: 10.1016/j.xphs.2015.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 09/22/2015] [Indexed: 02/06/2023]
Abstract
As part of a series of articles in this special issue describing 4 well-defined IgG1-Fc glycoforms as a model system for biosimilarity analysis (high mannose-Fc, Man5-Fc, GlcNAc-Fc and N297Q-Fc aglycosylated), the focus of this work is comparisons of their physical properties. A trend of decreasing apparent solubility (thermodynamic activity) by polyethylene glycol precipitation (pH 4.5, 6.0) and lower conformational stability by differential scanning calorimetry (pH 4.5) was observed with reducing size of the N297-linked oligosaccharide structures. Using multiple high-throughput biophysical techniques, the physical stability of the Fc glycoproteins was then measured in 2 formulations (NaCl and sucrose) across a wide range of temperatures (10°C-90°C) and pH (4.0-7.5) conditions. The data sets were used to construct 3-index empirical phase diagrams and radar charts to visualize the regions of protein structural stability. Each glycoform showed improved stability in the sucrose (vs. salt) formulation. The HM-Fc and Man5-Fc displayed the highest relative stability, followed by GlcNAc-Fc, with N297Q-Fc being the least stable. Thus, the overall physical stability profiles of the 4 IgG1-Fc glycoforms also show a correlation with oligosaccharide structure. These data sets are used to develop a mathematical model for biosimilarity analysis (as described in a companion article by Kim et al. in this issue).
Collapse
Affiliation(s)
- Apurva S More
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - Vishal M Toprani
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - Solomon Z Okbazghi
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047
| | - Jae H Kim
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - Thomas J Tolbert
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047
| | - David B Volkin
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047.
| |
Collapse
|
266
|
Zacchi LF, Schulz BL. N-glycoprotein macroheterogeneity: biological implications and proteomic characterization. Glycoconj J 2015; 33:359-76. [DOI: 10.1007/s10719-015-9641-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/04/2015] [Accepted: 11/20/2015] [Indexed: 10/22/2022]
|
267
|
Barnett GV, Qi W, Amin S, Lewis EN, Razinkov VI, Kerwin BA, Liu Y, Roberts CJ. Structural Changes and Aggregation Mechanisms for Anti-Streptavidin IgG1 at Elevated Concentration. J Phys Chem B 2015; 119:15150-63. [PMID: 26563591 DOI: 10.1021/acs.jpcb.5b08748] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Non-native protein aggregation may occur during manufacturing and storage of protein therapeutics, and this may decrease drug efficacy or jeopardize patient safety. From a regulatory perspective, changes in higher order structure due to aggregation are of particular interest but can be difficult to monitor directly at elevated protein concentrations. The present report focuses on non-native aggregation of antistreptavidin (AS) IgG1 at 30 mg/mL under solution conditions that prior work at dilute concentrations (e.g., 1 mg/mL) indicated would result in different aggregation mechanisms. Time-dependent aggregation and structural changes were monitored in situ with dynamic light scattering, small-angle neutron scattering, and Raman scattering and ex situ with far-UV circular dichroism and second-derivative UV spectroscopy. The effects of adding 0.15 M (∼5 w/w %) sucrose were also assessed. The addition of sucrose decreased monomer loss rates but did not change protein-protein interactions, aggregation mechanism(s), or aggregate structure and morphology. Consistent with prior results, altering the pD or salt concentration had the primary effect of changing the aggregation mechanism. Overall, the results provide a comparison of aggregate structure and morphology created via different growth mechanisms using orthogonal techniques and show that the techniques agree at least qualitatively. Interestingly, AS-IgG1 aggregates created at pD 5.3 with no added salt formed the smallest aggregates but had the largest structural changes compared to other solution conditions. The observation that the larger aggregates were also those with less structural perturbation compared to folded AS-IgG1 might be expected to extend to other proteins if the same strong electrostatic repulsions that mediate aggregate growth also mediate structural changes of the constituent proteins within aggregates.
Collapse
Affiliation(s)
- Gregory V Barnett
- Department of Chemical and Biomolecular Engineering, University of Delaware , Newark, Delaware 19716, United States
| | - Wei Qi
- Malvern Biosciences Incorporated, Columbia, Maryland 21046, United States
| | - Samiul Amin
- Malvern Biosciences Incorporated, Columbia, Maryland 21046, United States
| | - E Neil Lewis
- Malvern Biosciences Incorporated, Columbia, Maryland 21046, United States
| | - Vladimir I Razinkov
- Drug Product Development, Amgen Incorporated, Seattle, Washington 98119, United States
| | - Bruce A Kerwin
- Drug Product Development, Amgen Incorporated, Seattle, Washington 98119, United States
| | - Yun Liu
- Department of Chemical and Biomolecular Engineering, University of Delaware , Newark, Delaware 19716, United States.,Center for Neutron Science, National Institutes of Standards and Technology , Gaithersburg, Maryland 20899, United States
| | - Christopher J Roberts
- Department of Chemical and Biomolecular Engineering, University of Delaware , Newark, Delaware 19716, United States
| |
Collapse
|