1
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
2
|
Moatamedi N, Emamzadeh R, Sadeghi HMM, Akbari V. Bioprocess optimization of interferon β-1-a in Pichia pastoris and its improved inhibitory effect against hepatocellular carcinoma cells. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e18984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
3
|
Lundahl MLE, Fogli S, Colavita PE, Scanlan EM. Aggregation of protein therapeutics enhances their immunogenicity: causes and mitigation strategies. RSC Chem Biol 2021; 2:1004-1020. [PMID: 34458822 PMCID: PMC8341748 DOI: 10.1039/d1cb00067e] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Protein aggregation in biotherapeutics has been identified to increase immunogenicity, leading to immune-mediated adverse effects, such as severe allergic responses including anaphylaxis. The induction of anti-drug antibodies (ADAs) moreover enhances drug clearance rates, and can directly block therapeutic function. In this review, identified immune activation mechanisms triggered by protein aggregates are discussed, as well as physicochemical properties of aggregates, such as size and shape, which contribute to immunogenicity. Furthermore, factors which contribute to protein stability and aggregation are considered. Lastly, with these factors in mind, we encourage an innovative and multidisciplinary approach with regard to further research in the field, with the overall aim to avoid immunogenic aggregation in future drug development.
Collapse
Affiliation(s)
- Mimmi L E Lundahl
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin 2 Ireland
| | - Silvia Fogli
- Glycome Biopharma, Unit 4, Joyce House, Barrack Square, Ballincollig Co Cork P31 HW35 Ireland
| | - Paula E Colavita
- School of Chemistry and Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin 2 Ireland
| | - Eoin M Scanlan
- School of Chemistry and Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
4
|
Lee AC, Jeong Y, Lee S, Jang H, Zheng A, Kwon S, Repine JE. Nasopharyngeal Type-I Interferon for Immediately Available Prophylaxis Against Emerging Respiratory Viral Infections. Front Immunol 2021; 12:660298. [PMID: 34093548 PMCID: PMC8170395 DOI: 10.3389/fimmu.2021.660298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022] Open
Abstract
In addition to SARS-CoV-2 and its variants, emerging viruses that cause respiratory viral infections will continue to arise. Increasing evidence suggests a delayed, possibly suppressed, type 1 interferon (IFN-I) response occurs early during COVID-19 and other viral respiratory infections such as SARS and MERS. These observations prompt considering IFN-β as a prophylactic or early intervention for respiratory viral infections. A rationale for developing and testing intranasal interferon beta (IFN-β) as an immediately available intervention for new respiratory viral infections that will arise unexpectedly in the future is presented and supported by basic and clinical trial observations. IFN-β prophylaxis could limit the spread and consequences of an emerging respiratory viral infection in at-risk individuals while specific vaccines are being developed.
Collapse
Affiliation(s)
- Amos C. Lee
- Bio-MAX Institute, Seoul National University, Seoul, South Korea
| | - Yunjin Jeong
- Bio-MAX Institute, Seoul National University, Seoul, South Korea
| | - Sumin Lee
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, South Korea
| | - Haewook Jang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, South Korea
| | - Allen Zheng
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sunghoon Kwon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, South Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, South Korea
- Institute of Entrepreneurial Bio Convergence, Seoul National University, Seoul, South Korea
- Seoul National University Hospital Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Center for Medical Institute, Seoul National University Hospital, Seoul, South Korea
| | - John E. Repine
- Webb-Waring Center, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
5
|
Lee CG, Kim T, Hong S, Chu J, Kang JE, Park HG, Choi JY, Song K, Rha SY, Lee S, Choi JS, Kim SM, Jeong HM, Shin YK. Antibody-Based Targeting of Interferon-Beta-1a Mutein in HER2-Positive Cancer Enhances Antitumor Effects Through Immune Responses and Direct Cell Killing. Front Pharmacol 2021; 11:608774. [PMID: 33505314 PMCID: PMC7832035 DOI: 10.3389/fphar.2020.608774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/24/2020] [Indexed: 11/17/2022] Open
Abstract
Type I interferon (IFN) has been approved as an anticancer agent to treat some malignancies. However, IFNs have a short in vivo half-life, systemic toxicity, and poor biophysical properties, which prevent it from being widely used for cancer therapy. This study aimed to construct recombinant IFN-β-1a mutein immunocytokines that comprise a human epidermal growth factor receptor 2 (HER2)-targeting antibody and IFN-β muteins with an additional glycosylation, which can overcome the limitation of the cytokine itself. Hence, the molecular design aims to 1) enhance productivity and biophysical properties by adding secondary glycosylation in IFN-β, 2) increase the therapeutic index of IFN-β therapy by preferential retention at the tumor by possessing high affinity for HER2-expressing cancer cells, and 3) improve the pharmacokinetics and, thus, the convenience of IFN-β administration. The yield of trastuzumab-IFN-β mutein was higher than that of trastuzumab-wild-type IFN-β in the mammalian cell culture system. Trastuzumab-IFN-β mutein showed similar IFN activity and HER2-targeting ability equivalent to that of IFN-β mutein and trastuzumab, respectively. Trastuzumab-IFN-β mutein directly inhibited the growth of HER2-positive gastric cancer cell lines and was more effective than trastuzumab or IFN-β mutein alone. Trastuzumab-IFN-β mutein and IFN-β mutein displayed enhanced immune cell-mediated cytotoxicity. Collectively, trastuzumab-IFN-β mutein may have indirect immune cell-mediated antitumor effects and direct cell growth inhibitory effects. Tumor-targeting effect of trastuzumab-IFN-β mutein was analyzed using in vivo fluorescence imaging. The accumulation of trastuzumab-IFN-β mutein was observed in HER2-positive tumors rather than other tissues except the liver. To evaluate the both direct tumor growth inhibition effect and indirect immune cell-mediated antitumor effect, we tested the effect of trastuzumab-IFN-β mutein in HER2-positive cancer xenograft models using nude mice or humanized mice. Trastuzumab-IFN-β mutein could significantly enhance tumor regression when compared with trastuzumab or IFN-β mutein. In addition, an increase in tumor-infiltrating lymphocytes was observed in the trastuzumab-IFN-β mutein-treated group, implying that the tumor-targeting IFN-β may have an enhanced antitumor effect through increased immune response. Therefore, targeting IFN-β with an anti-HER2 monoclonal antibody makes the immunocytokine more potent than either agent alone. These novel findings suggest that trastuzumab-IFN-β mutein merits clinical evaluation as a new candidate of anticancer therapeutics.
Collapse
Affiliation(s)
- Chan Gyu Lee
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea.,Genopharm Inc., Seoul, South Korea
| | - TaeEun Kim
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Sungyoul Hong
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jongwan Chu
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | | | - Hee Geon Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | | | - Kyoung Song
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Soohyeon Lee
- Division of Oncology-Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, South Korea
| | - Sun Min Kim
- Department of Obstetrics and Gynecology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, and Seoul National University College of Medicine, Seoul, South Korea
| | | | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.,Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Bio-MAX/N-Bio, Seoul National University, Seoul, South Korea
| |
Collapse
|
6
|
Ibeanu N, Egbu R, Onyekuru L, Javaheri H, Tee Khaw P, R. Williams G, Brocchini S, Awwad S. Injectables and Depots to Prolong Drug Action of Proteins and Peptides. Pharmaceutics 2020; 12:E999. [PMID: 33096803 PMCID: PMC7589296 DOI: 10.3390/pharmaceutics12100999] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Proteins and peptides have emerged in recent years to treat a wide range of multifaceted diseases such as cancer, diabetes and inflammation. The emergence of polypeptides has yielded advancements in the fields of biopharmaceutical production and formulation. Polypeptides often display poor pharmacokinetics, limited permeability across biological barriers, suboptimal biodistribution, and some proclivity for immunogenicity. Frequent administration of polypeptides is generally required to maintain adequate therapeutic levels, which can limit efficacy and compliance while increasing adverse reactions. Many strategies to increase the duration of action of therapeutic polypeptides have been described with many clinical products having been developed. This review describes approaches to optimise polypeptide delivery organised by the commonly used routes of administration. Future innovations in formulation may hold the key to the continued successful development of proteins and peptides with optimal clinical properties.
Collapse
Affiliation(s)
- Nkiruka Ibeanu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Raphael Egbu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Lesley Onyekuru
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Hoda Javaheri
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Gareth R. Williams
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Steve Brocchini
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Sahar Awwad
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| |
Collapse
|
7
|
Stability and Activity of the Hyperglycosylated Human Interferon-β R27T Variant. Sci Rep 2020; 10:8412. [PMID: 32439982 PMCID: PMC7242330 DOI: 10.1038/s41598-020-65495-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/28/2020] [Indexed: 11/16/2022] Open
Abstract
A hyperglycosylated recombinant human interferon-β (rhIFN-β) R27T mutant was established to improve relapsing-remitting multiple sclerosis (RRMS) in our previous study. We focused on the stability of the R27T mutant throughout its production lifetime, including culture, purification, and storage before formulation prior to clinical use. Herein, we address the stability of this protein during optimized culture and purification processes. Additionally, we employed artificial stress conditions during culture and purification to characterize R27T instability. Although, among total R27T, relative native R27T ratio displayed transiently low even under optimized production process, the ratio was recovered by the end of the overall production process, suggesting that culture and purification processes are optimized. Artificial stress during culture and purification processes resulted in degradation of R27T acidic and basic variants, and mismatched disulfide bonds in no-aggregated forms as well as in the aggregated form. The presence of disulfide bond exchange without aggregation in the unfolded/misfolded state could be a novel finding for rhIFN-β products. The results provide meaningful information for the comprehensive evaluation of the stability of the R27T variant.
Collapse
|
8
|
Shayesteh M, Ghasemi F, Tabandeh F, Yakhchali B, Shakibaie M. Design, construction, and expression of recombinant human interferon beta gene in CHO-s cell line using EBV-based expression system. Res Pharm Sci 2020; 15:144-153. [PMID: 32582354 PMCID: PMC7306247 DOI: 10.4103/1735-5362.283814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/10/2019] [Accepted: 05/04/2020] [Indexed: 11/04/2022] Open
Abstract
Background and purpose Codon optimization has been considered as a powerful strategy to increase the expression level of protein therapeutics in mammalian cells. As an empirical approach to study the effects of the codon usage and GC content on heterologous gene expression in suspension adapted Chinese hamster ovary (CHO-s) cells, we redesigned the recombinant human interferon beta (rhIFN- β) gene based on the codon preference of the CHO cell in a way to increase the GC content in the third position of each codon. Experimental approach The nucleotide sequence of the codon-optimized rhIFN-β was synthesized in parallel with the wild-type and expressed transiently in CHO-s cells using Epstein-Bar virus (EBV)-based expression system. The protein expression of the rhIFN-β by codon-optimized and wild-type genes were quantified using ELISA test. Findings / Results The results indicated a 2.8-fold increase in the expression level of the biologically active form of the rhIFN-β by codon-optimized sequence. Conclusion and implications These results shed light on the capability of codon optimization to create a stable CHO cell for scaling up the production of recombinant therapeutics such as rhIFN-β.
Collapse
Affiliation(s)
- Mohadeseh Shayesteh
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, I.R. Iran
| | - Fahimeh Ghasemi
- Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, I.R. Iran
| | - Fatemeh Tabandeh
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, I.R. Iran
| | - Bagher Yakhchali
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, I.R. Iran
| | - Mehdi Shakibaie
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, I.R. Iran
| |
Collapse
|
9
|
Song K, Moon DB, Kim NY, Shin YK. Glycosylation Heterogeneity of Hyperglycosylated Recombinant Human Interferon-β (rhIFN-β). ACS OMEGA 2020; 5:6619-6627. [PMID: 32258897 PMCID: PMC7114697 DOI: 10.1021/acsomega.9b04385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/11/2020] [Indexed: 05/17/2023]
Abstract
We previously developed a biobetter version of rhIFN-β (R27T) that possesses an additional glycosylation site compared with rhIFN-β 1a. Herein, we characterized N-glycosylation heterogeneity of R27T, which includes both N-glycan site occupancy heterogeneity (macro-heterogeneity) and complexity of carbohydrate moieties (micro-heterogeneity). N-glycan site occupancy manifested as distinct differences in size and isoelectric point. The analysis of complex carbohydrate moieties of R27T involved the common biopharmaceutical glycosylation critical quality attributes such as core fucosylation, antennary composition, sialylation, N-acetyllactosamine extensions, linkages, and overall glycan profiles using weak anion-exchange and hydrophilic interaction high-performance liquid chromatography with 2-aminobenzoic acid-labeled N-glycans. The double-glycosylated form accounted for approx. 94% R27T, while the single-glycosylated form accounted for 6% R27T. N-glycans consisted of a mixture of bi-, tri-, and tetra-antennary glycans, some with N-acetyllactosamine extensions, but neither outer arm fucose nor α-galactose was detected. Sialic acid major variants, N-acetyl- and N-glycolyl-neuraminic acid, were more abundant in R27T than in Rebif. The major N-glycan, accounting for ∼42% of total N-glycans, had a di-sialylated, core-fucosylated bi-antennary structure.
Collapse
Affiliation(s)
- Kyoung Song
- LOGONE
Bio Convergence Research Foundation, Center
for Companion Diagnostics, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
- . Phone: +82-2-880-9187
| | - Dae Bong Moon
- IFEZ
Bio Analysis Center, Yeonsu-Gu, Incheon 21985, Republic of Korea
| | - Na Young Kim
- ABION
Inc., R&D Center, Guro, Seoul 13488, Korea
| | - Young Kee Shin
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
- Molecular
Medicine and Biopharmaceutical Sciences, Graduate School of Convergence
Science and Technology, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
- Bio-MAX/N-Bio, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
- . Phone: +82-2-880-9187
| |
Collapse
|
10
|
Balkhi SS, Hojati Z. The Comparison Between the Mutated HuIFN-β 27-101 and the Wild Type Interferon β: the Comprehensive In Silico Study to Evaluate the Effect of Mutations on IFN-β. Adv Pharm Bull 2019; 9:640-648. [PMID: 31857969 PMCID: PMC6912172 DOI: 10.15171/apb.2019.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/02/2019] [Accepted: 07/21/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose: Interferon beta (IFN-β) is used to combat multiple sclerosis (MS) disease. Creating R27T and V101F mutations (mHuIFN-β-27 and mHuIFN-β-101) is one of the tasks performed to improve human interferon beta (HuIFN-β) half-life, function and expression. In this work, the impact of R27T and V101F mutations in recombinant IFN-β on its binding to interferon receptors were studied by molecular docking. Methods: This work was performed through in silico study. The simulation of mutation was performed using the online Rosetta Backrub software and checked using server verify3D. Comparison of access to the solvent of the amino acids in the structures created was performed using the asaview online server. Also, the effect of mutations on the fold of the protein was reviewed by the online HOPE server. The molecular docking was performed between HuIFN-β and the external region of IFNAR receptor using the online ClusPro2 protein-protein docking server. Results: The comparison of the values of the negative binding energy (ΔGbind) obtained from protein-protein molecular docking between IFNAR receptor and HuIFN-β, mHuIFN-β-27, mHuIFN-β-101 and mHuIFN-β-27-101 ligands did not show a significant difference, and these differences do not see any meaningful relationship between them (P > 0.9999). Conclusion: Regarding these results, it can be concluded that these mutations do not have a negative effect on the composition of the complex rHuIFN-β/IFNAR. So, they do not interfere with the binding of the IFN-β to the receptor. It is concluded that the quality of the rHuIFN-β is improved by introducing these two mutations.
Collapse
Affiliation(s)
- Sayed Sharif Balkhi
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, Postal Code: 81746-73441 Isfahan, Iran
| | - Zohreh Hojati
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, Postal Code: 81746-73441 Isfahan, Iran
| |
Collapse
|
11
|
Katla S, Yoganand K, Hingane S, Ranjith Kumar C, Anand B, Sivaprakasam S. Novel glycosylated human interferon alpha 2b expressed in glycoengineered Pichia pastoris and its biological activity: N-linked glycoengineering approach. Enzyme Microb Technol 2019; 128:49-58. [DOI: 10.1016/j.enzmictec.2019.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/17/2019] [Accepted: 05/13/2019] [Indexed: 10/26/2022]
|
12
|
Chung JH, Hong SH, Seo N, Kim TS, An HJ, Lee P, Shin EC, Kim HM. Structure-based glycoengineering of interferon lambda 4 enhances its productivity and anti-viral potency. Cytokine 2019; 125:154833. [PMID: 31479875 PMCID: PMC7129780 DOI: 10.1016/j.cyto.2019.154833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 01/12/2023]
Abstract
Structure-based glycoengineering was applied to improve IFNλ4′s expression level. Model of IFNλ4 signaling complex was used to select de novo N-glycosylation sites. Glycoengineered IFNλ4 variants showed enhanced expression and anti-viral activity.
Interferon lambda 4 (IFNλ4) has been recently known and studied for its role in hepatitis C virus (HCV) infection, but its clinical potential is significantly hampered due to its poor expression in vitro. Our study reports the successful production of IFNλ4 from a mammalian cell line through a glycoengineering and structure-based approach. We introduced de novo N-glycosylation of IFNλ4, guided by structural analysis, and produced IFNλ4 variants in Expi293F that displayed improved expression and potency. To preserve the structure and functionality of IFNλ4, the model structure of the IFNλ4 signaling complex was analyzed and the N-glycosylation candidate sites were selected. The receptor binding activity of engineered IFNλ4 variants and their receptor-mediated signaling pathway were similar to the E. coli version of IFNλ4 (eIFNλ4), while the antiviral activity and induction levels of interferon-stimulated gene (ISG) were all more robust in our variants. Our engineered IFNλ4 variants may be further developed for clinical applications and utilized in basic research to decipher the immunological roles of IFNλ4.
Collapse
Affiliation(s)
- Jae-Hee Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Seon-Hui Hong
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Nari Seo
- Graduate School of Analytical Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Tae-Shin Kim
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Pedro Lee
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Eui-Cheol Shin
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Ho Min Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea.
| |
Collapse
|
13
|
Validation of a cell-based colorimetric reporter gene assay for the evaluation of Type I Interferons. ACTA ACUST UNITED AC 2019; 22:e00331. [PMID: 31061815 PMCID: PMC6487280 DOI: 10.1016/j.btre.2019.e00331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 11/20/2022]
Abstract
The biotherapeutic type I interferons (IFN-I) are indicated to treat several diseases. These products are regulated to guarantee safety and efficacy through critical quality attributes. For this purpose, the development of robust assays is required, followed by its validation to demonstrate their suitability for its intended purpose. Despite there are some commercial kits to evaluate IFN-I signaling, these are focused on measuring in vitro biological response instead of their validation, which is a pharmaceutical industry requirement. The aim of this work was to validate the HEK-Blue IFN-α/β system evaluating the biological activity of IFN-α/β under good laboratory practices, according to international standards. Our results demonstrated that HEK-Blue IFN-α/β system comply with accuracy (r2>0.95) precision (CV < 20%) and specificity for both IFN-α/β; confirming that this assay is robust for this biotherapeutics' evaluation. Thereby, this bioassay could be implemented as a complementary method to the classical anti-proliferative and anti-viral assays under quality control environments.
Collapse
|
14
|
Lee S, Son WS, Yang HB, Rajasekaran N, Kim SS, Hong S, Choi JS, Choi JY, Song K, Shin YK. A Glycoengineered Interferon-β Mutein (R27T) Generates Prolonged Signaling by an Altered Receptor-Binding Kinetics. Front Pharmacol 2019; 9:1568. [PMID: 30733680 PMCID: PMC6353837 DOI: 10.3389/fphar.2018.01568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/24/2018] [Indexed: 12/15/2022] Open
Abstract
The glycoengineering approach is used to improve biophysical properties of protein-based drugs, but its direct impact on binding affinity and kinetic properties for the glycoengineered protein and its binding partner interaction is unclear. Type I interferon (IFN) receptors, composed of IFNAR1 and IFNAR2, have different binding strengths, and sequentially bind to IFN in the dominant direction, leading to activation of signals and induces a variety of biological effects. Here, we evaluated receptor-binding kinetics for each state of binary and ternary complex formation between recombinant human IFN-β-1a and the glycoengineered IFN-β mutein (R27T) using the heterodimeric Fc-fusion technology, and compared biological responses between them. Our results have provided evidence that the additional glycan of R27T, located at the binding interface of IFNAR2, destabilizes the interaction with IFNAR2 via steric hindrance, and simultaneously enhances the interaction with IFNAR1 by restricting the conformational freedom of R27T. Consequentially, altered receptor-binding kinetics of R27T in the ternary complex formation led to a substantial increase in strength and duration of biological responses such as prolonged signal activation and gene expression, contributing to enhanced anti-proliferative activity. In conclusion, our findings reveal N-glycan at residue 25 of R27T is a crucial regulator of receptor-binding kinetics that changes biological activities such as long-lasting activation. Thus, we believe that R27T may be clinically beneficial for patients with multiple sclerosis.
Collapse
Affiliation(s)
- Saehyung Lee
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Woo Sung Son
- Department of Pharmacy, College of Pharmacy, CHA University, Pocheon, South Korea
| | - Ho Bin Yang
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Nirmal Rajasekaran
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Sung-Su Kim
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul, South Korea
| | - Sungyoul Hong
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, South Korea
| | | | - Kyoung Song
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul, South Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, South Korea.,Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Suwon, South Korea
| |
Collapse
|
15
|
Jing X, Hou Y, Hallett W, Sahajwalla CG, Ji P. Key Physicochemical Characteristics Influencing ADME Properties of Therapeutic Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:115-129. [PMID: 31482497 DOI: 10.1007/978-981-13-7709-9_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Therapeutic proteins are a rapidly growing class of drugs in clinical settings. The pharmacokinetics (PK) of therapeutic proteins relies on their absorption, distribution, metabolism, and excretion (ADME) properties. Moreover, the ADME properties of therapeutic proteins are impacted by their physicochemical characteristics. Comprehensive evaluation of these characteristics and their impact on ADME properties are critical to successful drug development. This chapter summarizes all relevant physicochemical characteristics and their effect on ADME properties of therapeutic proteins.
Collapse
Affiliation(s)
- Xing Jing
- U.S. Food and Drug Administration, Office of Clinical Pharmacology, DV II, Silver Spring, MD, USA.
| | - Yan Hou
- U.S. Food and Drug Administration, Office of Clinical Pharmacology, DV II, Silver Spring, MD, USA
| | - William Hallett
- U.S. Food and Drug Administration, Office of Clinical Pharmacology, DV II, Silver Spring, MD, USA
| | - Chandrahas G Sahajwalla
- U.S. Food and Drug Administration, Office of Clinical Pharmacology, DV II, Silver Spring, MD, USA
| | - Ping Ji
- U.S. Food and Drug Administration, Office of Clinical Pharmacology, DV II, Silver Spring, MD, USA
| |
Collapse
|
16
|
Zhou Q, Qiu H. The Mechanistic Impact of N-Glycosylation on Stability, Pharmacokinetics, and Immunogenicity of Therapeutic Proteins. J Pharm Sci 2018; 108:1366-1377. [PMID: 30471292 DOI: 10.1016/j.xphs.2018.11.029] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 01/03/2023]
Abstract
N-glycosylation is one of major post-translational modifications in nature, and it is essential for protein structure and function. As hydrophilic moieties of glycoproteins, N-glycans play important roles in protein stability. They protect the proteins against proteolytic degradation, aggregation, and thermal denaturation through maintaining optimal conformations. There are extensive evidences showing the involvement of N-glycans in the pharmacodynamics and pharmacokinetics of recombinant therapeutic proteins and antibodies. Highly sialylated complex-type glycans enable the longer serum half-lives of proteins against uptake through hepatic asialoglycoprotein receptor and mannose receptor for degradation in lysosomes. Moreover, the presence of nonhuman glycans results in clearance through pre-existing antibodies from serum and induces IgE-mediated anaphylaxis. N-glycans also facilitate or reduce the adverse immune responses of the proteins through interacting with multiple glycan-binding proteins, including those specific for mannose or mannose 6-phosphate. Due to the glycan impacts, a few therapeutic proteins were glycoengineered to improve the pharmacokinetics and stability. Thus, N-glycosylation should be extensively investigated and optimized for each individual protein for better efficacy and safety.
Collapse
Affiliation(s)
- Qun Zhou
- Biologics Research, Sanofi, 49 New York Avenue, Framingham, Massachusetts 01701.
| | - Huawei Qiu
- Biologics Research, Sanofi, 49 New York Avenue, Framingham, Massachusetts 01701
| |
Collapse
|
17
|
Buettner MJ, Shah SR, Saeui CT, Ariss R, Yarema KJ. Improving Immunotherapy Through Glycodesign. Front Immunol 2018; 9:2485. [PMID: 30450094 PMCID: PMC6224361 DOI: 10.3389/fimmu.2018.02485] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/08/2018] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is revolutionizing health care, with the majority of high impact "drugs" approved in the past decade falling into this category of therapy. Despite considerable success, glycosylation-a key design parameter that ensures safety, optimizes biological response, and influences the pharmacokinetic properties of an immunotherapeutic-has slowed the development of this class of drugs in the past and remains challenging at present. This article describes how optimizing glycosylation through a variety of glycoengineering strategies provides enticing opportunities to not only avoid past pitfalls, but also to substantially improve immunotherapies including antibodies and recombinant proteins, and cell-based therapies. We cover design principles important for early stage pre-clinical development and also discuss how various glycoengineering strategies can augment the biomanufacturing process to ensure the overall effectiveness of immunotherapeutics.
Collapse
Affiliation(s)
- Matthew J Buettner
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Christopher T Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Pharmacology/Toxicology Branch I, Division of Clinical Evaluation and Pharmacology/Toxicology, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
18
|
Synthesis and antiplasmodial activity of glyco-conjugate hybrids of phenylhydrazono-indolinones and glycosylated 1,2,3-triazolyl-methyl-indoline-2,3-diones. Eur J Med Chem 2018; 155:764-771. [DOI: 10.1016/j.ejmech.2018.06.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 12/23/2022]
|
19
|
Gugliotta A, Ceaglio N, Etcheverrigaray M, Kratje R, Oggero M. Strategies to Develop Therapeutic N- and O-Hyperglycosylated Proteins. Methods Mol Biol 2018; 1674:163-181. [PMID: 28921436 DOI: 10.1007/978-1-4939-7312-5_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Glycoengineering by N- and/or O-hyperglycosylation represents a procedure to introduce potential sites for adding N- and/or O-glycosyl structures to proteins with the aim of producing biotherapeutics with improved pharmacodynamic and pharmacokinetic properties. In this chapter, a detailed description of the steps routinely performed to generate new proteins having high content of N- and/or O-glycosyl moieties is carried out. The rational strategy involves the initial stage of designing N- and/or O-hyperglycosylated muteins to be expressed by mammalian cells and includes the upstream and downstream processing stages necessary to develop hyperglycosylated versions of the proteins of interest with the purpose of beginning the long road toward producing biobetters.
Collapse
Affiliation(s)
- Agustina Gugliotta
- UNL, CONICET, FBCB, Cell Culture Laboratory, Ciudad Universitaria UNL-C.C. 242, (S3000ZAA), Santa Fe, Pcia. de Santa Fe, Argentina
| | - Natalia Ceaglio
- CONICET, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Marina Etcheverrigaray
- UNL, CONICET, FBCB, Cell Culture Laboratory, Ciudad Universitaria UNL-C.C. 242, (S3000ZAA), Santa Fe, Pcia. de Santa Fe, Argentina
| | - Ricardo Kratje
- UNL, CONICET, FBCB, Cell Culture Laboratory, Ciudad Universitaria UNL-C.C. 242, (S3000ZAA), Santa Fe, Pcia. de Santa Fe, Argentina
| | - Marcos Oggero
- UNL, CONICET, FBCB, Cell Culture Laboratory, Ciudad Universitaria UNL-C.C. 242, (S3000ZAA), Santa Fe, Pcia. de Santa Fe, Argentina.
| |
Collapse
|
20
|
O'Flaherty R, Trbojević-Akmačić I, Greville G, Rudd PM, Lauc G. The sweet spot for biologics: recent advances in characterization of biotherapeutic glycoproteins. Expert Rev Proteomics 2017; 15:13-29. [PMID: 29130774 DOI: 10.1080/14789450.2018.1404907] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Glycosylation is recognized as a Critical Quality Attribute for therapeutic glycoproteins such as monoclonal antibodies, fusion proteins and therapeutic replacement enzymes. Hence, efficient and quantitative glycan analysis techniques have been increasingly important for their discovery, development and quality control. The aim of this review is to highlight relevant and recent advances in analytical technologies for characterization of biotherapeutic glycoproteins. Areas covered: The review gives an overview of the glycosylation trends of biotherapeutics approved in 2016 and 2017 by FDA. It describes current and novel analytical technologies for characterization of therapeutic glycoproteins and is explored in the context of released glycan, glycopeptide or intact glycoprotein analysis. Ultra performance liquid chromatography, mass spectrometry and capillary electrophoresis technologies are explored in this context. Expert commentary: There is a need for the biopharmaceutical industry to incorporate novel state of the art analytical technologies into existing and new therapeutic glycoprotein workflows for safer and more efficient biotherapeutics and for the improvement of future biotherapeutic design. Additionally, at present, there is no 'gold-standard' approach to address all the regulatory requirements and as such this will involve the use of orthogonal glycoanalytical technologies with a view to gain diagnostic information about the therapeutic glycoprotein.
Collapse
Affiliation(s)
- Róisín O'Flaherty
- a NIBRT GlycoScience Group , National Institute for Bioprocessing, Research and Training , Blackrock, Co. Dublin , Ireland
| | | | - Gordon Greville
- a NIBRT GlycoScience Group , National Institute for Bioprocessing, Research and Training , Blackrock, Co. Dublin , Ireland
| | - Pauline M Rudd
- a NIBRT GlycoScience Group , National Institute for Bioprocessing, Research and Training , Blackrock, Co. Dublin , Ireland
| | - Gordan Lauc
- b Genos Glycoscience Research Laboratory , 10000 , Zagreb , Croatia.,c Faculty of Pharmacy and Biochemistry , University of Zagreb , Zagreb , Croatia
| |
Collapse
|
21
|
Kim TE, Hong S, Song K, Park SH, Shin YK. Sensitization of glycoengineered interferon-β1a-resistant cancer cells by cFLIP inhibition for enhanced anti-cancer therapy. Oncotarget 2017; 8:13957-13970. [PMID: 28086218 PMCID: PMC5355153 DOI: 10.18632/oncotarget.14573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/27/2016] [Indexed: 12/30/2022] Open
Abstract
In this study, we examined the molecular mechanism underlying the resistance of cancer cells to R27T, a glycoengineered version of recombinant human interferon (IFN)-β1a, and sought to overcome R27T resistance through combination therapy. R27T has been shown to induce anti-proliferation and apoptosis in human OVCAR-3 and MCF-7 cells, but not in HeLa cells. R27T treatment increased caspase-8 activity and the consequent cleavage of caspase-8 and -3 in R27T-sensitive OVCAR-3 cells, but not in R27T-resistant HeLa cells. Conversely, R27T increased the expression of cellular FLICE-like inhibitory protein (cFLIP) in HeLa cells, but not in OVCAR-3 cells. The sensitization of HeLa cells with cFLIP small interfering RNA or 4,5,6,7-tetrabromobenzotriazole (TBB, an inhibitor of casein kinase-2) facilitated R27T-induced caspase activation, and consequently apoptosis. In OVCAR-3-xenografted mice, intraperitoneal administration of R27T showed 2.1-fold higher anti-tumor efficacy than did the control vehicle. The combined administration of R27T and TBB showed the greatest anti-tumor effect in HeLa tumor-bearing mice, reducing the relative tumor volume by 35.7% compared to that in R27T-treated mice. Taken together, our results suggest that R27T has potential as an anti-cancer drug, and combination therapy with cFLIP inhibitors may be an effective strategy for overcoming R27T resistance.
Collapse
Affiliation(s)
- Tae-Eun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungyoul Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung Song
- Abion Inc., R&D Center, Seoul 08394, Republic of Korea
| | - Sang-Ho Park
- Abion Inc., R&D Center, Seoul 08394, Republic of Korea.,GE Healthcare Korea, R&D Center, Incheon 21988, Republic of Korea
| | - Young Kee Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
22
|
Abstract
INTRODUCTION Many of the biotherapeutics approved or under development suffer from a short half-life necessitating frequent applications in order to maintain a therapeutic concentration over an extended period of time. The implementation of half-life extension strategies allows the generation of long-lasting therapeutics with improved pharmacokinetic and pharmacodynamic properties. AREAS COVERED This review gives an overview of the different half-life extension strategies developed over the past years and their application to generate next-generation biotherapeutics. It focuses on srategies already used in approved drugs and drugs that are in clinical development. These strategies include those aimed at increasing the hydrodynamic radius of the biotherapeutic and strategies which further implement recycling by the neonatal Fc receptor (FcRn). EXPERT OPINION Half-life extension strategies have become an integral part of development for many biotherapeutics. A diverse set of these strategies is available for the fine-tuning of half-life and adaption to the intended treatment modality and disease. Currently, half-life extension is dominated by strategies utilizing albumin binding or fusion, fusion to an immunoglobulin Fc region and PEGylation. However, a variety of alternative strategies, such as fusion of flexible polypeptide chains as PEG mimetic substitute, have reached advanced stages and offer further alternatives for half-life extension.
Collapse
Affiliation(s)
- Roland E Kontermann
- a Institute of Cell Biology and Immunology , University of Stuttgart , Stuttgart , Germany
| |
Collapse
|
23
|
Madhavan A, Sukumaran RK. Secreted expression of an active human interferon‐beta (HuIFNβ) in
Kluyveromyces lactis. Eng Life Sci 2016. [DOI: 10.1002/elsc.201500120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Affiliation(s)
- Aravind Madhavan
- Centre for Biofuels, Biotechnology Division CSIR‐National Institute for Interdisciplinary Science and Technology Trivandrum India
| | - Rajeev Kumar Sukumaran
- Centre for Biofuels, Biotechnology Division CSIR‐National Institute for Interdisciplinary Science and Technology Trivandrum India
| |
Collapse
|
24
|
Interferon Beta: From Molecular Level to Therapeutic Effects. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:343-72. [DOI: 10.1016/bs.ircmb.2016.06.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Kim NA, Song K, Lim DG, Hada S, Shin YK, Shin S, Jeong SH. Basal buffer systems for a newly glycosylated recombinant human interferon-β with biophysical stability and DoE approaches. Eur J Pharm Sci 2015. [DOI: 10.1016/j.ejps.2015.07.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|