1
|
Collins M, Ibeanu N, Grabowska WR, Awwad S, Khaw PT, Brocchini S, Khalili H. Bispecific FpFs: a versatile tool for preclinical antibody development. RSC Chem Biol 2024:d4cb00130c. [PMID: 39347456 PMCID: PMC11427889 DOI: 10.1039/d4cb00130c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024] Open
Abstract
We previously described FpFs 1̲ (Fab-PEG-Fab) as binding mimetics of IgGs. FpFs are prepared with di(bis-sulfone) conjugation reagents 3̲ that undergo disulfide rebridging conjugation with the accessible disulfide of each Fab (Scheme 1). We have now prepared bispecific FpFs 2̲ (bsFpF and Fab1-PEG-Fab2) as potential bispecific antibody mimetics with the intent that bsFpFs could be used in preclinical antibody development since sourcing bispecific antibodies may be challenging during preclinical research. The di(bis-sulfone) reagent 3̲ was first used to prepare a bsFpF 2̲ by the sequential conjugation of a first Fab and then a second Fab to another target (Scheme 2). Seeking to improve bsFpF synthesis, the asymmetric conjugation reagent, bis-sulfone bis-sulfide 1̲6̲, with different thiol conjugation reactivities at each terminus (Scheme 4) was examined and the bsFpFs appeared to be formed at similar conversion to the di(bis-sulfone) reagent 3̲. To explore the advantages of using common intermediates in the preparation of bsFpF families, we investigated bsFpF synthesis with a protein conjugation-ligation approach (Scheme 5). Reagents with a bis-sulfone moiety for conjugation on one PEG terminus and a ligation moiety on the other terminus were examined. Bis-sulfone PEG trans-cyclooctene (TCO) 2̲8̲ and bis-sulfone PEG tetrazine (Tz) 3̲0̲ were used to prepare several bsFpFs targeting various therapeutic targets (TNF-α, IL6R, IL17, and VEGF) and tissue affinity targets (hyaluronic acid and collagen II). Surface plasmon resonance (SPR) binding studies indicated that there was little difference between the dissociation rate constant (k d) for the unmodified Fab, mono-conjugated PEG-Fab and the corresponding Fab in a bsFpF. The Fab association rate (k a) in the bsFpF was slower than for PEG-Fab, which may be because of mass differences that influence SPR results. These observations suggest that each Fab will bind to its target independently of the other Fab and that bsFpF binding profiles can be estimated using the corresponding PEG-Fab conjugates.
Collapse
Affiliation(s)
- Matthew Collins
- School of Health, Sport and Bioscience, University of East London London UK
| | - Nkiru Ibeanu
- School of Pharmacy, University College London London UK
- 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 London UK
- 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
| | - Peng T 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
| | | | - Hanieh Khalili
- School of Pharmacy, University College London London UK
- School of Biomedical Science, University of West London London W5 5RF UK
| |
Collapse
|
2
|
Tao J, Dhanjee HH, Gribble MW, Kottisch V, Rodriguez J, Brown JS, Schmidt H, Juneja J, Denhez F, Lee PS, Lipovšek D, Krystek S, Zhang Y, Bousquet P, Zhang Y, Pentelute BL, Buchwald SL. Site-Specific Antibody Prodrugs via S-Arylation: a Bioconjugation Approach Toward Masked Tyrosine Analogues. J Am Chem Soc 2024; 146:20080-20085. [PMID: 39001844 DOI: 10.1021/jacs.4c04035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
The utility of antibody therapeutics is hampered by potential cross-reactivity with healthy tissue. Over the past decade, significant advances have been made in the design of activatable antibodies, which increase, or create altogether, the therapeutic window of a parent antibody. Of these, antibody prodrugs (pro-antibodies) are masked antibodies that have advanced the most for therapeutic use. They are designed to reveal the active, parent antibody only when encountering proteases upregulated in the microenvironment of the targeted disease tissue, thereby minimizing off-target activity. However, current pro-antibody designs are relegated to fusion proteins that append masking groups restricted to the use of only canonical amino acids, offering excellent control of the site of introduction, but with no authority over where the masking group is installed other than the N-terminus of the antibody. Here, we present a palladium-based bioconjugation approach for the site-specific introduction of a masked tyrosine mimic in the complementary determining region of the FDA approved antibody therapeutic ipilimumab used as a model system. The approach enables the introduction of a protease cleavable group tethered to noncanonical polymers (polyethylene glycol (PEG)) resulting in 47-fold weaker binding to cells expressing CTLA-4, the target antigen of ipilimumab. Upon exposure to tumor-associated proteases, the masking group is cleaved, unveiling a tyrosine-mimic (dubbed hydroxyphenyl cysteine (HPC)) that restores (>90% restoration) binding affinity to its target antigen.
Collapse
Affiliation(s)
- Jason Tao
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Heemal H Dhanjee
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Michael W Gribble
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Veronika Kottisch
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jacob Rodriguez
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Joseph S Brown
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Holly Schmidt
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Juhi Juneja
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Fabienne Denhez
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Peter S Lee
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Daša Lipovšek
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Stanley Krystek
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Yihong Zhang
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Patrick Bousquet
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Yong Zhang
- Research and Early Development, Bristol Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08544, United States
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, United States
| | - Stephen L Buchwald
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Zhang J, Bodenko V, Larkina M, Bezverkhniaia E, Xu T, Liao Y, Abouzayed A, Plotnikov E, Tretyakova M, Yuldasheva F, Belousov MV, Orlova A, Tolmachev V, Gräslund T, Vorobyeva A. Half-life extension via ABD-fusion leads to higher tumor uptake of an affibody-drug conjugate compared to PAS- and XTENylation. J Control Release 2024; 370:468-478. [PMID: 38697314 DOI: 10.1016/j.jconrel.2024.04.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
A critical parameter during the development of protein therapeutics is to endow them with suitable pharmacokinetic and pharmacodynamic properties. Small protein drugs are quickly eliminated by kidney filtration, and in vivo half-life extension is therefore often desired. Here, different half-life extension technologies were studied where PAS polypeptides (PAS300, PAS600), XTEN polypeptides (XTEN288, XTEN576), and an albumin binding domain (ABD) were compared for half-life extension of an anti-human epidermal growth factor receptor 2 (HER2) affibody-drug conjugate. The results showed that extension with the PAS or XTEN polypeptides or the addition of the ABD lowered the affinity for HER2 to some extent but did not negatively affect the cytotoxic potential. The half-lives in mice ranged from 7.3 h for the construct including PAS300 to 11.6 h for the construct including PAS600. The highest absolute tumor uptake was found for the construct including the ABD, which was 60 to 160% higher than the PASylated or XTENylated constructs, even though it did not have the longest half-life (9.0 h). A comparison of the tumor-to-normal-organ ratios showed the best overall performance of the ABD-fused construct. In conclusion, PASylation, XTENylation, and the addition of an ABD are viable strategies for half-life extension of affibody-drug conjugates, with the best performance observed for the construct including the ABD.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, Stockholm 114 17, Sweden
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Scientific and Educational Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Ekaterina Bezverkhniaia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Tianqi Xu
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Yunqi Liao
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Evgenii Plotnikov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Maria Tretyakova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Feruza Yuldasheva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Mikhail V Belousov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, Stockholm 114 17, Sweden.
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
4
|
Geng WC, Jiang ZT, Chen SL, Guo DS. Supramolecular interaction in the action of drug delivery systems. Chem Sci 2024; 15:7811-7823. [PMID: 38817563 PMCID: PMC11134347 DOI: 10.1039/d3sc04585d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/27/2024] [Indexed: 06/01/2024] Open
Abstract
Complex diseases and diverse clinical needs necessitate drug delivery systems (DDSs), yet the current performance of DDSs is far from ideal. Supramolecular interactions play a pivotal role in various aspects of drug delivery, encompassing biocompatibility, drug loading, stability, crossing biological barriers, targeting, and controlled release. Nevertheless, despite having some understanding of the role of supramolecular interactions in drug delivery, their incorporation is frequently overlooked in the design and development of DDSs. This perspective provides a brief analysis of the involved supramolecular interactions in the action of drug delivery, with a primary emphasis on the DDSs employed in the clinic, mainly liposomes and polymers, and recognized phenomena in research, such as the protein corona. The supramolecular interactions implicated in various aspects of drug delivery systems, including biocompatibility, drug loading, stability, spatiotemporal distribution, and controlled release, were individually analyzed and discussed. This perspective aims to trigger a comprehensive and systematic consideration of supramolecular interactions in the further development of DDSs. Supramolecular interactions embody the true essence of the interplay between the majority of DDSs and biological systems.
Collapse
Affiliation(s)
- Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Ze-Tao Jiang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Shi-Lin Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| |
Collapse
|
5
|
Zhu W, Tanday N, Lafferty RA, Flatt PR, Irwin N. Novel enzyme-resistant pancreatic polypeptide analogs evoke pancreatic beta-cell rest, enhance islet cell turnover, and inhibit food intake in mice. Biofactors 2024. [PMID: 38635341 DOI: 10.1002/biof.2059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Pancreatic polypeptide (PP) is a postprandial hormone secreted from pancreatic islets that activates neuropeptide Y4 receptors (NPY4Rs). PP is known to induce satiety but effects at the level of the endocrine pancreas are less well characterized. In addition, rapid metabolism of PP by dipeptidyl peptidase-4 (DPP-4) limits the investigation of the effects of the native peptide. Therefore, in the present study, five novel amino acid substituted and/or fatty acid derivatized PP analogs were synthesized, namely [P3]PP, [K13Pal]PP, [P3,K13Pal]PP, [N-Pal]PP, and [N-Pal,P3]PP, and their impact on pancreatic beta-cell function, as well as appetite regulation and glucose homeostasis investigated. All PP analogs displayed increased resistance to DPP-4 degradation. In addition, all peptides inhibited alanine-induced insulin secretion from BRIN-BD11 beta cells. Native PP and related analogs (10-8 and 10-6 M), and especially [P3]PP and [K13Pal]PP, significantly protected against cytokine-induced beta-cell apoptosis and promoted cellular proliferation, with effects dependent on the NPY4R for all peptides barring [N-Pal,P3]PP. In mice, all peptides, except [N-Pal]PP and [N-Pal,P3]PP, evoked a dose-dependent (25, 75, and 200 nmol/kg) suppression of appetite, with native PP and [P3]PP further augmenting glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) induced reductions of food intake. The PP peptides had no obvious detrimental effect on glucose tolerance and they did not noticeably impair the glucose-regulatory actions of GLP-1 or CCK. In conclusion, Pro3 amino acid substitution of PP, either alone or together with mid-chain acylation, creates PP analogs with benefits on beta-cell rest, islet cell turnover, and energy regulation that may be applicable to the treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Wuyun Zhu
- Diabetes Research Centre, Schools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster University, Coleraine, UK
| | - Neil Tanday
- Diabetes Research Centre, Schools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster University, Coleraine, UK
| | - Ryan A Lafferty
- Diabetes Research Centre, Schools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster University, Coleraine, UK
| | - Peter R Flatt
- Diabetes Research Centre, Schools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster University, Coleraine, UK
| | - Nigel Irwin
- Diabetes Research Centre, Schools of Biomedical Sciences and Pharmacy & Pharmaceutical Sciences, Ulster University, Coleraine, UK
| |
Collapse
|
6
|
Lee HJ, Tomasini-Johansson BR, Gupta N, Kwon GS. Fibronectin-targeted FUD and PEGylated FUD peptides for fibrotic diseases. J Control Release 2023; 360:69-81. [PMID: 37315694 PMCID: PMC10527082 DOI: 10.1016/j.jconrel.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) molecules. Fibronectin (FN) is a glycoprotein found in the blood and tissues, a key player in the assembly of ECM through interaction with cellular and extracellular components. Functional Upstream Domain (FUD), a peptide derived from an adhesin protein of bacteria, has a high binding affinity for the N-terminal 70-kDa domain of FN that plays a crucial role in FN polymerization. In this regard, FUD peptide has been characterized as a potent inhibitor of FN matrix assembly, reducing excessive ECM accumulation. Furthermore, PEGylated FUD was developed to prevent rapid elimination of FUD and enhance its systemic exposure in vivo. Herein, we summarize the development of FUD peptide as a potential anti-fibrotic agent and its application in experimental fibrotic diseases. In addition, we discuss how modification of the FUD peptide via PEGylation impacts pharmacokinetic profiles of the FUD peptide and can potentially contribute to anti-fibrosis therapy.
Collapse
Affiliation(s)
- Hye Jin Lee
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Bianca R Tomasini-Johansson
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, WI 53705, USA
| | - Nikesh Gupta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA; Carbone Cancer Center, University of Wisconsin - Madison, 600 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
7
|
Oleinik LA, Madonov PG, Pykhtina MB. Potential of Interferon Lambda as an Inhibitor of SARS-CoV-2. Mol Biol 2023; 57:291-298. [PMID: 37128210 PMCID: PMC10131541 DOI: 10.1134/s0026893323020152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/06/2022] [Accepted: 10/06/2022] [Indexed: 05/03/2023]
Abstract
This study provides an overview of scientific results on the feasibility of using type III interferons against SARS-CoV-2. We have analyzed data obtained from the PubMed electronic database for the period 2020‒2022. The results of our own studies of pharmacological substances based on recombinant IFN-λ1 and its pegylated form are also presented. Completed and ongoing investigations allow us to position IFN-λ as an effective therapeutic agent against SARS-CoV-2.
Collapse
Affiliation(s)
- L. A. Oleinik
- Research Institute of Clinical and Experimental Lymрhology—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - P. G. Madonov
- Research Institute of Clinical and Experimental Lymрhology—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - M. B. Pykhtina
- Research Institute of Clinical and Experimental Lymрhology—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
8
|
Yeung CWS, Periayah MH, Teo JYQ, Goh ETL, Chee PL, Loh WW, Loh XJ, Lakshminarayanan R, Lim JYC. Transforming Polyethylene into Water-Soluble Antifungal Polymers. Macromolecules 2023. [DOI: 10.1021/acs.macromol.2c01944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Celine W. S. Yeung
- Agency for Science, Technology and Research (A*STAR), Institute of Materials Research and Engineering (IMRE), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Mercy Halleluyah Periayah
- Singapore Eye Research Institute (SERI), The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| | - Jerald Y. Q. Teo
- Agency for Science, Technology and Research (A*STAR), Institute of Materials Research and Engineering (IMRE), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Eunice Tze Leng Goh
- Singapore Eye Research Institute (SERI), The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| | - Pei Lin Chee
- Agency for Science, Technology and Research (A*STAR), Institute of Materials Research and Engineering (IMRE), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Wei Wei Loh
- Agency for Science, Technology and Research (A*STAR), Institute of Materials Research and Engineering (IMRE), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Xian Jun Loh
- Agency for Science, Technology and Research (A*STAR), Institute of Materials Research and Engineering (IMRE), 2 Fusionopolis Way, Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Rajamani Lakshminarayanan
- Singapore Eye Research Institute (SERI), The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| | - Jason Y. C. Lim
- Agency for Science, Technology and Research (A*STAR), Institute of Materials Research and Engineering (IMRE), 2 Fusionopolis Way, Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
9
|
Goncalves AG, Hartzell EJ, Sullivan MO, Chen W. Recombinant protein polymer-antibody conjugates for applications in nanotechnology and biomedicine. Adv Drug Deliv Rev 2022; 191:114570. [PMID: 36228897 DOI: 10.1016/j.addr.2022.114570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/03/2022] [Accepted: 10/04/2022] [Indexed: 01/24/2023]
Abstract
Currently, there are over 100 antibody-based therapeutics on the market for the treatment of various diseases. The increasing importance of antibody treatment is further highlighted by the recent FDA emergency use authorization of certain antibody therapies for COVID-19 treatment. Protein-based materials have gained momentum for antibody delivery due to their biocompatibility, tunable chemistry, monodispersity, and straightforward synthesis and purification. In this review, we discuss progress in engineering the molecular features of protein-based biomaterials, in particular recombinant protein polymers, for introducing novel functionalities and enhancing the delivery properties of antibodies and related binding protein domains.
Collapse
Affiliation(s)
- Antonio G Goncalves
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States
| | - Emily J Hartzell
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States.
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States.
| |
Collapse
|
10
|
Rosen DB, Kvarnhammar AM, Laufer B, Knappe T, Karlsson JJ, Hong E, Lee YC, Thakar D, Zúñiga LA, Bang K, Sabharwal SS, Uppal K, Olling JD, Kjaergaard K, Kurpiers T, Schnabel M, Reich D, Glock P, Zettler J, Krusch M, Bernhard A, Heinig S, Konjik V, Wegge T, Hehn Y, Killian S, Viet L, Runz J, Faltinger F, Tabrizi M, Abel KL, Breinholt VM, Singel SM, Sprogøe K, Punnonen J. TransCon IL-2 β/γ: a novel long-acting prodrug with sustained release of an IL-2Rβ/γ-selective IL-2 variant with improved pharmacokinetics and potent activation of cytotoxic immune cells for the treatment of cancer. J Immunother Cancer 2022; 10:jitc-2022-004991. [PMID: 35817480 PMCID: PMC9274542 DOI: 10.1136/jitc-2022-004991] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Recombinant interleukin-2 (IL-2, aldesleukin) is an approved cancer immunotherapy but causes severe toxicities including cytokine storm and vascular leak syndrome (VLS). IL-2 promotes antitumor function of IL-2Rβ/γ+ natural killer (NK) cells and CD8+, CD4+ and gamma delta (γδ) T cells. However, IL-2 also potently activates immunosuppressive IL-2Rα+ regulatory T cells (Tregs) and IL-2Rα+ eosinophils and endothelial cells, which may promote VLS. Aldesleukin is rapidly cleared requiring frequent dosing, resulting in high Cmax likely potentiating toxicity. Thus, IL-2 cancer immunotherapy has two critical drawbacks: potent activation of undesired IL-2Rα+ cells and suboptimal pharmacokinetics with high Cmax and short half-life. METHODS TransCon IL-2 β/γ was designed to optimally address these drawbacks. To abolish IL-2Rα binding yet retain strong IL-2Rβ/γ activity, IL-2 β/γ was created by permanently attaching a small methoxy polyethylene glycol (mPEG) moiety in the IL-2Rα binding site. To improve pharmacokinetics, IL-2 β/γ was transiently attached to a 40 kDa mPEG carrier via a TransCon (transient conjugation) linker creating a prodrug, TransCon IL-2 β/γ, with sustained release of IL-2 β/γ. IL-2 β/γ was characterized in binding and primary cell assays while TransCon IL-2 β/γ was studied in tumor-bearing mice and cynomolgus monkeys. RESULTS IL-2 β/γ demonstrated selective and potent human IL-2Rβ/γ binding and activation without IL-2Rα interactions. TransCon IL-2 β/γ showed slow-release pharmacokinetics with a low Cmax and a long (>30 hours) effective half-life for IL-2 β/γ in monkeys. In mouse tumor models, TransCon IL-2 β/γ promoted CD8+ T cell and NK cell activation and antitumor activity. In monkeys, TransCon IL-2 β/γ induced robust activation and expansion of CD8+ T cells, NK cells and γδ T cells, relative to CD4+ T cells, Tregs and eosinophils, with no evidence of cytokine storm or VLS. Similarly, IL-2 β/γ enhanced proliferation and cytotoxicity of primary human CD8+ T cells, NK cells and γδ T cells. SUMMARY TransCon IL-2 β/γ is a novel long-acting prodrug with sustained release of an IL-2Rβ/γ-selective IL-2. It has remarkable and durable pharmacodynamic effects in monkeys and potential for improved clinical efficacy and tolerability compared with aldesleukin. TransCon IL-2 β/γ is currently being evaluated in a Phase 1/2 clinical trial (NCT05081609).
Collapse
Affiliation(s)
| | | | | | | | | | - Enping Hong
- Ascendis Pharma Inc, Redwood City, California, USA
| | - Yu-Chi Lee
- Ascendis Pharma Inc, Redwood City, California, USA
| | - Dhruv Thakar
- Ascendis Pharma Inc, Redwood City, California, USA
| | | | - Kathy Bang
- Ascendis Pharma Inc, Redwood City, California, USA
| | | | - Karan Uppal
- Ascendis Pharma Inc, Redwood City, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Laura Viet
- Ascendis Pharma GmbH, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Michot N, Guyochin A, Cinier M, Savignard C, Kitten O, Pascual MH, Pouzieux S, Ozoux ML, Verdier P, Vicat P, Dumas J. Albumin binding Nanofitins, a new scaffold to extend half-life of biologics - a case study with exenatide peptide. Peptides 2022; 152:170760. [PMID: 35150805 DOI: 10.1016/j.peptides.2022.170760] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 01/01/2023]
Abstract
A new strategy of peptide half-life extension has been evaluated. We investigated libraries of a small and very stable protein scaffold called Nanofitin, capable of high affinity for protein targets. We have identified Nanofitins targeting Human and mouse Serum Albumin, which could significantly improve the pharmacokinetics of an active associated peptide, mobilizing the patient's own albumin without external source. To demonstrate the impact of this approach on half-life extension, a genetic fusion of an Exenatide peptide with an Albumin Binding Nanofitin (ABNF) was performed. Specific activity of Exenatide-ABNF was measured and unaffected by the fusion. In vivo mice results provided convincing data (t½ of 8 min for Exenatide peptide compared to 20 h for Exenatide-ABNF) with sustained pharmacological activity over 3 days. This study constitutes a proof-of-concept of in vivo half-life extension of a biologic using an ABNF. Besides, the absence of cysteine in the Nanofitin scaffold, which is therefore devoid of structuring disulfide bonds, allows manufacturing in microbial cost effective systems.
Collapse
Affiliation(s)
- Nadine Michot
- Sanofi, Biologics Research, Vitry sur Seine, 94430, France
| | | | | | | | | | | | | | | | - Patrick Verdier
- Sanofi, Drug Safety & Animal Research, Alfortville 94430, France
| | - Pascale Vicat
- Sanofi, Drug Safety & Animal Research, Alfortville 94430, France
| | - Jacques Dumas
- Sanofi, Biologics Research, Vitry sur Seine, 94430, France
| |
Collapse
|
12
|
Seifert O, Kontermann RE. GlycoTAIL and FlexiTAIL as Half-Life Extension Modules for Recombinant Antibody Fragments. Molecules 2022; 27:molecules27103272. [PMID: 35630749 PMCID: PMC9143431 DOI: 10.3390/molecules27103272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/02/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Many therapeutic proteins are small in size and are rapidly cleared from circulation. Consequently, half-life extension strategies have emerged to improve pharmacokinetic properties, including fusion or binding to long-lasting serum proteins, chemical modifications with hydrophilic polymers such as PEGylation, or, more recently, fusion to PEG mimetic polypeptides. In the present study, two different PEG mimetic approaches, the GlycoTAIL and the FlexiTAIL, were applied to increase the hydrodynamic radius of antibody fragments of different sizes and valencies, including scFv, diabody, and scFv-EHD2 fusion proteins. The GlycoTAIL and FlexiTAIL sequences of varying lengths are composed of aliphatic and hydrophilic residues, with the GlycoTAIL furthermore comprising N-glycosylation sites. All modified proteins could be produced in a mammalian expression system without reducing stability and antigen binding, and all modified proteins exhibited a prolonged half-life and increased drug disposition in mice. The strongest effects were observed for proteins comprising a FlexiTAIL of 248 residues. Thus, the GlycoTAIL and FlexiTAIL sequences represent a flexible and modular system to improve the pharmacokinetic properties of proteins.
Collapse
Affiliation(s)
- Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany;
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, 70569 Stuttgart, Germany
- Correspondence:
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany;
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, 70569 Stuttgart, Germany
| |
Collapse
|
13
|
Hagemans IM, Wierstra PJ, Steuten K, Molkenboer-Kuenen JDM, van Dalen D, Ter Beest M, van der Schoot JMS, Ilina O, Gotthardt M, Figdor CG, Scheeren FA, Heskamp S, Verdoes M. Multiscale imaging of therapeutic anti-PD-L1 antibody localization using molecularly defined imaging agents. J Nanobiotechnology 2022; 20:64. [PMID: 35109860 PMCID: PMC8811974 DOI: 10.1186/s12951-022-01272-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND While immune checkpoint inhibitors such as anti-PD-L1 antibodies have revolutionized cancer treatment, only subgroups of patients show durable responses. Insight in the relation between clinical response, PD-L1 expression and intratumoral localization of PD-L1 therapeutics could improve patient stratification. Therefore, we present the modular synthesis of multimodal antibody-based imaging tools for multiscale imaging of PD-L1 to study intratumoral distribution of PD-L1 therapeutics. RESULTS To introduce imaging modalities, a peptide containing a near-infrared dye (sulfo-Cy5), a chelator (DTPA), an azide, and a sortase-recognition motif was synthesized. This peptide and a non-fluorescent intermediate were used for site-specific functionalization of c-terminally sortaggable mouse IgG1 (mIgG1) and Fab anti-PD-L1. To increase the half-life of the Fab fragment, a 20 kDa PEG chain was attached via strain-promoted azide-alkyne cycloaddition (SPAAC). Biodistribution and imaging studies were performed with 111In-labeled constructs in 4T1 tumor-bearing mice. Comparing our site-specific antibody-conjugates with randomly conjugated antibodies, we found that antibody clone, isotype and method of DTPA conjugation did not change tumor uptake. Furthermore, addition of sulfo-Cy5 did not affect the biodistribution. PEGylated Fab fragment displayed a significantly longer half-life compared to unPEGylated Fab and demonstrated the highest overall tumor uptake of all constructs. PD-L1 in tumors was clearly visualized by SPECT/CT, as well as whole body fluorescence imaging. Immunohistochemistry staining of tumor sections demonstrated that PD-L1 co-localized with the fluorescent and autoradiographic signal. Intratumoral localization of the imaging agent could be determined with cellular resolution using fluorescent microscopy. CONCLUSIONS A set of molecularly defined multimodal antibody-based PD-L1 imaging agents were synthesized and validated for multiscale monitoring of PD-L1 expression and localization. Our modular approach for site-specific functionalization could easily be adapted to other targets.
Collapse
Affiliation(s)
- Iris M Hagemans
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Peter J Wierstra
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kas Steuten
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Janneke D M Molkenboer-Kuenen
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Duco van Dalen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan M S van der Schoot
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Olga Ilina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
- Institute for Chemical Immunology, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Soleimanizadeh A, Dinter H, Schindowski K. Central Nervous System Delivery of Antibodies and Their Single-Domain Antibodies and Variable Fragment Derivatives with Focus on Intranasal Nose to Brain Administration. Antibodies (Basel) 2021; 10:antib10040047. [PMID: 34939999 PMCID: PMC8699001 DOI: 10.3390/antib10040047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
IgG antibodies are some of the most important biopharmaceutical molecules with a high market volume. In spite of the fact that clinical therapies with antibodies are broadly utilized in oncology, immunology and hematology, their delivery strategies and biodistribution need improvement, their limitations being due to their size and poor ability to penetrate into tissues. In view of their small size, there is a rising interest in derivatives, such as single-domain antibodies and single-chain variable fragments, for clinical diagnostic but also therapeutic applications. Smaller antibody formats combine several benefits for clinical applications and can be manufactured at reduced production costs compared with full-length IgGs. Moreover, such formats have a relevant potential for targeted drug delivery that directs drug cargo to a specific tissue or across the blood–brain barrier. In this review, we give an overview of the challenges for antibody drug delivery in general and focus on intranasal delivery to the central nervous system with antibody formats of different sizes.
Collapse
Affiliation(s)
- Arghavan Soleimanizadeh
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Faculty of Medicine, University of Ulm, 89081 Ulm, Germany
| | - Heiko Dinter
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Department of Pharmacy and Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Correspondence:
| |
Collapse
|
15
|
An engineered IL-2 reprogrammed for anti-tumor therapy using a semi-synthetic organism. Nat Commun 2021; 12:4785. [PMID: 34373459 PMCID: PMC8352909 DOI: 10.1038/s41467-021-24987-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/30/2021] [Indexed: 11/15/2022] Open
Abstract
The implementation of applied engineering principles to create synthetic biological systems promises to revolutionize medicine, but application of fundamentally redesigned organisms has thus far not impacted practical drug development. Here we utilize an engineered microbial organism with a six-letter semi-synthetic DNA code to generate a library of site-specific, click chemistry compatible amino acid substitutions in the human cytokine IL-2. Targeted covalent modification of IL-2 variants with PEG polymers and screening identifies compounds with distinct IL-2 receptor specificities and improved pharmacological properties. One variant, termed THOR-707, selectively engages the IL-2 receptor beta/gamma complex without engagement of the IL-2 receptor alpha. In mice, administration of THOR-707 results in large-scale activation and amplification of CD8+ T cells and NK cells, without Treg expansion characteristic of IL-2. In syngeneic B16-F10 tumor-bearing mice, THOR-707 enhances drug accumulation in the tumor tissue, stimulates tumor-infiltrating CD8+ T and NK cells, and leads to a dose-dependent reduction of tumor growth. These results support further characterization of the immune modulatory, anti-tumor properties of THOR-707 and represent a fundamental advance in the application of synthetic biology to medicine, leveraging engineered semi-synthetic organisms as cellular factories to facilitate discovery and production of differentiated classes of chemically modified biologics. The use of synthetic organisms could provide opportunities for discovery and advanced manufacturing of medical drugs. Here the authors use a semi-synthetic organism with an expanded genetic code to generate site-specific chemical modifications in human IL-2.
Collapse
|
16
|
Tully M, Dimde M, Weise C, Pouyan P, Licha K, Schirner M, Haag R. Polyglycerol for Half-Life Extension of Proteins-Alternative to PEGylation? Biomacromolecules 2021; 22:1406-1416. [PMID: 33792290 DOI: 10.1021/acs.biomac.0c01627] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since several decades, PEGylation is known to be the clinical standard to enhance pharmacokinetics of biotherapeutics. In this study, we introduce polyglycerol (PG) of different lengths and architectures (linear and hyperbranched) as an alternative polymer platform to poly(ethylene glycol) (PEG) for half-life extension (HLE). We designed site-selective N-terminally modified PG-protein conjugates of the therapeutic protein anakinra (IL-1ra, Kineret) and compared them systematically with PEG analogues of similar molecular weights. Linear PG and PEG conjugates showed comparable hydrodynamic sizes and retained their secondary structure, whereas binding affinity to IL-1 receptor 1 decreased with increasing polymer length, yet remained in the low nanomolar range for all conjugates. The terminal half-life of a 40 kDa linear PG-modified anakinra was extended 4-fold compared to the unmodified protein, close to its PEG analogue. Our results demonstrate similar performances of PEG- and PG-anakinra conjugates and therefore highlight the outstanding potential of polyglycerol as a PEG alternative for half-life extension of biotherapeutics.
Collapse
Affiliation(s)
- Michael Tully
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Mathias Dimde
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Christoph Weise
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Paria Pouyan
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Kai Licha
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Michael Schirner
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| | - Rainer Haag
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
17
|
Binder U, Skerra A. PASylated Thymosin α1: A Long-Acting Immunostimulatory Peptide for Applications in Oncology and Virology. Int J Mol Sci 2020; 22:ijms22010124. [PMID: 33374407 PMCID: PMC7795856 DOI: 10.3390/ijms22010124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/29/2022] Open
Abstract
Thymosin α1 (Tα1) is an immunostimulatory peptide for the treatment of hepatitis B virus (HBV) and hepatitis C virus (HCV) infections and used as an immune enhancer, which also offers prospects in the context of COVID-19 infections and cancer. Manufacturing of this N-terminally acetylated 28-residue peptide is demanding, and its short plasma half-life limits in vivo efficacy and requires frequent dosing. Here, we combined the PASylation technology with enzymatic in situ N-acetylation by RimJ to produce a long-acting version of Tα1 in Escherichia coli at high yield. ESI-MS analysis of the purified fusion protein indicated the expected composition without any signs of proteolysis. SEC analysis revealed a 10-fold expanded hydrodynamic volume resulting from the fusion with a conformationally disordered Pro/Ala/Ser (PAS) polypeptide of 600 residues. This size effect led to a plasma half-life in rats extended by more than a factor 8 compared to the original synthetic peptide due to retarded kidney filtration. Our study provides the basis for therapeutic development of a next generation thymosin α1 with prolonged circulation. Generally, the strategy of producing an N-terminally protected PASylated peptide solves three major problems of peptide drugs: (i) instability in the expression host, (ii) rapid degradation by serum exopeptidases, and (iii) low bioactivity because of fast renal clearance.
Collapse
Affiliation(s)
- Uli Binder
- XL-protein GmbH, Lise-Meitner-Str. 30, 85354 Freising, Germany
- Correspondence: (U.B.); (A.S.)
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
- Correspondence: (U.B.); (A.S.)
| |
Collapse
|
18
|
Optimizing the anti-tumor efficacy of protein-drug conjugates by engineering the molecular size and half-life. J Control Release 2020; 327:186-197. [DOI: 10.1016/j.jconrel.2020.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/21/2020] [Accepted: 08/03/2020] [Indexed: 11/19/2022]
|
19
|
Kaur N, Dilawari R, Kaur A, Sahni G, Rishi P. Recombinant expression, purification and PEGylation of Paneth cell peptide (cryptdin-2) with value added attributes against Staphylococcus aureus. Sci Rep 2020; 10:12164. [PMID: 32699335 PMCID: PMC7376037 DOI: 10.1038/s41598-020-69039-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022] Open
Abstract
Cryptdins are disulfide-rich cationic antimicrobial peptides secreted by mouse Paneth cells and are known to exhibit potent antimicrobial activity against various deadly pathogens. Keeping in view the extremely low yield obtained from mouse Paneth cells and high cost of synthetic peptide(s), herein, we have attempted to produce cryptdin-2 in Escherichia coli using recombinant technology. To avoid lethal effects of peptide on the host cells, cryptdin-2 was expressed as a fusion protein with thioredoxin as fusion partner which yielded 40 mg/L protein in the soluble fraction. Subsequently, mature cryptdin-2 was cleaved from the fusion partner and purified by cation exchange chromatography. Since conjugation of poly(ethylene) glycol (PEG) has been known to improve the biological properties of biomolecules, therefore, we further attempted to prepare PEG-conjugated variant of cryptdin-2 using thiol specific PEGylation. Though the antimicrobial activity of PEGylated cryptdin-2 was compromised to some extent, but it was found to have enhanced serum stability for longer duration as compared to its un-modified forms. Also, it was found to exhibit reduced toxicity to the host cells. Further, its synergism with gentamicin suggests that PEGylated cryptdin-2 can be used with conventional antibiotics, thereby indicating its possibility to be used as an adjunct therapy.
Collapse
Affiliation(s)
- Navneet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India.,CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, India
| | - Rahul Dilawari
- CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, India
| | - Amrita Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Girish Sahni
- CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
20
|
Powers NE, Swartzwelter B, Marchetti C, de Graaf DM, Lerchner A, Schlapschy M, Datar R, Binder U, Edwards CK, Skerra A, Dinarello CA. PASylation of IL-1 receptor antagonist (IL-1Ra) retains IL-1 blockade and extends its duration in mouse urate crystal-induced peritonitis. J Biol Chem 2020; 295:868-882. [PMID: 31819009 DOI: 10.1074/jbc.ra119.010340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/11/2019] [Indexed: 01/21/2023] Open
Abstract
Interleukin-1 (IL-1) is a key mediator of inflammation and immunity. Naturally-occurring IL-1 receptor antagonist (IL-1Ra) binds and blocks the IL-1 receptor-1 (IL-1R1), preventing signaling. Anakinra, a recombinant form of IL-1Ra, is used to treat a spectrum of inflammatory diseases. However, anakinra is rapidly cleared from the body and requires daily administration. To create a longer-lasting alternative, PASylated IL-1Ra (PAS-IL-1Ra) has been generated by in-frame fusion of a long, defined-length, N-terminal Pro/Ala/Ser (PAS) random-coil polypeptide with IL-1Ra. Here, we compared the efficacy of two PAS-IL-1Ra molecules, PAS600-IL-1Ra and PAS800-IL-1Ra (carrying 600 and 800 PAS residues, respectively), with that of anakinra in mice. PAS600-IL-1Ra displayed markedly extended blood plasma levels 3 days post-administration, whereas anakinra was undetectable after 24 h. We also studied PAS600-IL-1Ra and PAS800-IL-1Ra for efficacy in monosodium urate (MSU) crystal-induced peritonitis. 5 days post-administration, PAS800-IL-1Ra significantly reduced leukocyte influx and inflammatory markers in MSU-induced peritonitis, whereas equimolar anakinra administered 24 h before MSU challenge was ineffective. The 6-h pretreatment with equimolar anakinra or PAS800-IL-1Ra before MSU challenge similarly reduced inflammatory markers. In cultured A549 lung carcinoma cells, anakinra, PAS600-IL-1Ra, and PAS800-IL-Ra reduced IL-1α-induced IL-6 and IL-8 levels with comparable potency. In human peripheral blood mononuclear cells, these molecules suppressed Candida albicans-induced production of the cancer-promoting cytokine IL-22. Surface plasmon resonance analyses revealed significant binding between PAS-IL-1Ra and IL-1R1, although with a slightly lower affinity than anakinra. These results validate PAS-IL-1Ra as an active IL-1 antagonist with marked in vivo potency and a significantly extended half-life compared with anakinra.
Collapse
Affiliation(s)
- Nicholas E Powers
- Department of Medicine, University of Colorado, Aurora, Colorado 80045
| | | | - Carlo Marchetti
- Department of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Dennis M de Graaf
- Department of Medicine, University of Colorado, Aurora, Colorado 80045.,Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | | | - Martin Schlapschy
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Rajiv Datar
- DNX Biopharmaceuticals, Inc., San Diego, California 92121
| | - Uli Binder
- XL-protein GmbH, Lise-Meitner-Strasse 30, 85354 Freising, Germany
| | - Carl K Edwards
- DNX Biopharmaceuticals, Inc., San Diego, California 92121
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, Aurora, Colorado 80045.,Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
21
|
Powers NE, Swartzwelter B, Marchetti C, de Graaf DM, Lerchner A, Schlapschy M, Datar R, Binder U, Edwards CK, Skerra A, Dinarello CA. PASylation of IL-1 receptor antagonist (IL-1Ra) retains IL-1 blockade and extends its duration in mouse urate crystal-induced peritonitis. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49941-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
22
|
Nielsen T, Märcher A, Drobňáková Z, Hučko M, Štengl M, Balšánek V, Wiberg C, Nielsen PF, Nielsen TE, Gothelf KV, Cló E. Disulphide-mediated site-directed modification of proteins. Org Biomol Chem 2020; 18:4717-4722. [DOI: 10.1039/d0ob00861c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Site-directed addition of a single thiols handle to proteins by means of temporary disulphide rebridging of solvent exposed disulphides is obtained with a new labelling reagent.
Collapse
Affiliation(s)
- Thorbjørn Nielsen
- Interdisciplinary Nanoscience Center
- and the Dept. of Chemistry
- Aarhus University
- 8000 Aarhus C
- Denmark
| | - Anders Märcher
- Interdisciplinary Nanoscience Center
- and the Dept. of Chemistry
- Aarhus University
- 8000 Aarhus C
- Denmark
| | | | | | | | | | | | | | | | - Kurt V. Gothelf
- Interdisciplinary Nanoscience Center
- and the Dept. of Chemistry
- Aarhus University
- 8000 Aarhus C
- Denmark
| | | |
Collapse
|
23
|
Belén LH, Rangel-Yagui CDO, Beltrán Lissabet JF, Effer B, Lee-Estevez M, Pessoa A, Castillo RL, Farías JG. From Synthesis to Characterization of Site-Selective PEGylated Proteins. Front Pharmacol 2019; 10:1450. [PMID: 31920645 PMCID: PMC6930235 DOI: 10.3389/fphar.2019.01450] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Covalent attachment of therapeutic proteins to polyethylene glycol (PEG) is widely used for the improvement of its pharmacokinetic and pharmacological properties, as well as the reduction in reactogenicity and related side effects. This technique named PEGylation has been successfully employed in several approved drugs to treat various diseases, even cancer. Some methods have been developed to obtain PEGylated proteins, both in multiple protein sites or in a selected amino acid residue. This review focuses mainly on traditional and novel examples of chemical and enzymatic methods for site-selective PEGylation, emphasizing in N-terminal PEGylation, that make it possible to obtain products with a high degree of homogeneity and preserve bioactivity. In addition, the main assay methods that can be applied for the characterization of PEGylated molecules in complex biological samples are also summarized in this paper.
Collapse
Affiliation(s)
- Lisandra Herrera Belén
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| | - Carlota de Oliveira Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jorge F. Beltrán Lissabet
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| | - Brian Effer
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| | - Manuel Lee-Estevez
- Faculty of Health Sciences, Universidad Autónoma de Chile, Temuco, Chile
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Rodrigo L. Castillo
- Department of Internal Medicine East, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Jorge G. Farías
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
24
|
Syamala PPN, Soberats B, Görl D, Gekle S, Würthner F. Thermodynamic insights into the entropically driven self-assembly of amphiphilic dyes in water. Chem Sci 2019; 10:9358-9366. [PMID: 32110300 PMCID: PMC7017873 DOI: 10.1039/c9sc03103k] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/19/2019] [Indexed: 01/04/2023] Open
Abstract
Self-assembly of amphiphilic dyes and π-systems are more difficult to understand and to control in water compared to organic solvents due to the hydrophobic effect. Herein, we elucidate in detail the self-assembly of a series of archetype bolaamphiphiles bearing a naphthalene bisimide (NBI) π-core with appended oligoethylene glycol (OEG) dendrons of different size. By utilizing temperature-dependent UV-vis spectroscopy and isothermal titration calorimetry (ITC), we have dissected the enthalpic and entropic parameters pertaining to the molecules' self-assembly. All investigated compounds show an enthalpically disfavored aggregation process leading to aggregate growth and eventually precipitation at elevated temperature, which is attributed to the dehydration of oligoethylene glycol units and their concomitant conformational changes. Back-folded conformation of the side chains plays a major role, as revealed by molecular dynamics (MD) and two dimensional NMR (2D NMR) studies, in directing the association. The sterical effect imparted by the jacketing of monomers and dimers also changes the aggregation mechanism from isodesmic to weakly anti-cooperative.
Collapse
Affiliation(s)
- Pradeep P N Syamala
- Universität Würzburg , Institut für Organische Chemie , Am Hubland , 97074 Würzburg , Germany .
- Center for Nanosystems Chemistry & Bavarian Polymer Institute (BPI) , Universität Würzburg , Theodor-Boveri-Weg , 97074 Würzburg , Germany
| | - Bartolome Soberats
- Center for Nanosystems Chemistry & Bavarian Polymer Institute (BPI) , Universität Würzburg , Theodor-Boveri-Weg , 97074 Würzburg , Germany
| | - Daniel Görl
- Center for Nanosystems Chemistry & Bavarian Polymer Institute (BPI) , Universität Würzburg , Theodor-Boveri-Weg , 97074 Würzburg , Germany
| | - Stephan Gekle
- Universität Bayreuth , Biofluid Simulation and Modeling, Theoretische Physik VI & Bavarian Polymer Institute (BPI) , 95440 Bayreuth , Germany .
| | - Frank Würthner
- Universität Würzburg , Institut für Organische Chemie , Am Hubland , 97074 Würzburg , Germany .
- Center for Nanosystems Chemistry & Bavarian Polymer Institute (BPI) , Universität Würzburg , Theodor-Boveri-Weg , 97074 Würzburg , Germany
| |
Collapse
|
25
|
Brandl F, Merten H, Zimmermann M, Béhé M, Zangemeister-Wittke U, Plückthun A. Influence of size and charge of unstructured polypeptides on pharmacokinetics and biodistribution of targeted fusion proteins. J Control Release 2019; 307:379-392. [PMID: 31252038 DOI: 10.1016/j.jconrel.2019.06.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 01/19/2023]
Abstract
Alternative non-IgG binding proteins developed for therapy are small in size and, thus, are rapidly cleared from the circulation by renal filtration. To avoid repeated injection or continuous infusion for the maintenance of therapeutic serum concentrations, extensions of unfolded polypeptides have been developed to prolong serum half-life, but systematic, comparative studies investigating the influence of their size and charge on serum half-life, extravasation, tumor localization and excretion mechanisms have so far been lacking. Here we used a high-affinity Designed Ankyrin Repeat Protein (DARPin) targeting the tumor marker epithelial cell adhesion molecule (EpCAM) in a preclinical tumor xenograft model in mice, and fused it with a series of defined unstructured polypeptides. We used three different sizes of two previously described polypeptides, an uncharged one consisting of only Pro, Ala and Ser (termed PAS) and a charged one consisting of Pro, Ala, Ser, Thr, Gly, Glu (termed XTEN) and performed for the first time a precise comparative localization, distribution and extravasation study. Pharmacokinetic analysis showed a clear linear relationship between hydrodynamic radius and serum half-life across both polypeptides, reaching a half-life of up to 21 h in mice. Tumor uptake was EpCAM-dependent and directly proportional to half-life and size, showing an even tumor penetration for all fusion proteins without unspecific accumulation in non-target tissue. Unexpectedly, charge had no influence on any parameter, neither tumor nor tissue accumulation nor kidney elimination kinetics. Thus, both polypeptide types have a very similar potential for precise half-life modification and tumor targeting.
Collapse
Affiliation(s)
- Fabian Brandl
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland
| | - Hannes Merten
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Martina Zimmermann
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Martin Béhé
- Center of Radiopharmaceutical Sciences, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Uwe Zangemeister-Wittke
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland.
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
26
|
Kaur N, Sinha PK, Sahni G. Site-specific PEGylation of micro-plasmin for improved thrombolytic therapy through engineering enhanced resistance against serpin mediated inhibition. PLoS One 2019; 14:e0217234. [PMID: 31141522 PMCID: PMC6541275 DOI: 10.1371/journal.pone.0217234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/08/2019] [Indexed: 12/03/2022] Open
Abstract
The relatively rapid inhibition of microplasmin by α2-AP leads to short functional half-life of the molecule in vivo, causing inefficient clot dissolution, even after site-specific, local catheter-based delivery. Here, we describe a PEGylation approach for improving the therapeutic potential via improving the survival of microplasmin in presence of its cognate inhibitor, α2-AP, wherein a series of strategically designed cysteine analogs of micro-plasminogen were prepared and expressed in E. coli, and further modified by covalent grafting in vitro with PEG groups of different molecular sizes so as to select single or double PEG chains that increase the molecular weight and hydrodynamic radii of the conjugates, but with a minimal discernible effect on intrinsic plasmin activity and structural framework, as explored by amidolytic activity and CD-spectroscopy, respectively. Interestingly, some of the purified PEG-coupled proteins after conversion to their corresponding proteolytically active forms were found to exhibit significantly reduced inhibition rates (up to 2-fold) by α2-AP relative to that observed with wild-type microplasmin. These results indicate an interesting, and not often observed, effect of PEG groups through reduced/altered dynamics between protease and inhibitor, likely through a steric hindrance mechanism. Thus, the present study successfully identifies single- and double-site PEGylated muteins of microplasmin with significantly enhanced functional half-life through enhanced resistance to inactivation by its in vivo plasma inhibitor. Such an increased survival of bioactivity in situ, holds unmistakable potential for therapeutic exploitation, especially in ischemic strokes where a direct, catheter-based deposition within the cranium has been shown to be promising, but is currently limited by the very short in vivo bioactive half-life of the fibrin dissolving agent/s.
Collapse
Affiliation(s)
- Navneet Kaur
- CSIR-Institute of Microbial Technology, Chandigarh, India
- Panjab University, Chandigarh, India
| | - Prakash Kumar Sinha
- CSIR-Institute of Microbial Technology, Chandigarh, India
- Panjab University, Chandigarh, India
| | - Girish Sahni
- CSIR-Institute of Microbial Technology, Chandigarh, India
- * E-mail:
| |
Collapse
|
27
|
Strategies for the production of long-acting therapeutics and efficient drug delivery for cancer treatment. Biomed Pharmacother 2019; 113:108750. [DOI: 10.1016/j.biopha.2019.108750] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 11/21/2022] Open
|
28
|
Single-Domain Antibodies and Their Formatting to Combat Viral Infections. Antibodies (Basel) 2018; 8:antib8010001. [PMID: 31544807 PMCID: PMC6640686 DOI: 10.3390/antib8010001] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Since their discovery in the 1990s, single-domain antibodies (VHHs), also known as Nanobodies®, have changed the landscape of affinity reagents. The outstanding solubility, stability, and specificity of VHHs, as well as their small size, ease of production and formatting flexibility favor VHHs over conventional antibody formats for many applications. The exceptional ease by which it is possible to fuse VHHs with different molecular modules has been particularly explored in the context of viral infections. In this review, we focus on VHH formats that have been developed to combat viruses including influenza viruses, human immunodeficiency virus-1 (HIV-1), and human respiratory syncytial virus (RSV). Such formats may significantly increase the affinity, half-life, breadth of protection of an antiviral VHH and reduce the risk of viral escape. In addition, VHHs can be equipped with effector functions, for example to guide components of the immune system with high precision to sites of viral infection.
Collapse
|
29
|
Khantasup K, Saiviroonporn P, Jarussophon S, Chantima W, Dharakul T. Anti-EpCAM scFv gadolinium chelate: a novel targeted MRI contrast agent for imaging of colorectal cancer. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2018; 31:633-644. [DOI: 10.1007/s10334-018-0687-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/29/2018] [Accepted: 04/25/2018] [Indexed: 11/29/2022]
|
30
|
Fan K, Jiang B, Guan Z, He J, Yang D, Xie N, Nie G, Xie C, Yan X. Fenobody: A Ferritin-Displayed Nanobody with High Apparent Affinity and Half-Life Extension. Anal Chem 2018; 90:5671-5677. [PMID: 29634235 DOI: 10.1021/acs.analchem.7b05217] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nanobodies consist of a single domain variable fragment of a camelid heavy-chain antibody. Nanobodies have potential applications in biomedical fields because of their simple production procedures and low cost. Occasionally, nanobody clones of interest exhibit low affinities for their target antigens, which, together with their short half-life limit bioanalytical or therapeutic applications. Here, we developed a novel platform we named fenobody, in which a nanobody developed against H5N1 virus is displayed on the surface of ferritin in the form of a 24mer. We constructed a fenobody by substituting the fifth helix of ferritin with the nanobody. TEM analysis showed that nanobodies were displayed on the surface of ferritin in the form of 6 × 4 bundles, and that these clustered nanobodies are flexible for antigen binding in spatial structure. Comparing fenobodies with conventional nanobodies currently used revealed that the antigen binding apparent affinity of anti-H5N1 fenobody was dramatically increased (∼360-fold). Crucially, their half-life extension in a murine model was 10-fold longer than anti-H5N1 nanobody. In addition, we found that our fenobodies are highly expressed in Escherichia coli, and are both soluble and thermo-stable nanocages that self-assemble as 24-polymers. In conclusion, our results demonstrate that fenobodies have unique advantages over currently available systems for apparent affinity enhancement and half-life extension of nanobodies. Our fenobody system presents a suitable platform for various large-scale biotechnological processes and should greatly facilitate the application of nanobody technology in these areas.
Collapse
Affiliation(s)
- Kelong Fan
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology , Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101 , China
| | - Bing Jiang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology , Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101 , China.,University of Chinese Academy of Sciences , 19A Yuquan Road , Beijing 100049 , China
| | - Zhe Guan
- State Key Laboratory of Membrane Biology, Laboratory of Molecular Biophysics, School of Life Sciences , Peking University , Beijing 100871 , China
| | - Jiuyang He
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology , Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101 , China.,University of Chinese Academy of Sciences , 19A Yuquan Road , Beijing 100049 , China
| | - Dongling Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology , Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101 , China
| | - Ni Xie
- Institute of Translation Medicine , Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University , Shenzhen , 518035 , China
| | - Guohui Nie
- Institute of Translation Medicine , Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University , Shenzhen , 518035 , China
| | - Can Xie
- State Key Laboratory of Membrane Biology, Laboratory of Molecular Biophysics, School of Life Sciences , Peking University , Beijing 100871 , China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology , Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101 , China.,University of Chinese Academy of Sciences , 19A Yuquan Road , Beijing 100049 , China
| |
Collapse
|
31
|
Nanoparticle-macrophage interactions: A balance between clearance and cell-specific targeting. Bioorg Med Chem 2017; 25:4487-4496. [PMID: 28705434 DOI: 10.1016/j.bmc.2017.06.040] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 06/16/2017] [Accepted: 06/25/2017] [Indexed: 01/06/2023]
Abstract
The surface properties of nanoparticles (NPs) are a major factor that influences how these nanomaterials interact with biological systems. Interactions between NPs and macrophages of the reticuloendothelial system (RES) can reduce the efficacy of NP diagnostics and therapeutics. Traditionally, to limit NP clearance by the RES system, the NP surface is neutralized with molecules like poly(ethylene glycol) (PEG) which are known to resist protein adsorption and RES clearance. Unfortunately, PEG modification is not without drawbacks including difficulties with the synthesis and associations with immune reactions. To overcome some of these obstacles, we neutralized the NP surface by acetylation and compared this modification to PEGylation for RES clearance and tumor-specific targeting. We found that acetylation was comparable to PEGylation in reducing RES clearance. Additionally, we found that dendrimer acetylation did not impact folic acid (FA)-mediated targeting of tumor cells whereas PEG surface modification reduced the targeting ability of the NP. These results clarify the impact of different NP surface modifications on RES clearance and cell-specific targeting and provide insights into the design of more effective NPs.
Collapse
|
32
|
Danial M, Stauffer AN, Wurm FR, Root MJ, Klok HA. Site-Specific Polymer Attachment to HR2 Peptide Fusion Inhibitors against HIV-1 Decreases Binding Association Rates and Dissociation Rates Rather Than Binding Affinity. Bioconjug Chem 2016; 28:701-712. [PMID: 27737540 DOI: 10.1021/acs.bioconjchem.6b00540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A popular strategy for overcoming the limited plasma half-life of peptide heptad repeat 2 (HR2) fusion inhibitors against HIV-1 is conjugation with biocompatible polymers such as poly(ethylene glycol) (PEG). However, despite improved resistance to proteolysis and reduced renal elimination, covalent attachment of polymers often causes a loss in therapeutic potency. In this study, we investigated the molecular origins of the loss in potency upon conjugation of linear, midfunctional, and hyperbranched PEG-like polymers to peptides that inhibit HIV-1-host cell membrane fusion. Fluorescence binding assays revealed that polymer conjugation imparted mass transport limitations that manifested as coexistent slower association and dissociation rates from the gp41 target on HIV-1. Furthermore, reduced association kinetics rather than affinity disruption was responsible for the loss in antiviral potency. Finally, the binding assays indicated that the unmodified HR2-derived peptide demonstrated diffusion-limited binding. The observed high potency of the unmodified peptide in HIV-1 inhibition assays was therefore attributed to rapid peptide conformational changes upon binding to the gp41 prehairpin structure. This study emphasizes that the view in which polymer ligation to therapeutic peptides inadvertently leads to loss in potency due to a loss in binding affinity requires scientific verification on a case-by-case basis and that high peptide potency may be due to rapid target-binding events.
Collapse
Affiliation(s)
- Maarten Danial
- Institut des Matériaux and Institut des Sciences et Ingéniere Chimiques, École Polytechnique Fédérale de Lausanne, Laboratoire des Polymères , Bâtiment MXD, Station 12, 1015 Lausanne, Switzerland
| | - Angela N Stauffer
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University , 233 South 10th Street, Philadelphia, Pennsylvania 19107, U.S.A
| | - Frederik R Wurm
- Institut des Matériaux and Institut des Sciences et Ingéniere Chimiques, École Polytechnique Fédérale de Lausanne, Laboratoire des Polymères , Bâtiment MXD, Station 12, 1015 Lausanne, Switzerland
| | - Michael J Root
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University , 233 South 10th Street, Philadelphia, Pennsylvania 19107, U.S.A
| | - Harm-Anton Klok
- Institut des Matériaux and Institut des Sciences et Ingéniere Chimiques, École Polytechnique Fédérale de Lausanne, Laboratoire des Polymères , Bâtiment MXD, Station 12, 1015 Lausanne, Switzerland
| |
Collapse
|
33
|
Biosensor binding data and its applicability to the determination of active concentration. Biophys Rev 2016; 8:347-358. [PMID: 28510014 DOI: 10.1007/s12551-016-0219-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/13/2016] [Indexed: 10/20/2022] Open
Abstract
Protein concentration data are required for understanding protein interactions and are a prerequisite for the determination of affinity and kinetic properties. It is vital for the judgment of protein quality and for monitoring the effect of therapeutic agents. Protein concentration values are typically obtained by comparison to a standard and derived from a standard curve. The use of a protein standard is convenient, but may not give reliable results if samples and standards behave differently. In other cases, a standard preparation may not be available and has to be established and validated. Using surface plasmon resonance (SPR) biosensors, an alternative concentration method is possible. This method is called calibration-free concentration analysis (CFCA); it generates active concentration data directly and without the use of a standard. The active concentration of a protein is defined through its interaction with its binding partner. This concentration can differ from the total protein concentration if some protein fraction is incapable of binding. If a protein has several different binding sites, active concentration data can be established for each binding site using site-specific interaction partners. This review will focus on CFCA analysis. It will reiterate the theory of CFCA and describe how CFCA has been applied in different research segments. The major part of the review will, however, try to set expectations on CFCA and discuss how CFCA can be further developed for absolute and relative concentration measurements.
Collapse
|
34
|
Thieme V, Jolly N, Madsen AN, Bellmann-Sickert K, Schwartz TW, Holst B, Cox HM, Beck-Sickinger AG. High molecular weight PEGylation of human pancreatic polypeptide at position 22 improves stability and reduces food intake in mice. Br J Pharmacol 2016; 173:3208-3221. [PMID: 27545829 DOI: 10.1111/bph.13582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Human pancreatic polypeptide (hPP) is known to suppress appetite and food intake, thereby representing a potential therapeutic approach against obesity and associated metabolic disorders. The aim of this study was to improve hPP stability by covalent PEGylation with diverse molecular weight polyethylene glycols (PEGs) at two positions using promising lead structures while maintaining target activity. EXPERIMENTAL APPROACH Modified peptides were synthesized by combined solid-phase and solution-phase peptide synthesis. Their potency was investigated in constitutively expressing human epithelial cells and isolated human colonic mucosa as well as receptor-transfected artificial cell lines. Human blood plasma and porcine liver homogenates were used to examine the in vitro stability of the analogues. The most promising variants were injected s.c. in C57BL/6JRj mice to monitor fasting-induced food intake and bioavailability. KEY RESULTS In human epithelia and colonic mucosal preparations, activity of the modified hPP peptides depended on the core sequence and latency of the peptides was related to PEG size. Peptides modified with a 22 kDa PEG (PEG22) remained intact in blood plasma and on incubation with liver homogenates for more than 96 h. Finally, hPP2-36 , [K22 (PEG22)]hPP2-36 and [K22 (PEG22),Q34 ]hPP significantly reduced cumulative food intake in mice over 16 h after s.c. administration. CONCLUSIONS AND IMPLICATIONS Modification with PEG22 at position 22 stabilizes hPP significantly while extending its biological activities and could be used in drug development prospectively.
Collapse
Affiliation(s)
- V Thieme
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany
| | - N Jolly
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London, UK
| | - A N Madsen
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - K Bellmann-Sickert
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany
| | - T W Schwartz
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - B Holst
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - H M Cox
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London, UK
| | - A G Beck-Sickinger
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany.
| |
Collapse
|
35
|
Collins J, Kempe K, Wilson P, Blindauer CA, McIntosh MP, Davis TP, Whittaker MR, Haddleton DM. Stability Enhancing N-Terminal PEGylation of Oxytocin Exploiting Different Polymer Architectures and Conjugation Approaches. Biomacromolecules 2016; 17:2755-66. [PMID: 27419537 DOI: 10.1021/acs.biomac.6b00919] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Oxytocin, a cyclic nine amino acid neurohypophyseal hormone therapeutic, is effectively used in the control of postpartum hemorrhaging (PPH) and is on the WHO List of Essential Medicines. However, oxytocin has limited shelf life stability in aqueous solutions, particularly at temperatures in excess of 25 °C and injectable aqueous oxytocin formulations require refrigeration (<8 °C). This is particularly problematic in the hot climates often found in many developing countries where daytime temperatures can exceed 40 °C and where reliable cold-chain storage is not always achievable. The purpose of this study was to develop N-terminal amine targeted PEGylation strategies utilizing both linear PEG and polyPEG "comb" polymers as an effective method for stabilizing solution formulations of this peptide for prolonged storage in the absence of efficient cold-chain storage. The conjugation chemistries investigated herein include irreversible amine targeted conjugation methods utilizing NHS ester and aldehyde reductive amination chemistry. Additionally, one reversible conjugation method using a Schiff base approach was explored to allow for the release of the native peptide, thus, ensuring that biological activity remains unaffected. The reversibility of this approach was investigated for the different polymer architectures, alongside a nonpolymer oxytocin analogue to monitor how pH can tune native peptide release. Elevated temperature degradation studies of the polymer conjugates were evaluated to assess the stability of the PEGylated analogues in comparison to the native peptide in aqueous formulations to mimic storage conditions in developing nations and regions where storage under appropriate conditions is challenging.
Collapse
Affiliation(s)
- Jennifer Collins
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom
| | - Kristian Kempe
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Paul Wilson
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Claudia A Blindauer
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom
| | - Michelle P McIntosh
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Michael R Whittaker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - David M Haddleton
- Department of Chemistry, University of Warwick , CV4 7AL, Coventry, United Kingdom.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| |
Collapse
|
36
|
Ginn C, Choi JW, Brocchini S. Disulfide-bridging PEGylation during refolding for the more efficient production of modified proteins. Biotechnol J 2016; 11:1088-99. [DOI: 10.1002/biot.201600035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/07/2016] [Accepted: 05/17/2016] [Indexed: 01/18/2023]
Affiliation(s)
| | - Ji-won Choi
- PolyTherics Ltd; Babraham Research Campus, Babraham; Cambridge UK
| | | |
Collapse
|
37
|
Vernet E, Popa G, Pozdnyakova I, Rasmussen JE, Grohganz H, Giehm L, Jensen MH, Wang H, Plesner B, Nielsen HM, Jensen KJ, Berthelsen J, Sundström M, van de Weert M. Large-Scale Biophysical Evaluation of Protein PEGylation Effects: In Vitro Properties of 61 Protein Entities. Mol Pharm 2016; 13:1587-98. [DOI: 10.1021/acs.molpharmaceut.6b00049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Erik Vernet
- Novo Nordisk A/S, 2880 Bagsværd, Denmark
- The
Novo Nordisk Foundation Center for Protein Research (NNF CPR), Faculty
of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gina Popa
- The
Novo Nordisk Foundation Center for Protein Research (NNF CPR), Faculty
of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina Pozdnyakova
- The
Novo Nordisk Foundation Center for Protein Research (NNF CPR), Faculty
of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob E. Rasmussen
- Department
of Chemistry, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Holger Grohganz
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Giehm
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Malene H. Jensen
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Huabing Wang
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bitten Plesner
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanne M. Nielsen
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Knud J. Jensen
- Department
of Chemistry, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jens Berthelsen
- Costerton
Biofilm center, Department for Immunology and Microbiology, Faculty
of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Sundström
- The
Novo Nordisk Foundation Center for Protein Research (NNF CPR), Faculty
of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marco van de Weert
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Pegylated Trastuzumab Fragments Acquire an Increased in Vivo Stability but Show a Largely Reduced Affinity for the Target Antigen. Int J Mol Sci 2016; 17:491. [PMID: 27043557 PMCID: PMC4848947 DOI: 10.3390/ijms17040491] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/19/2016] [Accepted: 03/24/2016] [Indexed: 12/20/2022] Open
Abstract
PEGylation of biomolecules is a major approach to increase blood stream half-life, stability and solubility of biotherapeutics and to reduce their immunogenicity, aggregation potential and unspecific interactions with other proteins and tissues. Antibodies have generally long half-lives due to high molecular mass and stability toward proteases, however their size lowers to some extent their potential because of a reduced ability to penetrate tissues, especially those of tumor origin. Fab or otherwise engineered smaller fragments are an alternative but are less stable and are much less well retained in circulation. We have here investigated the effects of various PEGylations on the binding properties and in vivo half-life of Fab fragments derived from the enzymatic splitting of Trastuzumab. We find that PEGylation increases the half-life of the molecules but also strongly affects the ability to recognize the target antigen in a way that is dependent on the extent and position of the chemical modification. Data thus support the concept that polyethylene glycol (PEG) conjugation on Trastuzumab Fabs increases half-life but reduces their affinity and this is a fine balance, which must be carefully considered for the design of strategies based on the use of antibody fragments.
Collapse
|
39
|
Alric C, Aubrey N, Allard-Vannier É, di Tommaso A, Blondy T, Dimier-Poisson I, Chourpa I, Hervé-Aubert K. Covalent conjugation of cysteine-engineered scFv to PEGylated magnetic nanoprobes for immunotargeting of breast cancer cells. RSC Adv 2016. [DOI: 10.1039/c6ra06076e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Orientation- and site-directed covalent conjugation of cysteine-engineered scFv to PEGylated SPIONs allows antigen recognition while preserving colloidal properties of nanoprobes.
Collapse
Affiliation(s)
- Christophe Alric
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Nicolas Aubrey
- Université François Rabelais de Tours
- UMR1282 INRA ‘Infectiologie et Santé Publique’
- F 37000 Tours
- France
| | - Émilie Allard-Vannier
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Anne di Tommaso
- Université François Rabelais de Tours
- UMR1282 INRA ‘Infectiologie et Santé Publique’
- F 37000 Tours
- France
| | - Thibaut Blondy
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Isabelle Dimier-Poisson
- Université François Rabelais de Tours
- UMR1282 INRA ‘Infectiologie et Santé Publique’
- F 37000 Tours
- France
| | - Igor Chourpa
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| | - Katel Hervé-Aubert
- Université François Rabelais de Tours
- EA6295 ‘Nanomédicaments et Nanosondes’
- F 37200 Tours
- France
| |
Collapse
|
40
|
Gille H, Hülsmeyer M, Trentmann S, Matschiner G, Christian HJ, Meyer T, Amirkhosravi A, Audoly LP, Hohlbaum AM, Skerra A. Functional characterization of a VEGF-A-targeting Anticalin, prototype of a novel therapeutic human protein class. Angiogenesis 2015; 19:79-94. [DOI: 10.1007/s10456-015-9490-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 11/01/2015] [Indexed: 10/22/2022]
|
41
|
Plückthun A. Designed ankyrin repeat proteins (DARPins): binding proteins for research, diagnostics, and therapy. Annu Rev Pharmacol Toxicol 2015; 55:489-511. [PMID: 25562645 DOI: 10.1146/annurev-pharmtox-010611-134654] [Citation(s) in RCA: 406] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Designed ankyrin repeat proteins (DARPins) can recognize targets with specificities and affinities that equal or surpass those of antibodies, but because of their robustness and extreme stability, they allow a multitude of more advanced formats and applications. This review highlights recent advances in DARPin design, illustrates their properties, and gives some examples of their use. In research, they have been established as intracellular, real-time sensors of protein conformations and as crystallization chaperones. For future therapies, DARPins have been developed by advanced, structure-based protein engineering to selectively induce apoptosis in tumors by uncoupling surface receptors from their signaling cascades. They have also been used successfully for retargeting viruses. In ongoing clinical trials, DARPins have shown good safety and efficacy in macular degeneration diseases. These developments all ultimately exploit the high stability, solubility, and aggregation resistance of these molecules, permitting a wide range of conjugates and fusions to be produced and purified.
Collapse
Affiliation(s)
- Andreas Plückthun
- Department of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland;
| |
Collapse
|
42
|
Schneeweis LA, Obenauer-Kutner L, Kaur P, Yamniuk AP, Tamura J, Jaffe N, O'Mara BW, Lindsay S, Doyle M, Bryson J. Comparison of Ensemble and Single Molecule Methods for Particle Characterization and Binding Analysis of a PEGylated Single-Domain Antibody. J Pharm Sci 2015; 104:4015-4024. [PMID: 26343417 DOI: 10.1002/jps.24624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/29/2022]
Abstract
Domain antibodies (dAbs) are single immunoglobulin domains that form the smallest functional unit of an antibody. This study investigates the behavior of these small proteins when covalently attached to the polyethylene glycol (PEG) moiety that is necessary for extending the half-life of a dAb. The effect of the 40 kDa PEG on hydrodynamic properties, particle behavior, and receptor binding of the dAb has been compared by both ensemble solution and surface methods [light scattering, isothermal titration calorimetry (ITC), surface Plasmon resonance (SPR)] and single-molecule atomic force microscopy (AFM) methods (topography, recognition imaging, and force microscopy). The large PEG dominates the properties of the dAb-PEG conjugate such as a hydrodynamic radius that corresponds to a globular protein over four times its size and a much reduced association rate. We have used AFM single-molecule studies to determine the mechanism of PEG-dependent reductions in the effectiveness of the dAb observed by SPR kinetic studies. Recognition imaging showed that all of the PEGylated dAb molecules are active, suggesting that some may transiently become inactive if PEG sterically blocks binding. This helps explain the disconnect between the SPR, determined kinetically, and the force microscopy and ITC results that demonstrated that PEG does not change the binding energy.
Collapse
Affiliation(s)
- Lumelle A Schneeweis
- Protein Science and Structure, Bristol-Myers Squibb, Princeton, New Jersey 08543.
| | - Linda Obenauer-Kutner
- Biologic Process and Product Development, Bristol-Myers Squibb, Pennington, New Jersey 08534
| | - Parminder Kaur
- Biodesign Institute, Arizona State University, Tempe, Arizona 85287; Department of Physics, Arizona State University, Tempe, Arizona 85287; Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287
| | - Aaron P Yamniuk
- Protein Science and Structure, Bristol-Myers Squibb, Princeton, New Jersey 08543
| | - James Tamura
- Protein Science and Structure, Bristol-Myers Squibb, Princeton, New Jersey 08543
| | - Neil Jaffe
- Biologic Process and Product Development, Bristol-Myers Squibb, Pennington, New Jersey 08534
| | - Brian W O'Mara
- Biologic Process and Product Development, Bristol-Myers Squibb, Pennington, New Jersey 08534
| | - Stuart Lindsay
- Biodesign Institute, Arizona State University, Tempe, Arizona 85287; Department of Physics, Arizona State University, Tempe, Arizona 85287; Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287
| | - Michael Doyle
- Protein Science and Structure, Bristol-Myers Squibb, Princeton, New Jersey 08543
| | - James Bryson
- Protein Science and Structure, Bristol-Myers Squibb, Princeton, New Jersey 08543
| |
Collapse
|
43
|
Abstract
PEGylation is the covalent conjugation of PEG to therapeutic molecules. Protein PEGylation is a clinically proven approach for extending the circulation half-life and reducing the immunogenicity of protein therapeutics. Most clinically used PEGylated proteins are heterogeneous mixtures of PEG positional isomers conjugated to different residues on the protein main chain. Current research is focused to reduce product heterogeneity and to preserve bioactivity. Recent advances and possible future directions in PEGylation are described in this review. So far protein PEGylation has yielded more than 10 marketed products and in view of the lack of equally successful alternatives to extend the circulation half-life of proteins, PEGylation will still play a major role in drug delivery for many years to come.
Collapse
|
44
|
Tong X, Lee S, Bararpour L, Yang F. Long-Term Controlled Protein Release from Poly(Ethylene Glycol) Hydrogels by Modulating Mesh Size and Degradation. Macromol Biosci 2015; 15:1679-86. [PMID: 26259711 DOI: 10.1002/mabi.201500245] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/21/2015] [Indexed: 01/09/2023]
Abstract
Poly(ethylene glycol) (PEG)-based hydrogels are popular biomaterials for protein delivery to guide desirable cellular fates and tissue repair. However, long-term protein release from PEG-based hydrogels remains challenging. Here, we report a PEG-based hydrogel platform for long term protein release, which allows efficient loading of proteins via physical entrapment. Tuning hydrogel degradation led to increase in hydrogel mesh size and gradual release of protein over 60 days of with retained bioactivity. Importantly, this platform does not require the chemical modification of loaded proteins, and may serve as a versatile tool for long-term delivery of a wide range of proteins for drug-delivery and tissue-engineering applications.
Collapse
Affiliation(s)
- Xinming Tong
- Department of Orthopaedic Surgery, Stanford University, CA, 94305, USA
| | - Soah Lee
- Department of Materials Science and Engineering, Stanford University, CA, 94305, USA
| | - Layla Bararpour
- Department of Bioengineering, Stanford University, CA, 94305, USA
| | - Fan Yang
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, CA, 94305, USA.
| |
Collapse
|
45
|
Nanaware-Kharade N, Thakkar S, Gonzalez GA, Peterson EC. A Nanotechnology-Based Platform for Extending the Pharmacokinetic and Binding Properties of Anti-methamphetamine Antibody Fragments. Sci Rep 2015; 5:12060. [PMID: 26159352 PMCID: PMC4498229 DOI: 10.1038/srep12060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 06/08/2015] [Indexed: 12/02/2022] Open
Abstract
To address the need for effective medications to aid in the treatment of methamphetamine (METH) abuse, we used a nanotechnology approach to customize the in vivo behavior of an anti-METH single chain antibody (scFv7F9Cys). Anti-METH scFv7F9Cys was conjugated to dendrimer nanoparticles via a polyethylene glycol (PEG) linker to generate high-order conjugates termed dendribodies. We found that the high affinity (KD = 6.2 nM) and specificity for METH was unchanged after nanoparticle conjugation. The dendribodies were administered in an i.v. bolus to male Sprague Dawley rats after starting a s.c. infusion of METH. The PCKN values for clearance and volume of distribution of scFv7F9Cys after conjugation to dendrimers decreased 45 and 1.6-fold respectively, and the terminal elimination half-life increased 20-fold. Organ distribution of scFv7F9Cys and dendribody in blood and urine agreed well with the PCKN data. Renal clearance appeared to be the major route of elimination for both experimental medications. We have thus successfully developed a novel multivalent METH-binding nanomedicine by conjugating multiple anti-METH scFvs to dendrimer nanoparticles, extending the scFv half-life from 1.3 (±0.3) to 26 (±2.6) hr. These data suggest that the dendribody design could be a feasible platform for generating multivalent antibodies with customizable PCKN profiles.
Collapse
Affiliation(s)
- Nisha Nanaware-Kharade
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham St., # 611, Little Rock, Arkansas 72205, USA
| | - Shraddha Thakkar
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham St., # 750, Little Rock, Arkansas 72205, USA
| | - Guillermo A Gonzalez
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham St., # 611, Little Rock, Arkansas 72205, USA
| | - Eric C Peterson
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham St., # 611, Little Rock, Arkansas 72205, USA
| |
Collapse
|
46
|
Morath V, Bolze F, Schlapschy M, Schneider S, Sedlmayer F, Seyfarth K, Klingenspor M, Skerra A. PASylation of Murine Leptin Leads to Extended Plasma Half-Life and Enhanced in Vivo Efficacy. Mol Pharm 2015; 12:1431-42. [PMID: 25811325 DOI: 10.1021/mp5007147] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Leptin plays a central role in the control of energy homeostasis and appetite and, thus, has attracted attention for therapeutic approaches in spite of its limited pharmacological activity owing to the very short circulation in the body. To improve drug delivery and prolong plasma half-life, we have fused murine leptin with Pro/Ala/Ser (PAS) polypeptides of up to 600 residues, which adopt random coil conformation with expanded hydrodynamic volume in solution and, consequently, retard kidney filtration in a similar manner as polyethylene glycol (PEG). Relative to unmodified leptin, size exclusion chromatography and dynamic light scattering revealed an approximately 21-fold increase in apparent size and a much larger molecular diameter of around 18 nm for PAS(600)-leptin. High receptor-binding activity for all PASylated leptin versions was confirmed in BIAcore measurements and cell-based dual-luciferase assays. Pharmacokinetic studies in mice revealed a much extended plasma half-life after ip injection, from 26 min for the unmodified leptin to 19.6 h for the PAS(600) fusion. In vivo activity was investigated after single ip injection of equimolar doses of each leptin version. Strongly increased and prolonged hypothalamic STAT3 phosphorylation was detected for PAS(600)-leptin. Also, a reduction in daily food intake by up to 60% as well as loss in body weight of >10% lasting for >5 days was observed, whereas unmodified leptin was merely effective for 1 day. Notably, application of a PASylated superactive mouse leptin antagonist (SMLA) led to the opposite effects. Thus, PASylated leptin not only provides a promising reagent to study its physiological role in vivo but also may offer a superior drug candidate for clinical therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Arne Skerra
- ∥XL-protein GmbH, Lise-Meitner-Strasse 30, 85354 Freising, Germany
| |
Collapse
|
47
|
Podobnik B, Helk B, Smilović V, Škrajnar Š, Fidler K, Jevševar S, Godwin A, Williams P. Conjugation of PolyPEG to interferon alpha extends serum half-life while maintaining low viscosity of the conjugate. Bioconjug Chem 2015; 26:452-9. [PMID: 25629733 DOI: 10.1021/bc500523t] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The covalent attachment of poly(ethylene glycol) (PEG) to therapeutic proteins is a commonly used approach for extending in vivo half-lives. A potential limitation of this PEGylation strategy is the adverse effect of PEG on conjugate viscosity. Interferon-alpha (IFN) was conjugated via its N-terminal amino group by reductive amination to α-aldehyde functional comb-shaped PolyPEG polymers (50 and 70 kDa) and to linear PEG (30 kDa). In vitro potencies of the purified PEGylated IFN conjugates were measured by reporter gene assay using a HEK293P/ISRE-SEAP cell line. IFN levels were measured in rats following intravenous injection. Viscosities of various linear PEG and PolyPEG polymers along with the polymer-IFN conjugates were determined using a rotational rheometer with cone-and-plate geometry. In vitro potencies and half-lives of the PEGylated IFN conjugates were compared with those of the marketed branched PEG-IFN conjugate PEGASYS. Both PolyPEG-IFN conjugates retained a similar potency as that of the marketed comparator, whereas the linear PEG-IFN conjugate potency was greater. All conjugates showed extended half-lives compared to that of naked IFN, with the PolyPEG conjugates exhibiting the longest half-lives and the linear PEG conjugate, the shortest. Viscosity analysis showed that the linear PEG-IFN conjugate was over twice as viscous as both PolyPEG conjugates. Taken together, this work demonstrates the potential of PolyPEG conjugation to therapeutic proteins as a novel tool for optimizing pharmacokinetic profiles in a way that potentially allows administration of high-dose formulations because of lower conjugate viscosity.
Collapse
Affiliation(s)
- B Podobnik
- †Sandoz Biopharmaceuticals, Mengeš, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - B Helk
- ‡Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - V Smilović
- †Sandoz Biopharmaceuticals, Mengeš, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - Š Škrajnar
- †Sandoz Biopharmaceuticals, Mengeš, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - K Fidler
- †Sandoz Biopharmaceuticals, Mengeš, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - S Jevševar
- †Sandoz Biopharmaceuticals, Mengeš, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - A Godwin
- §PolyTherics Ltd, Babraham Research Campus, Babraham, Cambridge CB22 3AT, United Kingdom
| | - P Williams
- ∥PolyTherics Ltd, Unit 4, Vanguard Centre, University of Warwick Science Park, Coventry, CV4 7EZ, United Kingdom
| |
Collapse
|
48
|
|
49
|
Stefan N, Zimmermann M, Simon M, Zangemeister-Wittke U, Plückthun A. Novel prodrug-like fusion toxin with protease-sensitive bioorthogonal PEGylation for tumor targeting. Bioconjug Chem 2014; 25:2144-56. [PMID: 25350699 DOI: 10.1021/bc500468s] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Highly potent biotoxins like Pseudomonas exotoxin A (ETA) are attractive payloads for tumor targeting. However, despite replacement of the natural cell-binding domain of ETA by tumor-selective antibodies or alternative binding proteins like designed ankyrin repeat proteins (DARPins) the therapeutic window of such fusion toxins is still limited by target-independent cellular uptake, resulting in toxicity in normal tissues. Furthermore, the strong immunogenicity of the bacterial toxin precludes repeated administration in most patients. Site-specific modification to convert ETA into a prodrug-like toxin which is reactivated specifically in the tumor, and at the same time has a longer circulation half-life and is less immunogenic, is therefore appealing. To engineer a prodrug-like fusion toxin consisting of the anti-EpCAM DARPin Ec1 and a domain I-deleted variant of ETA (ETA″), we used strain-promoted azide alkyne cycloaddition for bioorthogonal conjugation of linear or branched polyethylene glycol (PEG) polymers at defined positions within the toxin moiety. Reversibility of the shielding was provided by a designed peptide linker containing the cleavage site for the rhinovirus 3C model protease. We identified two distinct sites, one within the catalytic domain and one close to the C-terminal KDEL sequence of Ec1-ETA″, simultaneous PEGylation of which resulted in up to 1000-fold lower cytotoxicity in EpCAM-positive tumor cells. Importantly, the potency of the fusion toxin was fully restored by proteolytic unveiling. Upon systemic administration in mice, PEGylated Ec1-ETA″ was much better tolerated than Ec1-ETA″; it showed a longer circulation half-life and an almost 10-fold increased area under the curve (AUC). Our strategy of engineering prodrug-like fusion toxins by bioorthogonal veiling opens new possibilities for targeting tumors with more specificity and efficacy.
Collapse
Affiliation(s)
- Nikolas Stefan
- Department of Biochemistry, Winterthurerstrasse 190, University of Zurich , CH-8057 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
50
|
Liebner R, Meyer M, Hey T, Winter G, Besheer A. Head to head comparison of the formulation and stability of concentrated solutions of HESylated versus PEGylated anakinra. J Pharm Sci 2014; 104:515-26. [PMID: 25445200 DOI: 10.1002/jps.24253] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/12/2014] [Accepted: 10/17/2014] [Indexed: 12/31/2022]
Abstract
Although PEGylation of biologics is currently the gold standard for half-life extension, the technology has a number of limitations, most importantly the non-biodegradability of PEG and the extremely high viscosity at high concentrations. HESylation is a promising alternative based on coupling to the biodegradable polymer hydroxyethyl starch (HES). In this study, we are comparing HESylation with PEGylation regarding the effect on the protein's physicochemical properties, as well as on formulation at high concentrations, where protein stability and viscosity can be compromised. For this purpose, the model protein anakinra is coupled to HES or PEG by reductive amination. Results show that coupling of HES or PEG had practically no effect on the protein's secondary structure, and that it reduced protein affinity by one order of magnitude, with HESylated anakinra more affine than the PEGylated protein. The viscosity of HESylated anakinra at protein concentrations up to 75 mg/mL was approximately 40% lower than that of PEG-anakinra. Both conjugates increased the apparent melting temperature of anakinra in concentrated solutions. Finally, HESylated anakinra was superior to PEG-anakinra regarding monomer recovery after 8 weeks of storage at 40°C. These results show that HESylating anakinra offers formulation advantages compared with PEGylation, especially for concentrated protein solutions.
Collapse
Affiliation(s)
- Robert Liebner
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximillians-University Munich, Munich, 81377, Germany
| | | | | | | | | |
Collapse
|