1
|
Cini JK, Kenney RT, Dexter S, McAndrew SJ, Eraslan RN, Brody R, Rezac DJ, Boohaker R, Lapi SE, Mohan P. SON-1010: an albumin-binding IL-12 fusion protein that improves cytokine half-life, targets tumors, and enhances therapeutic efficacy. Front Immunol 2024; 15:1493257. [PMID: 39697343 PMCID: PMC11652653 DOI: 10.3389/fimmu.2024.1493257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Background Cytokines have been promising cancer immunotherapeutics for decades, yet only two are licensed to date. Interleukin-12 (IL-12) is a potent regulator of cell-mediated immunity that activates NK cells and interferon-γ (IFNγ) production. It plays a central role in multiple pathways that can enhance cancer cell death and modify the tumor microenvironment (TME). Attempts to dose rIL-12 were initially successful but IFNγ toxicity in Phase 2 complicated further development in the late 1990s. Since then, better dosing strategies have been developed, but none have achieved the level of cancer control seen in preclinical models. We set out to develop a novel strategy to deliver fully functional IL-12 and other biologics to the TME by binding albumin, taking advantage of its ability to be concentrated and retained in the tumor. Methods Single-chain variable fragments (scFv) were identified from a human phage display library that bound human, mouse, and cynomolgus macaque serum albumin, both at physiologic and acidic conditions. These were taken through a series of steps to identify strongly binding molecules that don't interfere with the normal physiology of albumin to bind FcRn, giving it prolonged half-life in serum, along with SPARC/GP60, which allows albumin to target the TME. A final molecule was chosen and a single mutation was made that minimizes the potential for immunogenicity. This fully human albumin-binding (FHAB®) domain was characterized and manufacturing processes were developed to bring the first drug candidate into the clinic. Results Once identified, the murine form of mIL12-FHAB was studied preclinically to understand its mechanism of action and biodistribution. It was found to be much more efficient at blocking tumor growth compared to murine IL-12, while stimulating significant IFNγ production with minimal toxicity. SON-1010, which uses the human IL-12 sequence, passed through all of the characterization and required toxicology and is currently being studied in the clinic. Conclusions We identified and developed a platform technology with prolonged half-life that can target IL-12 and other immune modulators to the TME. Safety and efficacy are being studied using SON-1010 as monotherapy and in combination with checkpoint blockade strategies.
Collapse
Affiliation(s)
- John K. Cini
- Sonnet BioTherapeutics, Inc., Princeton, NJ, United States
| | | | - Susan Dexter
- Sonnet BioTherapeutics, Inc., Princeton, NJ, United States
| | | | | | - Rich Brody
- InfinixBio, Inc., Athens, OH, United States
| | | | | | - Suzanne E. Lapi
- Radiology, Chemistry, and Biomedical Engineering, University of Alabama, Birmingham, AL, United States
| | - Pankaj Mohan
- Sonnet BioTherapeutics, Inc., Princeton, NJ, United States
| |
Collapse
|
2
|
Skiba MA, Sterling SM, Rawson S, Zhang S, Xu H, Jiang H, Nemeth GR, Gilman MSA, Hurley JD, Shen P, Staus DP, Kim J, McMahon C, Lehtinen MK, Rockman HA, Barth P, Wingler LM, Kruse AC. Antibodies expand the scope of angiotensin receptor pharmacology. Nat Chem Biol 2024; 20:1577-1585. [PMID: 38744986 PMCID: PMC11561159 DOI: 10.1038/s41589-024-01620-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
G-protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small-molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue and cellular levels. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays and structural studies, we develop maternally selective heavy-chain-only antibody ('nanobody') antagonists against the angiotensin II type I receptor and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to angiotensin II type I receptor with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.
Collapse
Affiliation(s)
- Meredith A Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sarah M Sterling
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Cryo-EM Facility at MIT.nano, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shuhao Zhang
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Haoran Jiang
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Genevieve R Nemeth
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Morgan S A Gilman
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Joseph D Hurley
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Pengxiang Shen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Dean P Staus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
- Septerna, South San Francisco, CA, USA
| | - Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
| | - Conor McMahon
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Sanofi, Large Molecule Research, Cambridge, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Laura M Wingler
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Whitehead CA, Wines BD, Davies AM, McDonnell JM, Trist HM, Esparon SE, Hogarth PM. Stellabody: A novel hexamer-promoting mutation for improved IgG potency. Immunol Rev 2024; 328:438-455. [PMID: 39364646 DOI: 10.1111/imr.13400] [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] [Indexed: 10/05/2024]
Abstract
Advances in antibody engineering are being directed at the development of next generation immunotherapeutics with improved potency. Hexamerisation of IgG is a normal physiological aspect of IgG biology and recently described mutations that facilitate this process have a substantial impact upon monoclonal antibody behavior resulting in the elicitation of dramatically enhanced complement-dependent cytotoxicity, Fc receptor function, and enhanced antigen binding effects, such as targeted receptor agonism or microbe neutralization. Whereas the discovery of IgG hexamerisation enhancing mutations has largely focused on residues with exposure at the surface of the Fc-Fc and CH2-CH3 interfaces, our unique approach is the engineering of the mostly buried residue H429 in the CH3 domain. Selective substitution at position 429 forms the basis of Stellabody technology, where the choice of amino acid results in distinct hexamerisation outcomes. H429F results in monomeric IgG that hexamerises after target binding, so called "on-target" hexamerisation, while the H429Y mutant forms pH-sensitive hexamers in-solution prior to antigen binding. Moreover, Stellabody technologies are broadly applicable across the family of antibody-based biologic therapeutics, including conventional mAbs, bispecific mAbs, and Ig-like biologics such as Fc-fusions, with applications in diverse diseases.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Bruce D Wines
- Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anna M Davies
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, London, UK
| | - James M McDonnell
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, London, UK
| | - Halina M Trist
- Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Sandra E Esparon
- Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - P Mark Hogarth
- Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Almeida CF, Gully BS, Jones CM, Kedzierski L, Gunasinghe SD, Rice MT, Berry R, Gherardin NA, Nguyen TT, Mok YF, Reijneveld JF, Moody DB, Van Rhijn I, La Gruta NL, Uldrich AP, Rossjohn J, Godfrey DI. Direct recognition of an intact foreign protein by an αβ T cell receptor. Nat Commun 2024; 15:8816. [PMID: 39394178 PMCID: PMC11470135 DOI: 10.1038/s41467-024-51897-3] [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: 12/19/2021] [Accepted: 08/21/2024] [Indexed: 10/13/2024] Open
Abstract
αβ T cell receptors (αβTCRs) co-recognise antigens when bound to Major Histocompatibility Complex (MHC) or MHC class I-like molecules. Additionally, some αβTCRs can bind non-MHC molecules, but how much intact antigen reactivities are achieved remains unknown. Here, we identify an αβ T cell clone that directly recognises the intact foreign protein, R-phycoerythrin (PE), a multimeric (αβ)6γ protein complex. This direct αβTCR-PE interaction occurs in an MHC-independent manner, yet triggers T cell activation and bound PE with an affinity comparable to αβTCR-peptide-MHC interactions. The crystal structure reveals how six αβTCR molecules simultaneously engage the PE hexamer, mediated by the complementarity-determining regions (CDRs) of the αβTCR. Here, the αβTCR mainly binds to two α-helices of the globin fold in the PE α-subunit, which is analogous to the antigen-binding platform of the MHC molecule. Using retrogenic mice expressing this TCR, we show that it supports intrathymic T cell development, maturation, and exit into the periphery as mature CD4/CD8 double negative (DN) T cells with TCR-mediated functional capacity. Accordingly, we show how an αβTCR can recognise an intact foreign protein in an antibody-like manner.
Collapse
MESH Headings
- Animals
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Mice
- Phycoerythrin/metabolism
- Phycoerythrin/chemistry
- Lymphocyte Activation/immunology
- Protein Binding
- Crystallography, X-Ray
- Mice, Inbred C57BL
- Humans
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/metabolism
- Models, Molecular
Collapse
Affiliation(s)
- Catarina F Almeida
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin S Gully
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Claerwen M Jones
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lukasz Kedzierski
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Sachith D Gunasinghe
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- European Molecular Biology Laboratory (EMBL) Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| | - Michael T Rice
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Richard Berry
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Nicholas A Gherardin
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Trang T Nguyen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Yee-Foong Mok
- Melbourne Protein Characterisation Platform, Bio21 Molecular Science and Biotechnology Institute, Melbourne, VIC, Australia
| | - Josephine F Reijneveld
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - D Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Nicole L La Gruta
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Adam P Uldrich
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK.
| | - Dale I Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Zakrzewicz A, Vanderheyden K, Galaly Y, Feldhoff S, Sips M, Brinkhaus M, Tikkanen R. Binding to the neonatal Fc receptor enhances the pathogenicity of anti-desmoglein-3 antibodies in keratinocytes. Front Immunol 2024; 15:1473637. [PMID: 39450168 PMCID: PMC11499148 DOI: 10.3389/fimmu.2024.1473637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
The neonatal Fc receptor (FcRn) is important for numerous cellular processes that involve antibody recycling and trafficking. A major function of FcRn is IgG recycling and half-life prolongation, and FcRn blockade results in a reduction of autoantibodies in IgG-mediated autoimmune diseases. In epithelial cells, FcRn functions in processes different from IgG recycling, such as antibody transcytosis in intestinal cells. In pemphigus vulgaris, an autoimmune disease of the epidermis, IgG autoantibodies directed against desmosomal adhesion proteins, especially desmoglein-3 and -1, cause loss of keratinocyte adhesion. We have previously demonstrated that FcRn blockade with efgartigimod, a human Fc fragment with enhanced FcRn binding, significantly reduces the keratinocyte monolayer fragmentation caused by anti-desmoglein-3 antibodies. This points to a direct function of FcRn in keratinocytes, beyond IgG recycling, but the mechanisms have not yet been elucidated in detail. Here, we show that FcRn binding is required for the full pathogenicity of recombinant anti-desmoglein-3 antibodies in keratinocytes, and that antibodies that exhibit enhanced or reduced FcRn affinity due to targeted substitutions in their Fc region, as well as F(ab')2 fragments not binding to FcRn display different degrees of pathogenicity. Blockade of FcRn by efgartigimod only shows a protective effect on keratinocyte adhesion against antibodies capable of binding to FcRn. Furthermore, antibody-induced degradation of desmoglein-3 in keratinocytes does not depend on FcRn, demonstrating that desmoglein-3 degradation and acantholysis are functionally disconnected processes. Our data suggest that the role of FcRn in autoimmune diseases is likely to be versatile and cell-type dependent, thus stressing the importance of further studies on FcRn function in autoimmune diseases.
Collapse
Affiliation(s)
- Anna Zakrzewicz
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | | | - Yad Galaly
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Simon Feldhoff
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | | | | | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| |
Collapse
|
6
|
Bryniarski MA, Tuhin MTH, Acker TM, Wakefield DL, Sethaputra PG, Cook KD, Soto M, Ponce M, Primack R, Jagarapu A, LaGory EL, Conner KP. Cellular Neonatal Fc Receptor Recycling Efficiencies can Differentiate Target-Independent Clearance Mechanisms of Monoclonal Antibodies. J Pharm Sci 2024; 113:2879-2894. [PMID: 38906252 DOI: 10.1016/j.xphs.2024.06.013] [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: 02/08/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
In vivo clearance mechanisms of therapeutic monoclonal antibodies (mAbs) encompass both target-mediated and target-independent processes. Two distinct determinants of overall mAb clearance largely separate of target-mediated influences are non-specific cellular endocytosis and subsequent pH-dependent mAb recycling mediated by the neonatal Fc receptor (FcRn), where inter-mAb variability in the efficiency of both processes is observed. Here, we implemented a functional cell-based FcRn recycling assay via Madin-Darby canine kidney type II cells stably co-transfected with human FcRn and its light chain β2-microglobulin. Next, a series of pH-dependent internalization studies using a model antibody demonstrated proper function of the human FcRn complex. We then applied our cellular assays to assess the contribution of both FcRn and non-specific interactions in the cellular turnover for a panel of 8 clinically relevant mAbs exhibiting variable human pharmacokinetic behavior. Our results demonstrate that the interplay of non-specific endocytosis rates, pH-dependent non-specific interactions, and engagement with FcRn all contribute to the overall recycling efficiency of therapeutic monoclonal antibodies. The predictive capacity of our assay approach was highlighted by successful identification of all mAbs within our panel possessing clearance in humans greater than 5 mL/day/kg. These results demonstrate that a combination of cell-based in vitro assays can properly resolve individual mechanisms underlying the overall in vivo recycling efficiency and non-target mediated clearance of therapeutic mAbs.
Collapse
Affiliation(s)
- Mark A Bryniarski
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA.
| | - Md Tariqul Haque Tuhin
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Timothy M Acker
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Devin L Wakefield
- Research Biomics, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Panijaya Gemy Sethaputra
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Kevin D Cook
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Marcus Soto
- Pharmacokinetics & Drug Metabolism, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Manuel Ponce
- Pharmacokinetics & Drug Metabolism, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Ronya Primack
- Pharmacokinetics & Drug Metabolism, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Aditya Jagarapu
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Edward L LaGory
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA
| | - Kip P Conner
- Pharmacokinetics and Drug Metabolism, Amgen Research, 750 Gateway Blvd, Suite 100, South San Francisco, CA 94080, USA.
| |
Collapse
|
7
|
Lee J, Amatya R, Kim KE, Park YH, Hong E, Djayanti K, Min KA, Roh GS, Shin MC. Genetically engineered long-acting Esculentin-2CHa(1-30) fusion protein with potential applicability for the treatment of NAFLD. J Control Release 2024; 372:699-712. [PMID: 38925336 DOI: 10.1016/j.jconrel.2024.06.061] [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: 12/23/2023] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Esculentin-2CHa(1-30) (‟ESC") has been reported as a potent anti-diabetic peptide with little toxicity. However, its very short plasma residence time severely limits the therapeutic efficacy. To address this issue, we genetically engineered a fusion protein of tandem trimeric ESC with an albumin binding domain (ABD) and a fusion partner, SUMO (named ‟SUMO-3×ESC-ABD"). The SUMO-3×ESC-ABD, successfully produced from E. coli, showed low cellular and hemolytic toxicity while displaying potent activities for the amelioration of hyperglycemia as well as non-alcoholic fatty liver disease (NAFLD) in vitro. In animal studies, the estimated plasma half-life of SUMO-3×ESC-ABD was markedly longer (427-fold) than that of the ESC peptide. In virtue of the extended plasma residence, the SUMO-3×ESC-ABD could produce significant anti-hyperglycemic effects that lasted for >2 days, while both the ESC or ESC-ABD peptides elicited little effects. Further, twice-weekly treatment for 10 weeks, the SUMO-3×ESC-ABD displayed significant improvement in blood glucose control with a reduction in body weight. Most importantly, a significant improvement in the conditions of NAFLD was observed in the SUMO-3×ESC-ABD-treated mice. Along the systemic effects (by improved glucose tolerance and body weight reduction), direct inhibition of the hepatocyte lipid uptake was suggested as the major mechanism of the anti-NAFLD effects. Overall, this study demonstrated the utility of the long-acting SUMO-3×ESC-ABD as a potent drug candidate for the treatment of NAFLD.
Collapse
Affiliation(s)
- Jaewoong Lee
- Department of Anatomy and Convergence Medical Science, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727, Republic of Korea
| | - Reeju Amatya
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam 52828, Republic of Korea
| | - Kyung Eun Kim
- Department of Anatomy and Convergence Medical Science, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727, Republic of Korea
| | - Young-Hoon Park
- New Drug Development Center, Daegu, Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Eunmi Hong
- New Drug Development Center, Daegu, Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Krismala Djayanti
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 50834, Republic of Korea
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 50834, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727, Republic of Korea.
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam 52828, Republic of Korea.
| |
Collapse
|
8
|
Ge S, Dias ACP, Zhang X. Chimerism of avian IgY-scFv and truncated IgG-Fc: A novel strategy in cross-species antibody generation and enhancement. Immunology 2024; 172:46-60. [PMID: 38247105 DOI: 10.1111/imm.13752] [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: 06/10/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Chicken single-chain fragment variable (IgY-scFv) is a functional fragment and an emerging development in genetically engineered antibodies with a wide range of biomedical applications. However, scFvs have considerably shorter serum half-life due to the absence of antibody Fc region compared with the full-length antibody, and usually requires continuous intravenous administration for efficacy. A promising approach to overcome this limitation is to fuse scFv with immunoglobulin G (IgG) Fc region, for better recognition and mediation by the neonatal Fc receptor (FcRn) in the host. In this study, engineered mammalian ΔFc domains (CH2, CH3, and intact Fc region) were fused with anti-canine parvovirus-like particles avian IgY-scFv to produce chimeric antibodies and expressed in the HEK293 cell expression system. The obtained scFv-CH2, scFv-CH3, and scFv-Fc can bind with antigen specifically and dose-dependently. Surface plasmon resonance investigation confirmed that scFv-CH2, scFv-CH3, and scFv-Fc had different degrees of binding to FcRn, with scFv-Fc showing the highest affinity. scFv-Fc had a significantly longer half-life in mice compared with the unfused scFv. The identified ΔFcs are promising for the development of engineered Fc-based therapeutic antibodies and proteins with longer half-lives. The avian IgY-scFv-mammalian IgG Fc region opens up new avenues for antibody engineering, and it is a novel strategy to enhance the rapid development and screening of functional antibodies in veterinary and human medicine.
Collapse
Affiliation(s)
- Shikun Ge
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
- College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Alberto Carlos Pires Dias
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
| | - Xiaoying Zhang
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
- College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
9
|
Spiteri VA, Doutch J, Rambo RP, Bhatt JS, Gor J, Dalby PA, Perkins SJ. Using atomistic solution scattering modelling to elucidate the role of the Fc glycans in human IgG4. PLoS One 2024; 19:e0300964. [PMID: 38557973 PMCID: PMC10984405 DOI: 10.1371/journal.pone.0300964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Human immunoglobulin G (IgG) exists as four subclasses IgG1-4, each of which has two Fab subunits joined by two hinges to a Fc subunit. IgG4 has the shortest hinge with 12 residues. The Fc subunit has two glycan chains, but the importance of glycosylation is not fully understood in IgG4. Here, to evaluate the stability and structure of non-glycosylated IgG4, we performed a multidisciplinary structural study of glycosylated and deglycosylated human IgG4 A33 for comparison with our similar study of human IgG1 A33. After deglycosylation, IgG4 was found to be monomeric by analytical ultracentrifugation; its sedimentation coefficient of 6.52 S was reduced by 0.27 S in reflection of its lower mass. X-ray and neutron solution scattering showed that the overall Guinier radius of gyration RG and its cross-sectional values after deglycosylation were almost unchanged. In the P(r) distance distribution curves, the two M1 and M2 peaks that monitor the two most common distances within IgG4 were unchanged following deglycosylation. Further insight from Monte Carlo simulations for glycosylated and deglycosylated IgG4 came from 111,382 and 117,135 possible structures respectively. Their comparison to the X-ray and neutron scattering curves identified several hundred best-fit models for both forms of IgG4. Principal component analyses showed that glycosylated and deglycosylated IgG4 exhibited different conformations from each other. Within the constraint of unchanged RG and M1-M2 values, the glycosylated IgG4 models showed more restricted Fc conformations compared to deglycosylated IgG4, but no other changes. Kratky plots supported this interpretation of greater disorder upon deglycosylation, also observed in IgG1. Overall, these more variable Fc conformations may demonstrate a generalisable impact of deglycosylation on Fc structures, but with no large conformational changes in IgG4 unlike those seen in IgG1.
Collapse
Affiliation(s)
- Valentina A. Spiteri
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - James Doutch
- ISIS Facility, STFC Rutherford Appleton Laboratory, Harwell Campus, Didcot, Oxfordshire, United Kingdom
| | - Robert P. Rambo
- Diamond Light Source Ltd., Diamond House, Harwell Science and Innovation Campus, Chilton, Didcot, Oxfordshire, United Kingdom
| | - Jayesh S. Bhatt
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Jayesh Gor
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Paul A. Dalby
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Stephen J. Perkins
- Division of Biosciences, Department of Structural and Molecular Biology, University College London, London, United Kingdom
| |
Collapse
|
10
|
Li D, Ding L, Chen Y, Wang Z, Zeng Z, Ma X, Huang H, Li H, Qian X, Yang Z, Zhu H. Exploration of radionuclide labeling of a novel scFv-Fc fusion protein targeting CLDN18.2 for tumor diagnosis and treatment. Eur J Med Chem 2024; 266:116134. [PMID: 38266552 DOI: 10.1016/j.ejmech.2024.116134] [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: 12/01/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 01/26/2024]
Abstract
PURPOSE Claudin 18.2 (CLDN18.2), due to its highly selective expression in tumor cells, has made breakthrough progress in clinical research and is expected to be integrated into routine tumor diagnosis and treatment. METHODS In this research, we obtained an scFv-Fc fusion protein (SF106) targeting CLDN18.2 through hybridoma technology. The scFv-Fc fusion protein was labeled with radioactive isotopes (124I and 177Lu) to generate the radio-probes. The targeting and specificity of the radio-probes were tested in cellular models, and its diagnostic and therapeutic potential was further evaluated in tumor-bearing models. RESULTS The molecular probes [124I]I-SF106 and [177Lu]Lu-DOTA-SF106 possess high radiochemical purity (RCP, 98.18 ± 0.93 % and 97.05 ± 1.1 %) and exhibit good stability in phosphate buffer saline and 5 % human serum albumin (92.44 ± 4.68 % and 91.03 ± 2.42 % at 120 h). [124I]I-SF106 uptake in cells expressing CLDN18.2 was well targeted and specific, and the dissociation constant was 17.74 nM [124I]I-SF106 micro-PET imaging showed that the maximum standardized uptake value (SUVmax) was significantly higher than CLDN18.2-negative tumors (1.83 ± 0.02 vs. 1.23 ± 0.04, p < 0.001). The maximum uptake was attained in tumors expressing CLDN18.2 at 48 h after injection. [124I]I-SF106 and [177Lu]Lu-DOTA-SF106 dosimetric study showed that the effective dose in humans complies with the medical safety standards required for their clinical application. The results of treatment experiments showed that 3 MBq of [177Lu]Lu-DOTA-SF106 in CLDN18.2-expressing tumor-bearing mice could significantly inhibit tumor growth. CONCLUSION These results indicate that radionuclide-labeled scFv-Fc molecular probes ([124I]I-SF106 and [177Lu]Lu-DOTA-SF106) provide a new possibility for the diagnosis and treatment of CLDN18.2-positive cancer patients in clinical practice.
Collapse
Affiliation(s)
- Dapeng Li
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lei Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yan Chen
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zilei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Ziqing Zeng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaopan Ma
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Haifeng Huang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550025, Guizhou, China
| | - Hongjun Li
- Suzhou Transcenta Therapeutics Co., Ltd, Suzhou, 215000, China
| | - Xueming Qian
- Suzhou Transcenta Therapeutics Co., Ltd, Suzhou, 215000, China.
| | - Zhi Yang
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Hua Zhu
- Medical College, Guizhou University, Guiyang, 550025, Guizhou, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
11
|
Dylewski JF, Haddad G, Blaine J. Exploiting the neonatal Fc receptor to treat kidney disease. Kidney Int 2023; 105:S0085-2538(23)00747-0. [PMID: 39491149 PMCID: PMC11068363 DOI: 10.1016/j.kint.2023.09.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 05/07/2024]
Abstract
The neonatal Fc receptor (FcRn) was initially discovered as the receptor that allowed passive immunity in newborns by transporting maternal IgG through the placenta and enterocytes. Since its initial discovery, FcRn has been found to exist throughout all stages of life and in many different cell types. Beyond passive immunity, FcRn is necessary for intrinsic albumin and IgG recycling and is important for antigen processing and presentation. Given its multiple important roles, FcRn has been utilized in many disease treatments including a new class of agents that were developed to inhibit FcRn for treatment of a variety of autoimmune diseases. Certain cell populations within the kidney also express high levels of this receptor. Specifically, podocytes, proximal tubule epithelial cells, and vascular endothelial cells have been found to utilize FcRn. In this review, we summarize what is known about FcRn and its function within the kidney. We also discuss how FcRn has been used for therapeutic benefit, including how newer FcRn inhibiting agents are being used to treat autoimmune diseases. Lastly, we will discuss what renal diseases may respond to FcRn inhibitors and how further work studying FcRn within the kidney may lead to therapies for kidney diseases.
Collapse
Affiliation(s)
- James F Dylewski
- Division of Nephrology, Denver Health Medical Center, Denver, CO, USA; Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - George Haddad
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Judith Blaine
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
12
|
Bou-Jaoudeh M, Mimoun A, Delignat S, Peyron I, Capdevila L, Daventure V, Deligne C, Dimitrov JD, Christophe OD, Denis CV, Lenting PJ, Proulle V, Lacroix-Desmazes S. Imlifidase, a new option to optimize the management of patients with hemophilia A on emicizumab. J Thromb Haemost 2023; 21:2776-2783. [PMID: 37473843 DOI: 10.1016/j.jtha.2023.06.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/07/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Emicizumab is a bispecific, chimeric, humanized immunoglobulin G (IgG)4 that mimics the procoagulant activity of factor (F) VIII (FVIII). Its long half-life and subcutaneous route of administration have been life-changing in treating patients with hemophilia A (HA) with or without FVIII inhibitors. However, emicizumab only partially mimics FVIII activity; it prevents but does not treat acute bleeds. Emergency management is particularly complicated in patients with FVIII inhibitors receiving emicizumab prophylaxis in whom exogenous FVIII is inefficient. We have shown recently that Imlifidase (IdeS), a bacterial IgG-degrading enzyme, efficiently eliminates human anti-FVIII IgG in a mouse model of severe HA with inhibitors and opens a therapeutic window for the administration of exogenous FVIII. OBJECTIVES To investigate the impact of IdeS treatment in inhibitor-positive HA mice injected with emicizumab. METHODS IdeS was injected to HA mice reconstituted with human neutralizing anti-FVIII IgG and treated with emicizumab. RESULTS IdeS hydrolyzed emicizumab in vitro and in vivo, albeit, at slower rates than another recombinant human monoclonal IgG4. While F(ab')2 fragments were rapidly cleared from the circulation, thus leading to a rapid loss of emicizumab procoagulant activity, low amounts of single-cleaved intermediate IgG persisted for several days. Moreover, the IdeS-mediated elimination of the neutralizing anti-FVIII IgG and restoration of the hemostatic efficacy of exogenous FVIII were not impaired by the presence of emicizumab and polyclonal human IgG in inhibitor-positive HA mice. CONCLUSION Our results suggest that IdeS could be administered to inhibitor-positive patients with HA receiving emicizumab prophylaxis to improve and ease the management of breakthrough bleeds or programmed major surgeries.
Collapse
Affiliation(s)
- Melissa Bou-Jaoudeh
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Angelina Mimoun
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sandrine Delignat
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Ivan Peyron
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Ladislas Capdevila
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France; Service d'Hématologie Biologique et Unité Fonctionnelle d'Hémostase, Hôpital Cochin, AP-HP Centre, Université Paris Cité, Paris, France
| | - Victoria Daventure
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Claire Deligne
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jordan D Dimitrov
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Olivier D Christophe
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Cécile V Denis
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Peter J Lenting
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Valérie Proulle
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France; Service d'Hématologie Biologique et Unité Fonctionnelle d'Hémostase, Hôpital Cochin, AP-HP Centre, Université Paris Cité, Paris, France
| | - Sébastien Lacroix-Desmazes
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France.
| |
Collapse
|
13
|
Abdeldaim DT, Schindowski K. Fc-Engineered Therapeutic Antibodies: Recent Advances and Future Directions. Pharmaceutics 2023; 15:2402. [PMID: 37896162 PMCID: PMC10610324 DOI: 10.3390/pharmaceutics15102402] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Monoclonal therapeutic antibodies have revolutionized the treatment of cancer and other diseases. Fc engineering aims to enhance the effector functions or half-life of therapeutic antibodies by modifying their Fc regions. Recent advances in the Fc engineering of modern therapeutic antibodies can be considered the next generation of antibody therapy. Various strategies are employed, including altering glycosylation patterns via glycoengineering and introducing mutations to the Fc region, thereby enhancing Fc receptor or complement interactions. Further, Fc engineering strategies enable the generation of bispecific IgG-based heterodimeric antibodies. As Fc engineering techniques continue to evolve, an expanding portfolio of Fc-engineered antibodies is advancing through clinical development, with several already approved for medical use. Despite the plethora of Fc-based mutations that have been analyzed in in vitro and in vivo models, we focus here in this review on the relevant Fc engineering strategies of approved therapeutic antibodies to finetune effector functions, to modify half-life and to stabilize asymmetric bispecific IgGs.
Collapse
Affiliation(s)
- Dalia T. Abdeldaim
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, 88400 Biberach, Germany;
| |
Collapse
|
14
|
Skiba MA, Sterling SM, Rawson S, Gilman MS, Xu H, Nemeth GR, Hurley JD, Shen P, Staus DP, Kim J, McMahon C, Lehtinen MK, Wingler LM, Kruse AC. Antibodies Expand the Scope of Angiotensin Receptor Pharmacology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.23.554128. [PMID: 37662341 PMCID: PMC10473732 DOI: 10.1101/2023.08.23.554128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
G protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue, and cellular level. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays, and structural studies, we develop maternally selective heavy chain-only antibody ("nanobody") antagonists against the angiotensin II type I receptor (AT1R) and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to AT1R with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.
Collapse
Affiliation(s)
- Meredith A. Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah M. Sterling
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Morgan S.A. Gilman
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Huixin Xu
- Department of Pathology, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Genevieve R. Nemeth
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph D. Hurley
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Pengxiang Shen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Dean P. Staus
- Department of Medicine and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Jihee Kim
- Department of Medicine and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Conor McMahon
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Maria K. Lehtinen
- Department of Pathology, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Laura M. Wingler
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Andrew C. Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Pejchal R, Cooper AB, Brown ME, Vásquez M, Krauland EM. Profiling the Biophysical Developability Properties of Common IgG1 Fc Effector Silencing Variants. Antibodies (Basel) 2023; 12:54. [PMID: 37753968 PMCID: PMC10526015 DOI: 10.3390/antib12030054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023] Open
Abstract
Therapeutic antibodies represent the most significant modality in biologics, with around 150 approved drugs on the market. In addition to specific target binding mediated by the variable fragments (Fvs) of the heavy and light chains, antibodies possess effector functions through binding of the constant region (Fc) to Fcγ receptors (FcγR), which allow immune cells to attack and kill target cells using a variety of mechanisms. However, for some applications, including T-cell-engaging bispecifics, this effector function is typically undesired. Mutations within the lower hinge and the second constant domain (CH2) of IgG1 that comprise the FcγR binding interface reduce or eliminate effector function ("Fc silencing") while retaining binding to the neonatal Fc receptor (FcRn), important for normal antibody pharmacokinetics (PKs). Comprehensive profiling of biophysical developability properties would benefit the choice of constant region variants for development. Here, we produce a large panel of representative mutations previously described in the literature and in many cases in clinical or approved molecules, generate select combinations thereof, and characterize their binding and biophysical properties. We find that some commonly used CH2 mutations, including D265A and P331S, are effective in reducing binding to FcγR but significantly reduce stability, promoting aggregation, particularly under acidic conditions commonly employed in manufacturing. We highlight mutation sets that are particularly effective for eliminating Fc effector function with the retention of WT-like stability, including L234A, L235A, and S267K (LALA-S267K), L234A, L235E, and S267K (LALE-S267K), L234A, L235A, and P329A (LALA-P329A), and L234A, L235E, and P329G (LALE-P329G).
Collapse
Affiliation(s)
- Robert Pejchal
- Adimab LLC, Lebanon, NH 03766, USA; (M.E.B.); (M.V.); (E.M.K.)
| | | | | | | | | |
Collapse
|
16
|
Pyzik M, Kozicky LK, Gandhi AK, Blumberg RS. The therapeutic age of the neonatal Fc receptor. Nat Rev Immunol 2023; 23:415-432. [PMID: 36726033 PMCID: PMC9891766 DOI: 10.1038/s41577-022-00821-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 02/03/2023]
Abstract
IgGs are essential soluble components of the adaptive immune response that evolved to protect the body from infection. Compared with other immunoglobulins, the role of IgGs is distinguished and enhanced by their high circulating levels, long half-life and ability to transfer from mother to offspring, properties that are conferred by interactions with neonatal Fc receptor (FcRn). FcRn binds to the Fc portion of IgGs in a pH-dependent manner and protects them from intracellular degradation. It also allows their transport across polarized cells that separate tissue compartments, such as the endothelium and epithelium. Further, it is becoming apparent that FcRn functions to potentiate cellular immune responses when IgGs, bound to their antigens, form IgG immune complexes. Besides the protective role of IgG, IgG autoantibodies are associated with numerous pathological conditions. As such, FcRn blockade is a novel and effective strategy to reduce circulating levels of pathogenic IgG autoantibodies and curtail IgG-mediated diseases, with several FcRn-blocking strategies on the path to therapeutic use. Here, we describe the current state of knowledge of FcRn-IgG immunobiology, with an emphasis on the functional and pathological aspects, and an overview of FcRn-targeted therapy development.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa K Kozicky
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amit K Gandhi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Digestive Diseases Center, Boston, MA, USA.
| |
Collapse
|
17
|
Trastoy B, Du JJ, Cifuente JO, Rudolph L, García-Alija M, Klontz EH, Deredge D, Sultana N, Huynh CG, Flowers MW, Li C, Sastre DE, Wang LX, Corzana F, Mallagaray A, Sundberg EJ, Guerin ME. Mechanism of antibody-specific deglycosylation and immune evasion by Streptococcal IgG-specific endoglycosidases. Nat Commun 2023; 14:1705. [PMID: 36973249 PMCID: PMC10042849 DOI: 10.1038/s41467-023-37215-3] [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: 06/19/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
Bacterial pathogens have evolved intricate mechanisms to evade the human immune system, including the production of immunomodulatory enzymes. Streptococcus pyogenes serotypes secrete two multi-modular endo-β-N-acetylglucosaminidases, EndoS and EndoS2, that specifically deglycosylate the conserved N-glycan at Asn297 on IgG Fc, disabling antibody-mediated effector functions. Amongst thousands of known carbohydrate-active enzymes, EndoS and EndoS2 represent just a handful of enzymes that are specific to the protein portion of the glycoprotein substrate, not just the glycan component. Here, we present the cryoEM structure of EndoS in complex with the IgG1 Fc fragment. In combination with small-angle X-ray scattering, alanine scanning mutagenesis, hydrolytic activity measurements, enzyme kinetics, nuclear magnetic resonance and molecular dynamics analyses, we establish the mechanisms of recognition and specific deglycosylation of IgG antibodies by EndoS and EndoS2. Our results provide a rational basis from which to engineer novel enzymes with antibody and glycan selectivity for clinical and biotechnological applications.
Collapse
Affiliation(s)
- Beatriz Trastoy
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain.
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain.
| | - Jonathan J Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Javier O Cifuente
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Lorena Rudolph
- University of Lübeck, Center of Structural and Cell Biology in Medicine (CSCM), Institute of Chemistry and Metabolomics, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Mikel García-Alija
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Erik H Klontz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Daniel Deredge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Nazneen Sultana
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Chau G Huynh
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Maria W Flowers
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742, USA
| | - Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20742, USA
| | - Francisco Corzana
- Departamento Química and Centro de Investigación en Síntesis Quı́mica, Universidad de La Rioja, 26006, Rioja, Spain
| | - Alvaro Mallagaray
- University of Lübeck, Center of Structural and Cell Biology in Medicine (CSCM), Institute of Chemistry and Metabolomics, Ratzeburger Allee 160, 23562, Lübeck, Germany.
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain.
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain.
| |
Collapse
|
18
|
Planchais C, Reyes‐Ruiz A, Lacombe R, Zarantonello A, Lecerf M, Revel M, Roumenina LT, Atanasov BP, Mouquet H, Dimitrov JD. Evolutionary trajectory of receptor binding specificity and promiscuity of the spike protein of SARS-CoV-2. Protein Sci 2022; 31:e4447. [PMID: 36305765 PMCID: PMC9597384 DOI: 10.1002/pro.4447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 01/27/2023]
Abstract
SARS-CoV-2 infects cells by attachment to its receptor-the angiotensin converting enzyme 2 (ACE2). Regardless of the wealth of structural data, little is known about the physicochemical mechanism of interactions of the viral spike (S) protein with ACE2 and how this mechanism has evolved during the pandemic. Here, we applied experimental and computational approaches to characterize the molecular interaction of S proteins from SARS-CoV-2 variants of concern (VOC). Data on kinetics, activation-, and equilibrium thermodynamics of binding of the receptor binding domain (RBD) from VOC with ACE2 as well as data from computational protein electrostatics revealed a profound remodeling of the physicochemical characteristics of the interaction during the evolution. Thus, as compared to RBDs from Wuhan strain and other VOC, Omicron RBD presented as a unique protein in terms of conformational dynamics and types of non-covalent forces driving the complex formation with ACE2. Viral evolution resulted in a restriction of the RBD structural dynamics, and a shift to a major role of polar forces for ACE2 binding. Further, we investigated how the reshaping of the physicochemical characteristics of interaction affects the binding specificity of S proteins. Data from various binding assays revealed that SARS-CoV-2 Wuhan and Omicron RBDs manifest capacity for promiscuous recognition of unrelated human proteins, but they harbor distinct reactivity patterns. These findings might contribute for mechanistic understanding of the viral tropism and capacity to evade immune responses during evolution.
Collapse
Affiliation(s)
- Cyril Planchais
- Laboratory of Humoral ImmunologyInstitut Pasteur, Université Paris Cité, INSERM U1222ParisFrance
| | - Alejandra Reyes‐Ruiz
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Robin Lacombe
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Alessandra Zarantonello
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Maxime Lecerf
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Margot Revel
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Lubka T. Roumenina
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Boris P. Atanasov
- Institute of Organic Chemistry, Bulgarian Academy of SciencesSofiaBulgaria
| | - Hugo Mouquet
- Laboratory of Humoral ImmunologyInstitut Pasteur, Université Paris Cité, INSERM U1222ParisFrance
| | - Jordan D. Dimitrov
- Centre de Recherche des CordeliersINSERM, CNRS, Sorbonne Université, Université de ParisParisFrance
| |
Collapse
|
19
|
An Fc variant with two mutations confers prolonged serum half-life and enhanced effector functions on IgG antibodies. Exp Mol Med 2022; 54:1850-1861. [PMID: 36319752 PMCID: PMC9628495 DOI: 10.1038/s12276-022-00870-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/07/2022] Open
Abstract
The pH-selective interaction between the immunoglobulin G (IgG) fragment crystallizable region (Fc region) and the neonatal Fc receptor (FcRn) is critical for prolonging the circulating half-lives of IgG molecules through intracellular trafficking and recycling. By using directed evolution, we successfully identified Fc mutations that improve the pH-dependent binding of human FcRn and prolong the serum persistence of a model IgG antibody and an Fc-fusion protein. Strikingly, trastuzumab-PFc29 and aflibercept-PFc29, a model therapeutic IgG antibody and an Fc-fusion protein, respectively, when combined with our engineered Fc (Q311R/M428L), both exhibited significantly higher serum half-lives in human FcRn transgenic mice than their counterparts with wild-type Fc. Moreover, in a cynomolgus monkey model, trastuzumab-PFc29 displayed a superior pharmacokinetic profile to that of both trastuzumab-YTE and trastuzumab-LS, which contain the well-validated serum half-life extension Fcs YTE (M252Y/S254T/T256E) and LS (M428L/N434S), respectively. Furthermore, the introduction of two identified mutations of PFc29 (Q311R/M428L) into the model antibodies enhanced both complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity activity, which are triggered by the association between IgG Fc and Fc binding ligands and are critical for clearing cancer cells. In addition, the effector functions could be turned off by combining the two mutations of PFc29 with effector function-silencing mutations, but the antibodies maintained their excellent pH-dependent human FcRn binding profile. We expect our Fc variants to be an excellent tool for enhancing the pharmacokinetic profiles and potencies of various therapeutic antibodies and Fc-fusion proteins.
Collapse
|
20
|
The Fab region of IgG impairs the internalization pathway of FcRn upon Fc engagement. Nat Commun 2022; 13:6073. [PMID: 36241613 PMCID: PMC9568614 DOI: 10.1038/s41467-022-33764-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Binding to the neonatal Fc receptor (FcRn) extends serum half-life of IgG, and antagonizing this interaction is a promising therapeutic approach in IgG-mediated autoimmune diseases. Fc-MST-HN, designed for enhanced FcRn binding capacity, has not been evaluated in the context of a full-length antibody, and the structural properties of the attached Fab regions might affect the FcRn-mediated intracellular trafficking pathway. Here we present a comprehensive comparative analysis of the IgG salvage pathway between two full-size IgG1 variants, containing wild type and MST-HN Fc fragments, and their Fc-only counterparts. We find no evidence of Fab-regions affecting FcRn binding in cell-free assays, however, cellular assays show impaired binding of full-size IgG to FcRn, which translates into improved intracellular FcRn occupancy and intracellular accumulation of Fc-MST-HN compared to full size IgG1-MST-HN. The crystal structure of Fc-MST-HN in complex with FcRn provides a plausible explanation why the Fab disrupts the interaction only in the context of membrane-associated FcRn. Importantly, we find that Fc-MST-HN outperforms full-size IgG1-MST-HN in reducing IgG levels in cynomolgus monkeys. Collectively, our findings identify the cellular membrane context as a critical factor in FcRn biology and therapeutic targeting.
Collapse
|
21
|
Monoclonal Antibody Engineering and Design to Modulate FcRn Activities: A Comprehensive Review. Int J Mol Sci 2022; 23:ijms23179604. [PMID: 36077002 PMCID: PMC9455995 DOI: 10.3390/ijms23179604] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 01/03/2023] Open
Abstract
Understanding the biological mechanisms underlying the pH-dependent nature of FcRn binding, as well as the various factors influencing the affinity to FcRn, was concurrent with the arrival of the first recombinant IgG monoclonal antibodies (mAbs) and IgG Fc-fusion proteins in clinical practice. IgG Fc–FcRn became a central subject of interest for the development of these drugs for the comfort of patients and good clinical responses. In this review, we describe (i) mAb mutations close to and outside the FcRn binding site, increasing the affinity for FcRn at acidic pH and leading to enhanced mAb half-life and biodistribution, and (ii) mAb mutations increasing the affinity for FcRn at acidic and neutral pH, blocking FcRn binding and resulting, in vivo, in endogenous IgG degradation. Mutations modifying FcRn binding are discussed in association with pH-dependent modulation of antigen binding and (iii) anti-FcRn mAbs, two of the latest innovations in anti-FcRn mAbs leading to endogenous IgG depletion. We discuss the pharmacological effects, the biological consequences, and advantages of targeting IgG–FcRn interactions and their application in human therapeutics.
Collapse
|
22
|
Biophysical differences in IgG1 Fc-based therapeutics relate to their cellular handling, interaction with FcRn and plasma half-life. Commun Biol 2022; 5:832. [PMID: 35982144 PMCID: PMC9388496 DOI: 10.1038/s42003-022-03787-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/01/2022] [Indexed: 01/07/2023] Open
Abstract
Antibody-based therapeutics (ABTs) are used to treat a range of diseases. Most ABTs are either full-length IgG1 antibodies or fusions between for instance antigen (Ag)-binding receptor domains and the IgG1 Fc fragment. Interestingly, their plasma half-life varies considerably, which may relate to how they engage the neonatal Fc receptor (FcRn). As such, there is a need for an in-depth understanding of how different features of ABTs affect FcRn-binding and transport behavior. Here, we report on how FcRn-engagement of the IgG1 Fc fragment compare to clinically relevant IgGs and receptor domain Fc fusions, binding to VEGF or TNF-α. The results reveal FcRn-dependent intracellular accumulation of the Fc, which is in line with shorter plasma half-life than that of full-length IgG1 in human FcRn-expressing mice. Receptor domain fusion to the Fc increases its half-life, but not to the extent of IgG1. This is mirrored by a reduced cellular recycling capacity of the Fc-fusions. In addition, binding of cognate Ag to ABTs show that complexes of similar size undergo cellular transport at different rates, which could be explained by the biophysical properties of each ABT. Thus, the study provides knowledge that should guide tailoring of ABTs regarding optimal cellular sorting and plasma half-life. Analysis of clinically approved antibody-based therapeutics reveals different structural designs, such as full-length IgG1 or Fc-fusions, entail distinct biophysical properties that affect FcRn binding, intracellular transport and plasma half-life.
Collapse
|
23
|
Wei W, Corbeil CR, Gaudreault F, Deprez C, Purisima EO, Sulea T. Antibody mutations favoring
pH
‐dependent binding in solid tumor microenvironments: Insights from large‐scale structure‐based calculations. Proteins 2022; 90:1538-1546. [DOI: 10.1002/prot.26340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/26/2022] [Accepted: 03/23/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Wanlei Wei
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Christopher R. Corbeil
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Francis Gaudreault
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Christophe Deprez
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Enrico O. Purisima
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Traian Sulea
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| |
Collapse
|
24
|
House RV, Broge TA, Suscovich TJ, Snow DM, Tomic MT, Nonet G, Bajwa K, Zhu G, Martinez Z, Hackett K, Earnhart CG, Dorsey NM, Hopkins SA, Natour DS, Davis HD, Anderson MS, Gainey MR, Cobb RR. Evaluation of strategies to modify Anti-SARS-CoV-2 monoclonal antibodies for optimal functionality as therapeutics. PLoS One 2022; 17:e0267796. [PMID: 35657812 PMCID: PMC9165815 DOI: 10.1371/journal.pone.0267796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 04/15/2022] [Indexed: 01/08/2023] Open
Abstract
The current global COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a public health crisis with more than 168 million cases reported globally and more than 4.5 million deaths at the time of writing. In addition to the direct impact of the disease, the economic impact has been significant as public health measures to contain or reduce the spread have led to country wide lockdowns resulting in near closure of many sectors of the economy. Antibodies are a principal determinant of the humoral immune response to COVID-19 infections and may have the potential to reduce disease and spread of the virus. The development of monoclonal antibodies (mAbs) represents a therapeutic option that can be produced at large quantity and high quality. In the present study, a mAb combination mixture therapy was investigated for its capability to specifically neutralize SARS-CoV-2. We demonstrate that each of the antibodies bind the spike protein and neutralize the virus, preventing it from infecting cells in an in vitro cell-based assay, including multiple viral variants that are currently circulating in the human population. In addition, we investigated the effects of two different mutations in the Fc portion (YTE and LALA) of the antibody on Fc effector function and the ability to alleviate potential antibody-dependent enhancement of disease. These data demonstrate the potential of a combination of two mAbs that target two different epitopes on the SARS-CoV2 spike protein to provide protection against SARS-CoV-2 infection in humans while extending serum half-life and preventing antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
| | | | | | - Doris M. Snow
- Ology Bioservices, Frederick, MD, United States of America
| | - Milan T. Tomic
- Research and Development, Ology Bioservices, Inc., Alameda, CA, United States of America
| | - Genevieve Nonet
- Research and Development, Ology Bioservices, Inc., Alameda, CA, United States of America
| | - Kamaljit Bajwa
- Research and Development, Ology Bioservices, Inc., Alameda, CA, United States of America
| | - Guangyu Zhu
- Research and Development, Ology Bioservices, Inc., Alameda, CA, United States of America
| | - Zachary Martinez
- Research and Development, Ology Bioservices, Inc., Alameda, CA, United States of America
| | - Kyal Hackett
- Ology Bioservices, Frederick, MD, United States of America
| | - Christopher G. Earnhart
- US Department of Defense, Joint Program Executive Office for Chemical, Biological, Radiological, Nuclear Defense (JPEO-CBRND), Washington, DC, United States of America
| | - Nicole M. Dorsey
- US Department of Defense, Joint Program Executive Office for Chemical, Biological, Radiological, Nuclear Defense (JPEO-CBRND), Washington, DC, United States of America
| | | | - Dalia S. Natour
- Battelle Biomedical Research Center, West Jefferson, Columbus, Ohio, United States of America
| | - Heather D. Davis
- Battelle Biomedical Research Center, West Jefferson, Columbus, Ohio, United States of America
| | - Michael S. Anderson
- Battelle Biomedical Research Center, West Jefferson, Columbus, Ohio, United States of America
| | - Melicia R. Gainey
- Battelle Biomedical Research Center, West Jefferson, Columbus, Ohio, United States of America
| | - Ronald R. Cobb
- Process Development, Ology Bioservices, Alachua, FL, United States of America
| |
Collapse
|
25
|
Di Trani CA, Cirella A, Arrizabalaga L, Fernandez-Sendin M, Bella A, Aranda F, Melero I, Berraondo P. Overcoming the limitations of cytokines to improve cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:107-141. [PMID: 35777862 DOI: 10.1016/bs.ircmb.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytokines are pleiotropic soluble proteins used by immune cells to orchestrate a coordinated response against pathogens and malignancies. In cancer immunotherapy, cytokine-based drugs can be developed potentiating pro-inflammatory cytokines or blocking immunosuppressive cytokines. However, the complexity of the mechanisms of action of cytokines requires the use of biotechnological strategies to minimize systemic toxicity, while potentiating the antitumor response. Sequence mutagenesis, fusion proteins and gene therapy strategies are employed to enhance the half-life in circulation, target the desired bioactivity to the tumor microenvironment, and to optimize the therapeutic window of cytokines. In this review, we provide an overview of the different strategies currently being pursued in pre-clinical and clinical studies to make the most of cytokines for cancer immunotherapy.
Collapse
Affiliation(s)
- Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Leire Arrizabalaga
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Angela Bella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
26
|
Singh SK, Kumar D, Nagpal S, Dubey SK, Rathore AS. A Charge Variant of Bevacizumab Offers Enhanced FcRn-Dependent Pharmacokinetic Half-Life and Efficacy. Pharm Res 2022; 39:851-865. [PMID: 35355206 DOI: 10.1007/s11095-022-03236-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/14/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Lysine variants of monoclonal antibodies (mAbs) result from incomplete clipping of the C-terminal lysine residues of the heavy chain. Although the structure of the lysine variants has been determined for several mAb products, a detailed study that investigates the impact of lysine charge variants on PK/PD and preclinical safety is yet to be published. OBJECTIVE An in-depth investigation of the impact of C- terminal lysine clipping of mAbs on safety and efficacy for bevacizumab charge variants. METHOD Charge variant isolation using semi-preparative chromatography is followed by a comparative analysis of FcRn binding, pharmacokinetics, and pharmacodynamics in relevant animal models. RESULTS K1 variant exhibited improved FcRn binding affinity (4-fold), half-life (1.3-fold), and anti-tumor activity (1.3-fold) as compared to the K0 (main) product. However, the K2 variant, even though exhibited higher FcRn affinity (2-fold), displayed lower half-life (1.6-fold) and anti-tumor activity at medium and low doses. Differential proteomic analysis revealed that seven pathways (such as glycolysis, gluconeogenesis, carbon metabolism, synthesis of amino acids) were significantly enriched. Higher efficacy of the K1 variant is likely due to higher bioavailability of the drug, leading to complete downregulation of the pathways that facilitate catering of the energy requirements of the proliferating tumor cells. On the contrary, the K2 variant exhibits a shorter half-life, resulting only in partial reduction in the metabolic/energy requirements of the growing tumor cells. CONCLUSION Overall, we conclude that the mAb half-life, dosage, and efficacy of a biotherapeutic product are significantly impacted by the charge variant profile of a biotherapeutic product.
Collapse
Affiliation(s)
- Sumit K Singh
- School of Biochemical Engineering, IIT(BHU), Varanasi, India
| | - Deepak Kumar
- Department of Chemical Engineering, IIT, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology, Hauz Khas, New Delhi, 110016, India
| | | | - Sunil K Dubey
- R&D Healthcare Division, Emami Limited, Kolkata, India
| | - Anurag S Rathore
- Department of Chemical Engineering, IIT, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
27
|
Lee T, Kim JH, Kwon SJ, Seo JW, Park SH, Kim J, Jin J, Hong JH, Kang HJ, Sharma C, Choi JH, Chung SJ. Site-Selective Antibody-Drug Conjugation by a Proximity-Driven S to N Acyl Transfer Reaction on a Therapeutic Antibody. J Med Chem 2022; 65:5751-5759. [PMID: 35319890 DOI: 10.1021/acs.jmedchem.2c00084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immunoglobulin Gs (IgGs) contain many Lys and Cys residues, which results in an unwanted complex product mixture with conventional drug conjugation methods. We selectively acylated the ε-NH2 of K248 on trastuzumab using an IgG Fc-binding peptide (FcBP) equipped with a 5-norbornene-2-carboxylic acid thioester (AbClick-1). AbClick-1 locates its thioester close to the ε-NH2 of K248 while binding to trastuzumab. Consequently, the thioester underwent proximity-driven selective acylation of ε-NH2 through an S to N acyl transfer reaction. Furthermore, N-tert-butyl maleimide accelerated the cross-linking reaction with an approximately 95% yield of the desired product by scavenging the byproduct (FcBP-SH). Only K248 was modified selectively with the 5-norbornene-2-carbonyl group, which was further modified by click reaction to afford an antibody-drug conjugate (ADC) with two drugs per antibody. The resulting ADCs showed remarkable in vitro and in vivo anticancer activity. Our results demonstrate that a thioester is a promising chemical entity for proximity-driven site-selective conjugation of antibodies.
Collapse
Affiliation(s)
- TaeJin Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea.,AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea
| | - Ju Hwan Kim
- AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea
| | - Se Jeong Kwon
- School of Pharmacy, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea.,AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea
| | - Jin Woo Seo
- School of Pharmacy, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea.,AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea
| | - Sun Hee Park
- School of Pharmacy, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea.,AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea
| | - Jinyoung Kim
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Republic of Korea
| | - Jonghwa Jin
- Department of Convergence Technical Support, New Drug Development Center, 123 Osongsaengmyeng-ro, Cheongju, Chungbuk 28160, Republic of Korea
| | - Ji Hye Hong
- Department of Convergence Technical Support, New Drug Development Center, 123 Osongsaengmyeng-ro, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hyo Jin Kang
- AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea
| | - Chiranjeev Sharma
- Department of Biopharmaceutical Convergence, Graduate School, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea
| | - Ji Hoon Choi
- School of Pharmacy, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea
| | - Sang J Chung
- School of Pharmacy, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea.,AbTis Company Ltd., A-815, Suwon Venture Valley II, 142-10, Saneop-ro156beon-gil, Gwonseon-gu, Suwon, Gyeonggi-do 16648, Republic of Korea.,Department of Biopharmaceutical Convergence, Graduate School, Sungkyunkwan University, 2066 Seoburo, Jangangu, Suwon 16419, Republic of Korea
| |
Collapse
|
28
|
Grevys A, Frick R, Mester S, Flem-Karlsen K, Nilsen J, Foss S, Sand KMK, Emrich T, Fischer JAA, Greiff V, Sandlie I, Schlothauer T, Andersen JT. Antibody variable sequences have a pronounced effect on cellular transport and plasma half-life. iScience 2022; 25:103746. [PMID: 35118359 PMCID: PMC8800109 DOI: 10.1016/j.isci.2022.103746] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 01/05/2022] [Indexed: 11/15/2022] Open
Abstract
Monoclonal IgG antibodies are the fastest growing class of biologics, but large differences exist in their plasma half-life in humans. Thus, to design IgG antibodies with favorable pharmacokinetics, it is crucial to identify the determinants of such differences. Here, we demonstrate that the variable region sequences of IgG antibodies greatly affect cellular uptake and subsequent recycling and rescue from intracellular degradation by endothelial cells. When the variable sequences are masked by the cognate antigen, it influences both their transport behavior and binding to the neonatal Fc receptor (FcRn), a key regulator of IgG plasma half-life. Furthermore, we show how charge patch differences in the variable domains modulate both binding and transport properties and that a short plasma half-life, due to unfavorable charge patches, may partly be overcome by Fc-engineering for improved FcRn binding. IgG variable region sequences greatly affect cellular uptake and recycling Variable region charge patches affect FcRn binding and transport The presence of cognate antigen modulates cellular transport and FcRn binding Fc-engineering for improved FcRn binding can overcome unfavorable charge patches
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
- Corresponding author
| | - Rahel Frick
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Simone Mester
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Karine Flem-Karlsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Stian Foss
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Thomas Emrich
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | | | - Victor Greiff
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Corresponding author
| |
Collapse
|
29
|
Gstöttner C, Hook M, Christopeit T, Knaupp A, Schlothauer T, Reusch D, Haberger M, Wuhrer M, Domínguez-Vega E. Affinity Capillary Electrophoresis-Mass Spectrometry as a Tool to Unravel Proteoform-Specific Antibody-Receptor Interactions. Anal Chem 2021; 93:15133-15141. [PMID: 34739220 PMCID: PMC8600502 DOI: 10.1021/acs.analchem.1c03560] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Monoclonal antibody (mAb) pharmaceuticals consist of a plethora of different proteoforms with different functional characteristics, including pharmacokinetics and pharmacodynamics, requiring their individual assessment. Current binding techniques do not distinguish between coexisting proteoforms requiring tedious production of enriched proteoforms. Here, we have developed an approach based on mobility shift-affinity capillary electrophoresis-mass spectrometry (ACE-MS), which permitted us to determine the binding of coexisting mAb proteoforms to Fc receptors (FcRs). For high-sensitivity MS analysis, we used a sheathless interface providing adequate mAb sensitivity allowing functional characterization of mAbs with a high sensitivity and dynamic range. As a model system, we focused on the interaction with the neonatal FcR (FcRn), which determines the half-life of mAbs. Depending on the oxidation status, proteoforms exhibited different electrophoretic mobility shifts in the presence of FcRn, which could be used to determine their affinity. We confirmed the decrease of the FcRn affinity with antibody oxidation and observed a minor glycosylation effect, with higher affinities for galactosylated glycoforms. Next to relative binding, the approach permits the determination of individual KD values in solution resulting in values of 422 and 139 nM for double-oxidized and non-oxidized variants. Hyphenation with native MS provides unique capabilities for simultaneous heterogeneity assessment for mAbs, FcRn, and complexes formed. The latter provides information on binding stoichiometry revealing 1:1 and 1:2 for antibody/FcRn complexes. The use of differently engineered Fc-only constructs allowed distinguishing between symmetric and asymmetric binding. The approach opens up unique possibilities for proteoform-resolved antibody binding studies to FcRn and can be extended to other FcRs and protein interactions.
Collapse
Affiliation(s)
- Christoph Gstöttner
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Michaela Hook
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Tony Christopeit
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Alexander Knaupp
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Tilman Schlothauer
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Dietmar Reusch
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Markus Haberger
- Pharma Technical Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333ZA, The Netherlands
| |
Collapse
|
30
|
Pannek A, Houghton FJ, Verhagen AM, Dower SK, Hinde E, Gleeson PA. Dynamics of intracellular neonatal Fc receptor-ligand interactions in primary macrophages using biophysical fluorescence techniques. Mol Biol Cell 2021; 33:ar6. [PMID: 34731029 PMCID: PMC8886815 DOI: 10.1091/mbc.e21-02-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is responsible for the recycling of endocytosed albumin and IgG, and contributes to their long plasma half-life. We recently identified an FcRn-dependent recycling pathway from macropinosomes in macrophages; however, little is known about the dynamics of intracellular FcRn–ligand interactions to promote recycling. Here we demonstrate a multiplexed biophysical fluorescent microscopy approach to resolve the spatiotemporal dynamics of albumin–FcRn interactions in living bone marrow–derived macrophages (BMDMs). We used the phasor approach to fluorescence lifetime imaging microscopy (FLIM) of Förster resonance energy transfer (FRET) to detect the interaction of a FcRn–mCherry fusion protein with endocytosed Alexa Fluor 488–labeled human serum albumin (HSA–AF488) in BMDMs, and raster image correlation spectroscopy (RICS) analysis of single fluorescent-labeled albumin molecules to monitor the diffusion kinetics of internalized albumin. Our data identified a major fraction of immobile HSA–AF488 molecules in endosomal structures of human FcRn-positive mouse macrophages and an increase in FLIM-FRET following endocytosis, including detection of FRET in tubular-like structures. A nonbinding mutant of albumin showed minimum FLIM-FRET and high mobility. These data reveal the kinetics of FcRn–ligand binding within endosomal structures for recruitment into transport carriers for recycling. These approaches have wide applicability for analyses of intracellular ligand–receptor interactions.
Collapse
Affiliation(s)
- Andreas Pannek
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010.,Institute of Experimental Immunology, University of Bonn, Venusberg Campus, D-53127, Germany
| | - Fiona J Houghton
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010
| | - Anne M Verhagen
- CSL Limited, Research, Bio21 Molecular Science and Biotechnology Institute, Victoria 3010, Australia
| | - Steven K Dower
- CSL Limited, Research, Bio21 Molecular Science and Biotechnology Institute, Victoria 3010, Australia
| | - Elizabeth Hinde
- School of Physics and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia.,Department of Biochemistry and Pharmacology, The University of Melbourne, Victoria 3010, Australia
| | - Paul A Gleeson
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010
| |
Collapse
|
31
|
In vivo pharmacokinetic enhancement of monomeric Fc and monovalent bispecific designs through structural guidance. Commun Biol 2021; 4:1048. [PMID: 34497355 PMCID: PMC8426389 DOI: 10.1038/s42003-021-02565-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/18/2021] [Indexed: 11/08/2022] Open
Abstract
In a biologic therapeutic landscape that requires versatility in targeting specificity, valency and half-life modulation, the monomeric Fc fusion platform holds exciting potential for the creation of a class of monovalent protein therapeutics that includes fusion proteins and bispecific targeting molecules. Here we report a structure-guided approach to engineer monomeric Fc molecules to adapt multiple versions of half-life extension modifications. Co-crystal structures of these monomeric Fc variants with Fc neonatal receptor (FcRn) shed light into the binding interactions that could serve as a guide for engineering the half-life of antibody Fc fragments. These engineered monomeric Fc molecules also enabled the generation of a novel monovalent bispecific molecular design, which translated the FcRn binding enhancement to improvement of in vivo serum half-life. Lu Shan et al. present a structure-guided approach to engineer a monovalent form of the fragment crystallizable (Fc) region of an IgG4 antibody to adapt multiple versions of half-life extension modifications and bispecific targeting. Additionally, they report co-crystal structures of the variants bound to the Fc neonatal receptor that allow insights into the binding interactions.
Collapse
|
32
|
Azevedo C, Pinto S, Benjakul S, Nilsen J, Santos HA, Traverso G, Andersen JT, Sarmento B. Prevention of diabetes-associated fibrosis: Strategies in FcRn-targeted nanosystems for oral drug delivery. Adv Drug Deliv Rev 2021; 175:113778. [PMID: 33887405 DOI: 10.1016/j.addr.2021.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/29/2021] [Accepted: 04/16/2021] [Indexed: 01/02/2023]
Abstract
Diabetes mellitus is a chronic disease with an elevated risk of micro- and macrovascular complications, such as fibrosis. To prevent diabetes-associated fibrosis, the symptomatology of diabetes must be controlled, which is commonly done by subcutaneous injection of antidiabetic peptides. To minimize the pain and distress associated with such injections, there is an urgent need for non-invasive oral transmucosal drug delivery strategies. However, orally administered peptide-based drugs are exposed to harsh conditions in the gastrointestinal tract and poorly cross the selective intestinal epithelium. Thus, targeting of drugs to receptors expressed in epithelial cells, such as the neonatal Fc receptor (FcRn), may therefore enhance uptake and transport through mucosal barriers. This review compiles how in-depth studies of FcRn biology and engineering of receptor-binding molecules may pave the way for design of new classes of FcRn-targeted nanosystems. Tailored strategies may open new avenues for oral drug delivery and provide better treatment options for diabetes and, consequently, fibrosis prevention.
Collapse
|
33
|
Sun Y, Estevez A, Schlothauer T, Wecksler AT. Antigen physiochemical properties allosterically effect the IgG Fc-region and Fc neonatal receptor affinity. MAbs 2021; 12:1802135. [PMID: 32795110 PMCID: PMC7531492 DOI: 10.1080/19420862.2020.1802135] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is a key membrane protein that plays an integral role in serum immunoglobulin (IgG) recycling, which extends the half-life of antibody. In addition, FcRn is known to traffic antigen-bound immunoglobulins (Ag-IgGs), and to interact with immune complexes to facilitate the antigen cross-presentation of peptides derived from the immune complexes in antigen-presenting cells (APCs). Studies on the IgG-FcRn molecular interactions have primarily focused on the Fc region, and only recently have shown the potential impact of the antigen-binding fragment physiochemical properties on FcRn binding. However, the effect of the antigen physiochemical properties on IgG structure as it relates to Ag-IgG-FcRn binding is not well understood. Here we used an IgG-peptide antigen complex as a model system to investigate the structural effects of the antigen's physiochemical properties on the IgG structure, and the subsequent effects of Ag-IgG-FcRn interactions. We used hydroxyl radical footprinting-mass spectrometry to investigate the structural impact on an IgG upon antigen binding, and observed that the physicochemical properties of the antigen differentially induce conformational changes in the IgG FcRn binding region. The extent of these structural changes directly correlates to the magnitude of the affinity differences between the Ag-IgG complexes and FcRn. Moreover, the antigen's physicochemical properties differentially induce structural differences within the Ag-IgG-FcRn ternary complex. We also provide electron microscopy data that shows corroborating Fab-FcRn interactions, and confirms the hypothesis of potential 2:1 FcRn:IgG binding stoichiometry. These data demonstrate antigen-induced Fc structural rearrangements affect both the affinity toward FcRn and the trimeric antigen-IgG-FcRn complex, providing novel molecular insights in the first steps toward understanding interactions of FcRn-containing large(r)-sized immune complex.
Collapse
Affiliation(s)
- Yue Sun
- Protein Analytical Chemistry, Genentech Inc ., South San Francisco, CA, USA
| | - Alberto Estevez
- Structural Biology, Genentech Inc ., South San Francisco, CA, USA
| | - Tilman Schlothauer
- Roche Pharma Research & Early Development, Roche Innovation Center Munich , Penzberg, Germany.,Biological Technologies, Genentech Inc ., South San Francisco, CA, USA
| | - Aaron T Wecksler
- Protein Analytical Chemistry, Genentech Inc ., South San Francisco, CA, USA
| |
Collapse
|
34
|
A structural perspective on the design of decoy immune modulators. Pharmacol Res 2021; 170:105735. [PMID: 34146695 DOI: 10.1016/j.phrs.2021.105735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Therapeutic mAbs have dominated the class of immunotherapeutics in general and immune checkpoint inhibitors in particular. The high specificity of mAbs to the target molecule as well as their extended half-life and (or) the effector functions raised by the Fc part are some of the important aspects that contribute to the success of this class of therapeutics. Equally potential candidates are decoys and their fusions that can address some of the inherent limitations of mAbs, like immunogenicity, resistance development, low bio-availability and so on, besides maintaining the advantages of mAbs. The decoys are molecules that trap the ligands and prevent them from interacting with the signaling receptors. Although a few FDA-approved decoy immune modulators are very successful, the potential of this class of drugs is yet to be fully realized. Here, we review various strategies employed in fusion protein therapeutics with a focus on the design of decoy immunomodulators from the structural perspective and discuss how the information on protein structure and function can strategically guide the development of next-generation immune modulators.
Collapse
|
35
|
Fieux M, Le Quellec S, Bartier S, Coste A, Louis B, Giroudon C, Nourredine M, Bequignon E. FcRn as a Transporter for Nasal Delivery of Biologics: A Systematic Review. Int J Mol Sci 2021; 22:6475. [PMID: 34204226 PMCID: PMC8234196 DOI: 10.3390/ijms22126475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022] Open
Abstract
FcRn plays a major role in regulating immune homeostasis, but it is also able to transport biologics across cellular barriers. The question of whether FcRn could be an efficient transporter of biologics across the nasal epithelial barrier is of particular interest, as it would allow a less invasive strategy for the administration of biologics in comparison to subcutaneous, intramuscular or intravenous administrations, which are often used in clinical practice. A focused systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. It was registered on the international prospective register of systematic reviews PROSPERO, which helped in identifying articles that met the inclusion criteria. Clinical and preclinical studies involving FcRn and the nasal delivery of biologics were screened, and the risk of bias was assessed across studies using the Oral Health Assessment Tool (OHAT). Among the 12 studies finally included in this systematic review (out of the 758 studies screened), 11 demonstrated efficient transcytosis of biologics through the nasal epithelium. Only three studies evaluated the potential toxicity of biologics' intranasal delivery, and they all showed that it was safe. This systematic review confirmed that FcRn is expressed in the nasal airway and the olfactory epithelium, and that FcRn may play a role in IgG and/or IgG-derived molecule-transcytosis across the airway epithelium. However, additional research is needed to better characterize the pharmacokinetic and pharmacodynamic properties of biologics after their intranasal delivery.
Collapse
Affiliation(s)
- Maxime Fieux
- Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Service d’ORL, D’otoneurochirurgie et de Chirurgie Cervico-Faciale, Pierre Bénite, CEDEX, F-69495 Lyon, France
- Université de Lyon, Université Lyon 1, F-69003 Lyon, France; (S.L.Q.); (M.N.)
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
| | - Sandra Le Quellec
- Université de Lyon, Université Lyon 1, F-69003 Lyon, France; (S.L.Q.); (M.N.)
- Hospices Civils de Lyon, Hôpital Cardiologique Louis Pradel, Unité D’hémostase Clinique, CEDEX, F-69500 Bron, France
- EA 4609 Hémostase et Cancer, Université Claude Bernard Lyon 1, F-69372 Lyon, France
- Hospices Civils de Lyon, Centre de Biologie et de Pathologie Est, Service D’hématologie Biologique, CEDEX, F-69500 Bron, France
| | - Sophie Bartier
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
- Service d’ORL, de Chirurgie Cervico Faciale, Hôpital Henri Mondor, Assistance Publique des Hôpitaux de Paris, F-94000 Créteil, France
| | - André Coste
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
- Service d’ORL, de Chirurgie Cervico Faciale, Centre Hospitalier Intercommunal de Créteil, F-94010 Créteil, France
| | - Bruno Louis
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
| | - Caroline Giroudon
- Hospices Civils de Lyon, Service de la Documentation Centrale, CEDEX, F-69424 Lyon, France;
| | - Mikail Nourredine
- Université de Lyon, Université Lyon 1, F-69003 Lyon, France; (S.L.Q.); (M.N.)
- Hospices Civils de Lyon, Service de Biostatistique et Bioinformatique, F-69003 Lyon, France
- CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR 5558, F-69100 Villeurbanne, France
| | - Emilie Bequignon
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Créteil, France; (S.B.); (A.C.); (B.L.); (E.B.)
- CNRS ERL 7000, F-94010 Créteil, France
- Service d’ORL, de Chirurgie Cervico Faciale, Centre Hospitalier Intercommunal de Créteil, F-94010 Créteil, France
| |
Collapse
|
36
|
Suzuki T, Hashii N, Tada M, Ishii-Watabe A. The influence of antibody engineering on Fc conformation and Fc receptor binding properties: Analysis of FcRn-binding engineered antibodies and an Fc fusion protein. MAbs 2021; 13:1923366. [PMID: 34030575 PMCID: PMC8158039 DOI: 10.1080/19420862.2021.1923366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Therapeutic immunoglobulin G (IgG) antibodies have comparatively long half-lives because the neonatal Fc receptor (FcRn) binds to the IgG Fc at acidic pH in the endosome and protects IgG from degradation. To further prolong the half-lives, amino acid-substituted antibodies having high affinity to FcRn are being developed, and one such therapeutic antibody (ravulizumab) has been approved. In this study, we investigated the binding property to FcγR and the conformation of seven FcRn affinity-modulated adalimumab variants to clarify the impact of the amino acid substitutions on the function and conformation of IgG Fc. The amino acid substitutions in T254-P261 caused a change in deuterium uptake into some regions of Fc in HDX-MS analysis, but those at T311, M432 and N438 did not cause such a change. The conformations around F245-L255 (FLFPPKPKDTL) were particularly influenced by the amino acid substitution in M256-P261, and the conformational changes of this region were correlated with the decrease of the affinity to FcγRIIIa. Additionally, we investigated the conformational difference of Fc between a Fc fusion protein (etanercept) and a native IgG (adalimumab). Although the Fc fusion proteins were expected to have similar FcRn affinity to IgGs, the affinity of etanercept to FcRn was lower than that of adalimumab, and its half-life was shorter than those of the IgG antibodies. Differences in deuterium uptakes were observed in the two regions where they were also detected in the adalimumab variants, and the conformational differences appeared to be an important factor for the low FcRn affinity of etanercept.
Collapse
Affiliation(s)
- Takuo Suzuki
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | - Noritaka Hashii
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | - Minoru Tada
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| |
Collapse
|
37
|
Ke C, Ma Y, Pan D, Wan Z, Feng T, Yu D, Liu X, Wang H, Du M, Huang L, Zhang Y, Du L, Wang X, Li K, Yu D, Zhang M, Huang J, Qu J, Ren L, Hu Y, Cao G, Hu X, Wu S, Han H, Zhao Y. FcRn is not the receptor mediating the transfer of serum IgG to colostrum in pigs. Immunology 2021; 163:448-459. [PMID: 33738807 DOI: 10.1111/imm.13328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/02/2023] Open
Abstract
In contrast to humans or rabbits, in which maternal IgG is transmitted to offspring prenatally via the placenta or the yolk sac, large domestic animals such as pigs, cows and sheep transmit IgG exclusively through colostrum feeding after delivery. The extremely high IgG content in colostrum is absorbed by newborns via the small intestine. Although it is widely accepted that the neonatal Fc receptor, FcRn, is the receptor mediating IgG transfer across both the placenta and small intestine, it remains unclear whether FcRn also mediates serum IgG transfer across the mammary barrier to colostrum/milk, especially in large domestic animals. In this study, using a FcRn knockout pig model generated with a CRISPR-Cas9-based approach, we clearly demonstrate that FcRn is not responsible for the IgG transfer from serum to colostrum in pigs, although like in other mammals, it is involved in IgG homeostasis and mediates IgG absorption in the small intestine of newborns.
Collapse
Affiliation(s)
- Cuncun Ke
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China.,Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Yonghe Ma
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Dengke Pan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Zihui Wan
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Tao Feng
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Dawei Yu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Xiaojuan Liu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Haitao Wang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Minjie Du
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Linhua Huang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yifu Zhang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lijuan Du
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Xifeng Wang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Kongpan Li
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Di Yu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Ming Zhang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Jinwei Huang
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Junwei Qu
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Liming Ren
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Yanzhong Hu
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Gengsheng Cao
- Henan Engineering Laboratory for Mammary Bioreactor, School of Life Science, Henan University, Kaifeng, China
| | - Xiaoxiang Hu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Sen Wu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Haitang Han
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Yaofeng Zhao
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| |
Collapse
|
38
|
Faid V, Leblanc Y, Berger M, Seifert A, Bihoreau N, Chevreux G. C-terminal lysine clipping of IgG1: impact on binding to human FcγRIIIa and neonatal Fc receptors. Eur J Pharm Sci 2021; 159:105730. [PMID: 33493670 DOI: 10.1016/j.ejps.2021.105730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 01/01/2023]
Abstract
Monoclonal antibodies (mAbs) display numerous structural attributes, some of them may impact their safety and/or efficacy profiles. C-terminal lysine clipping is a common phenomenon occurring during the bioproduction of mAbs and leads to variable amounts of final process-related charge variants. If Fc-glycosylation has been by far the most documented critical quality attribute (CQA), the potential impacts of mAb C-terminal lysine content is far less reported, particularly on the ability of these basic variants to bind human Fc receptors. To address this question, three charge variant species having zero (K0), one (K1) and two (K2) C-terminal lysine(s) were isolated with high purity from an in-house human IgG1 by preparative strong-cation exchange (SCX) chromatography. A comprehensive biophysical characterization of these three fractions was undertaken, demonstrating their high similarity in terms of structural homogeneity, with a particular attention paid on their respective N-glycosylation profiles. The binding affinity of the fractions to human FcγRIIIa-Val176 was assessed both by affinity chromatography and surface plasmon resonance (SPR), and to human neonatal Fc receptor (FcRn) by affinity chromatography. Results demonstrate that the three charge variants did not show any significant binding difference for the two tested human Fc receptors, translating certainly to comparable biological properties. As a consequence, C-terminal lysine clipping of the present therapeutic IgG1 should not impact both FcRn-dependent pharmacokinetic profiles and FcγRIIIa-driven cytotoxic activities. The methods used in this study can be widely applied to other IgG1 to define criticality of the C-terminal lysine clipping as a CQA.
Collapse
Affiliation(s)
- Valegh Faid
- Analytical Department, LFB Biotechnologies, 3 avenue des Tropiques, 91958 Courtaboeuf (Les Ulis), France.
| | - Yann Leblanc
- Analytical Department, LFB Biotechnologies, 3 avenue des Tropiques, 91958 Courtaboeuf (Les Ulis), France
| | - Marie Berger
- Analytical Department, LFB Biotechnologies, 3 avenue des Tropiques, 91958 Courtaboeuf (Les Ulis), France
| | - Alexander Seifert
- Analytical Department, LFB Biotechnologies, 3 avenue des Tropiques, 91958 Courtaboeuf (Les Ulis), France
| | - Nicolas Bihoreau
- Analytical Department, LFB Biotechnologies, 3 avenue des Tropiques, 91958 Courtaboeuf (Les Ulis), France
| | - Guillaume Chevreux
- Analytical Department, LFB Biotechnologies, 3 avenue des Tropiques, 91958 Courtaboeuf (Les Ulis), France
| |
Collapse
|
39
|
Qi T, Cao Y. In Translation: FcRn across the Therapeutic Spectrum. Int J Mol Sci 2021; 22:3048. [PMID: 33802650 PMCID: PMC8002405 DOI: 10.3390/ijms22063048] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
As an essential modulator of IgG disposition, the neonatal Fc receptor (FcRn) governs the pharmacokinetics and functions many therapeutic modalities. In this review, we thoroughly reexamine the hitherto elucidated biological and thermodynamic properties of FcRn to provide context for our assessment of more recent advances, which covers antigen-binding fragment (Fab) determinants of FcRn affinity, transgenic preclinical models, and FcRn targeting as an immune-complex (IC)-clearing strategy. We further comment on therapeutic antibodies authorized for treating SARS-CoV-2 (bamlanivimab, casirivimab, and imdevimab) and evaluate their potential to saturate FcRn-mediated recycling. Finally, we discuss modeling and simulation studies that probe the quantitative relationship between in vivo IgG persistence and in vitro FcRn binding, emphasizing the importance of endosomal transit parameters.
Collapse
Affiliation(s)
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA;
| |
Collapse
|
40
|
Hubbard JJ, Pyzik M, Rath T, Kozicky LK, Sand KMK, Gandhi AK, Grevys A, Foss S, Menzies SC, Glickman JN, Fiebiger E, Roopenian DC, Sandlie I, Andersen JT, Sly LM, Baker K, Blumberg RS. FcRn is a CD32a coreceptor that determines susceptibility to IgG immune complex-driven autoimmunity. J Exp Med 2021; 217:151942. [PMID: 32658257 PMCID: PMC7537387 DOI: 10.1084/jem.20200359] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022] Open
Abstract
IgG immune complexes (ICs) promote autoimmunity through binding fragment crystallizable (Fc) γ-receptors (FcγRs). Of these, the highly prevalent FcγRIIa (CD32a) histidine (H)-131 variant (CD32aH) is strongly linked to human autoimmune diseases through unclear mechanisms. We show that, relative to the CD32a arginine (R)-131 (CD32aR) variant, CD32aH more avidly bound human (h) IgG1 IC and formed a ternary complex with the neonatal Fc receptor (FcRn) under acidic conditions. In primary human and mouse cells, both CD32a variants required FcRn to induce innate and adaptive immune responses to hIgG1 ICs, which were augmented in the setting of CD32aH. Conversely, FcRn induced responses to IgG IC independently of classical FcγR, but optimal responses required FcRn and FcγR. Finally, FcRn blockade decreased inflammation in a rheumatoid arthritis model without reducing circulating autoantibody levels, providing support for FcRn’s direct role in IgG IC-associated inflammation. Thus, CD32a and FcRn coregulate IgG IC-mediated immunity in a manner favoring the CD32aH variant, providing a novel mechanism for its disease association.
Collapse
Affiliation(s)
- Jonathan J Hubbard
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Michal Pyzik
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Timo Rath
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lisa K Kozicky
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kine M K Sand
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Amit K Gandhi
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Algirdas Grevys
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stian Foss
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Susan C Menzies
- Division of Gastroenterology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan N Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Edda Fiebiger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Laura M Sly
- Division of Gastroenterology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kristi Baker
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Harvard Digestive Diseases Center, Boston, MA
| |
Collapse
|
41
|
Ko S, Jo M, Jung ST. Recent Achievements and Challenges in Prolonging the Serum Half-Lives of Therapeutic IgG Antibodies Through Fc Engineering. BioDrugs 2021; 35:147-157. [PMID: 33608823 PMCID: PMC7894971 DOI: 10.1007/s40259-021-00471-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2021] [Indexed: 01/02/2023]
Abstract
Association of FcRn molecules to the Fc region of IgG in acidified endosomes and subsequent dissociation of the interaction in neutral pH serum enables IgG molecules to be recycled for prolonged serum persistence after internalization by endothelial cells, rather than being degraded in the serum and in the lysosomes inside the cells. Exploiting this intracellular trafficking and recycling mechanism, many researchers have engineered the Fc region to further extend the serum half-lives of therapeutic antibodies by optimizing the pH-dependent IgG Fc-FcRn interaction, and have generated various Fc variants exhibiting significantly improved circulating half-lives of therapeutic IgG antibodies. In order to estimate pharmacokinetic profiles of IgG Fc variants in human serum, not only a variety of in vitro techniques to determine the equilibrium binding constants and instantaneous rate constants for pH-dependent FcRn binding, but also diverse in vivo animal models including wild-type mouse, human FcRn transgenic mouse (Tg32 and Tg276), humanized mouse (Scarlet), or cynomolgus monkey have been harnessed. Currently, multiple IgG Fc variants that have been validated for their prolonged therapeutic potency in preclinical models have been successfully entered into human clinical trials for cancer, infectious diseases, and autoimmune diseases.
Collapse
Affiliation(s)
- Sanghwan Ko
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.,Institute of Human Genetics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Migyeong Jo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea. .,Institute of Human Genetics, Korea University College of Medicine, Seoul, Republic of Korea. .,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea. .,Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea. .,Biomedical Research Center, Korea University Anam Hospital, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Sivaccumar J, Sandomenico A, Vitagliano L, Ruvo M. Monoclonal Antibodies: A Prospective and Retrospective View. Curr Med Chem 2021; 28:435-471. [PMID: 32072887 DOI: 10.2174/0929867327666200219142231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Monoclonal Antibodies (mAbs) represent one of the most important classes of biotherapeutic agents. They are used to cure many diseases, including cancer, autoimmune diseases, cardiovascular diseases, angiogenesis-related diseases and, more recently also haemophilia. They can be highly varied in terms of format, source, and specificity to improve efficacy and to obtain more targeted applications. This can be achieved by leaving substantially unchanged the basic structural components for paratope clustering. OBJECTIVES The objective was to trace the most relevant findings that have deserved prestigious awards over the years, to report the most important clinical applications and to emphasize their latest emerging therapeutic trends. RESULTS We report the most relevant milestones and new technologies adopted for antibody development. Recent efforts in generating new engineered antibody-based formats are briefly reviewed. The most important antibody-based molecules that are (or are going to be) used for pharmacological practice have been collected in useful tables. CONCLUSION The topics here discussed prove the undisputed role of mAbs as innovative biopharmaceuticals molecules and as vital components of targeted pharmacological therapies.
Collapse
Affiliation(s)
- Jwala Sivaccumar
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Luigi Vitagliano
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| |
Collapse
|
43
|
Klaus T, Deshmukh S. pH-responsive antibodies for therapeutic applications. J Biomed Sci 2021; 28:11. [PMID: 33482842 PMCID: PMC7821552 DOI: 10.1186/s12929-021-00709-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/15/2021] [Indexed: 11/29/2022] Open
Abstract
Therapeutic antibodies are instrumental in improving the treatment outcome for certain disease conditions. However, to enhance their efficacy and specificity, many efforts are continuously made. One of the approaches that are increasingly explored in this field are pH-responsive antibodies capable of binding target antigens in a pH-dependent manner. We reviewed suitability and examples of these antibodies that are functionally modulated by the tumor microenvironment. Provided in this review is an update about antigens targeted by pH-responsive, sweeping, and recycling antibodies. Applicability of the pH-responsive antibodies in the engineering of chimeric antigen receptor T-cells (CAR-T) and in improving drug delivery to the brain by the enhanced crossing of the blood-brain barrier is also discussed. The pH-responsive antibodies possess strong treatment potential. They emerge as next-generation programmable engineered biologic drugs that are active only within the targeted biological space. Thus, they are valuable in targeting acidified tumor microenvironment because of improved spatial persistence and reduced on-target off-tumor toxicities. We predict that the programmable pH-dependent antibodies become powerful tools in therapies of cancer.
Collapse
Affiliation(s)
- Tomasz Klaus
- Research and Development Department, Pure Biologics, Inc., Dunska 11, 54427, Wrocław, Poland
| | - Sameer Deshmukh
- Research and Development Department, Pure Biologics, Inc., Dunska 11, 54427, Wrocław, Poland.
| |
Collapse
|
44
|
Aaen KH, Anthi AK, Sandlie I, Nilsen J, Mester S, Andersen JT. The neonatal Fc receptor in mucosal immune regulation. Scand J Immunol 2021; 93:e13017. [PMID: 33351196 DOI: 10.1111/sji.13017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/11/2020] [Accepted: 12/21/2020] [Indexed: 01/01/2023]
Abstract
The neonatal Fc receptor (FcRn) was first recognized for its role in transfer of maternal IgG to the foetus or newborn, providing passive immunity early in life. However, it has become clear that the receptor is versatile, widely expressed and plays an indispensable role in both immunological and non-immunological processes throughout life. The receptor rescues immunoglobulin G (IgG) and albumin from intracellular degradation and shuttles the ligands across polarized cell barriers, in all cases via a pH-dependent binding-and-release mechanism. These processes secure distribution and high levels of both IgG and albumin throughout the body. At mucosal sites, FcRn transports IgG across polarized epithelial cells where it retrieves IgG in complex with luminal antigens that is delivered to tissue-localized immune cells. In dendritic cells (DCs), FcRn orchestrates processing of IgG-opsonized immune complexes (ICs) in concert with classical Fcγ receptors, which results in antigen presentation and cross-presentation of antigenic peptides on MHC class II and I to CD4+ and CD8+ T cells, respectively. Hence, FcRn regulates transport of the ligands within and across different types of cells, but also processing of IgG-ICs by immune cells. As such, the receptor is involved in immune surveillance and protection against infections. In this brief review, we highlight how FcRn expressed by hematopoietic and non-hematopoietic cells contributes to immune regulation at mucosal barriers-biology that can be utilized in development of biologics and subunit vaccines for non-invasive delivery.
Collapse
Affiliation(s)
- Kristin Hovden Aaen
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Aina Karen Anthi
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Inger Sandlie
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jeannette Nilsen
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Simone Mester
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
45
|
Wang Q, Hai W, Shi S, Peng J, Xu Y. Oral uptake and persistence of the FnAb-8 protein characterized by in situ radio-labeling and PET/CT imaging. Asian J Pharm Sci 2020; 15:752-758. [PMID: 33363630 PMCID: PMC7750799 DOI: 10.1016/j.ajps.2020.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 11/28/2022] Open
Abstract
The absorption of peptides and proteins delivered orally is minimum because of the intestine epithelial barrier. There are few known active transport mechanisms for macromolecules including the neonatal Fc Receptor (FcRn) for the absorption and secretion of IgGs in infant and adult intestine. We had previously described the FnAb-8 protein that could bind to hFcRn tightly at pH 6.0 but barely at pH 7.4. In this study, we examined its uptake, biodistribution and pharmacokinetics after peroral administration in both wild-type and human FcRn transgenic (Tg) mice. FnAb-8 was modified to contain trans-cyclooctene (TCO) which could interact with 18F labeled tetrazine in situ via the bioorthogonal inverse-electron-demand Diels−Alder reaction. We showed that FnAb-8 had a tendency to distribute and persist in the Tg mice intestine for an extended duration of time. It could also be absorbed into the circulation and distributed systemically over a long period of time up to 172 h. The improvement in oral uptake and concentration in the intestine tissue may be valuable for designing oral delivery of biopharmaceuticals, especially for diseases involving the gastric intestinal tissue.
Collapse
Affiliation(s)
- Qian Wang
- School of Pharmacy and Chemistry, DaLi University, Dali 671000, China
| | - Wangxi Hai
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sanyuan Shi
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinliang Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuhong Xu
- School of Pharmacy and Chemistry, DaLi University, Dali 671000, China
| |
Collapse
|
46
|
Liu R, Oldham RJ, Teal E, Beers SA, Cragg MS. Fc-Engineering for Modulated Effector Functions-Improving Antibodies for Cancer Treatment. Antibodies (Basel) 2020; 9:E64. [PMID: 33212886 PMCID: PMC7709126 DOI: 10.3390/antib9040064] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022] Open
Abstract
The majority of monoclonal antibody (mAb) therapeutics possess the ability to engage innate immune effectors through interactions mediated by their fragment crystallizable (Fc) domain. By delivering Fc-Fc gamma receptor (FcγR) and Fc-C1q interactions, mAb are able to link exquisite specificity to powerful cellular and complement-mediated effector functions. Fc interactions can also facilitate enhanced target clustering to evoke potent receptor signaling. These observations have driven decades-long research to delineate the properties within the Fc that elicit these various activities, identifying key amino acid residues and elucidating the important role of glycosylation. They have also fostered a growing interest in Fc-engineering whereby this knowledge is exploited to modulate Fc effector function to suit specific mechanisms of action and therapeutic purposes. In this review, we document the insight that has been generated through the study of the Fc domain; revealing the underpinning structure-function relationships and how the Fc has been engineered to produce an increasing number of antibodies that are appearing in the clinic with augmented abilities to treat cancer.
Collapse
Affiliation(s)
- Rena Liu
- GlaxoSmithKline Research and Development, Stevenage SG1 2NY, UK;
| | - Robert J. Oldham
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Emma Teal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| |
Collapse
|
47
|
Rossini S, Noé R, Daventure V, Lecerf M, Justesen S, Dimitrov JD. V Region of IgG Controls the Molecular Properties of the Binding Site for Neonatal Fc Receptor. THE JOURNAL OF IMMUNOLOGY 2020; 205:2850-2860. [PMID: 33077645 DOI: 10.4049/jimmunol.2000732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/17/2020] [Indexed: 01/09/2023]
Abstract
Neonatal Fc receptor (FcRn) has a key role in the homeostasis of IgG. Despite its physiological and clinical importance, the interaction of IgG and FcRn remains not completely comprehended. Thus, IgG molecules with identical constant portions but with minor differences in their V regions have been demonstrated to interact with FcRn with a considerable heterogeneity in the binding affinity. To understand this discrepancy, we dissected the physicochemical mechanism of the interaction of 10 human IgG1 to human FcRn. The interactions of two Abs in the presence of their cognate Ags were also examined. Data from activation and equilibrium thermodynamics analyses as well as pH dependence of the kinetics revealed that the V region of IgG could modulate a degree of conformational changes and binding energy of noncovalent contacts at the FcRn binding interface. These results suggest that the V domains modulate FcRn binding site in Fc by allosteric effects. These findings contribute for a deeper understanding of the mechanism of IgG-FcRn interaction. They might also be of relevance for rational engineering of Abs for optimizing their pharmacokinetic properties.
Collapse
Affiliation(s)
- Sofia Rossini
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; and
| | - Rémi Noé
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; and
| | - Victoria Daventure
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; and
| | - Maxime Lecerf
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; and
| | | | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; and
| |
Collapse
|
48
|
Pilati D, Howard KA. Albumin-based drug designs for pharmacokinetic modulation. Expert Opin Drug Metab Toxicol 2020; 16:783-795. [DOI: 10.1080/17425255.2020.1801633] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Diego Pilati
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C Denmark
| | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C Denmark
| |
Collapse
|
49
|
Leblanc Y, Berger M, Seifert A, Bihoreau N, Chevreux G. Human serum albumin presents isoform variants with altered neonatal Fc receptor interactions. Protein Sci 2020; 28:1982-1992. [PMID: 31583777 DOI: 10.1002/pro.3733] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 12/23/2022]
Abstract
Human serum albumin (HSA) is the most abundant protein in plasma and presents the particularity, with IgG, to have an extraordinary long serum half-life conferred by its interaction with the neonatal Fc receptor (FcRn). If the impact of IgG post-translational modifications (PTMs) on FcRn binding is well documented, it is far less reported for HSA despite numerous PTMs occurring on the protein in plasma. HSA is susceptible to numerous degradation reactions in plasma, because of aging, oxidative stress or liver and pancreas related pathologies. In the present study, we combined FcRn affinity chromatography and mass spectrometry to investigate the impact of HSA PTMs upon FcRn binding. This methodology presents the advantage to distinguish the effect of a single modification from a plasma HSA preparation made of a mixture of different isoforms. Cys34 oxidation, Lys525 glycation, and Leu585 C-terminal truncation, which are modifications related to several pathological conditions, were demonstrated to act negatively on HSA-FcRn interaction. The HSA-FcRn binding alteration generated by these modifications is consistent with their vicinity with the interaction interface of the two proteins. Results were discussed regarding altered half-life of HSA observed in several disease states and pave the way toward new understandings of the hypoalbuminemia pathogenesis. SIGNIFICANCE STATEMENT: In this study, we investigated the impact of several post-translational modifications of HSA toward its ability to bind to the neonatal Fc receptor using in vitro affinity chromatography, mass spectrometry, and surface plasmon resonance. Cys34 oxidation, Lys525 glycation, and Leu585 C-terminal truncation were demonstrated to decrease HSA-FcRn binding. These modifications occurring in circulating HSA were discussed in relation to several pathologies as well as for the use of HSA as a therapeutic protein.
Collapse
Affiliation(s)
- Yann Leblanc
- Analytical Department of LFB Biotechnologies, Courtabœuf, France
| | - Marie Berger
- Analytical Department of LFB Biotechnologies, Courtabœuf, France
| | | | - Nicolas Bihoreau
- Analytical Department of LFB Biotechnologies, Courtabœuf, France
| | | |
Collapse
|
50
|
Stutz C, Blein S. A single mutation increases heavy-chain heterodimer assembly of bispecific antibodies by inducing structural disorder in one homodimer species. J Biol Chem 2020; 295:9392-9408. [PMID: 32404368 PMCID: PMC7363136 DOI: 10.1074/jbc.ra119.012335] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/08/2020] [Indexed: 12/31/2022] Open
Abstract
We previously reported efficient heavy-chain assembly of heterodimeric bispecific antibodies by exchanging the interdomain protein interface of the human IgG1 CH3 dimer with the protein interface of the constant α and β domains of the human T-cell receptor, a technology known as bispecific engagement by antibodies based on the T-cell receptor (BEAT). Efficient heterodimerization in mammalian cell transient transfections was observed, but levels were influenced by the nature of the binding arms, particularly in the Fab-scFv-Fc format. In this study, we report a single amino acid change that significantly and consistently improved the heterodimerization rate of this format (≥95%) by inducing partial disorder in one homodimer species without affecting the heterodimer. Correct folding and assembly of the heterodimer were confirmed by the high-resolution (1.88-1.98 Å) crystal structure presented here. Thermal stability and 1-anilinonaphthalene-8-sulfonic acid-binding experiments, comparing original BEAT, mutated BEAT, and "knobs-into-holes" interfaces, suggested a cooperative assembly process of heavy chains in heterodimers. The observed gain in stability of the interfaces could be classified in the following rank order: mutated BEAT > original BEAT > knobs-into-holes. We therefore propose that the superior cooperativity found in BEAT interfaces is the key driver of their greater performance. Furthermore, we show how the mutated BEAT interface can be exploited for the routine preparation of drug candidates, with minimal risk of homodimer contamination using a single Protein A chromatography step.
Collapse
Affiliation(s)
- Cian Stutz
- Department of Antibody Engineering, Ichnos Sciences S.A., Biopôle Lausanne-Epalinges, Epalinges, Switzerland
| | - Stanislas Blein
- Department of Antibody Engineering, Ichnos Sciences S.A., Biopôle Lausanne-Epalinges, Epalinges, Switzerland
| |
Collapse
|