1
|
Foss S, Sakya SA, Aguinagalde L, Lustig M, Shaughnessy J, Cruz AR, Scheepmaker L, Mathiesen L, Ruso-Julve F, Anthi AK, Gjølberg TT, Mester S, Bern M, Evers M, Bratlie DB, Michaelsen TE, Schlothauer T, Sok D, Bhattacharya J, Leusen J, Valerius T, Ram S, Rooijakkers SHM, Sandlie I, Andersen JT. Human IgG Fc-engineering for enhanced plasma half-life, mucosal distribution and killing of cancer cells and bacteria. Nat Commun 2024; 15:2007. [PMID: 38453922 PMCID: PMC10920689 DOI: 10.1038/s41467-024-46321-9] [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: 04/07/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Monoclonal IgG antibodies constitute the fastest growing class of therapeutics. Thus, there is an intense interest to design more potent antibody formats, where long plasma half-life is a commercially competitive differentiator affecting dosing, frequency of administration and thereby potentially patient compliance. Here, we report on an Fc-engineered variant with three amino acid substitutions Q311R/M428E/N434W (REW), that enhances plasma half-life and mucosal distribution, as well as allows for needle-free delivery across respiratory epithelial barriers in human FcRn transgenic mice. In addition, the Fc-engineered variant improves on-target complement-mediated killing of cancer cells as well as both gram-positive and gram-negative bacteria. Hence, this versatile Fc technology should be broadly applicable in antibody design aiming for long-acting prophylactic or therapeutic interventions.
Collapse
Affiliation(s)
- Stian Foss
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Siri A Sakya
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Leire Aguinagalde
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marta Lustig
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Jutamas Shaughnessy
- Department of Medicine, Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ana Rita Cruz
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lisette Scheepmaker
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Line Mathiesen
- Department of Public Health, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fulgencio Ruso-Julve
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Aina Karen Anthi
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Torleif Tollefsrud Gjølberg
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Simone Mester
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Malin Bern
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Mitchell Evers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Diane B Bratlie
- Infection Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Terje E Michaelsen
- Infection Immunology, Norwegian Institute of Public Health, Oslo, Norway
- Department of Chemical Pharmacy, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Munich, Germany
| | - Devin Sok
- International AIDS Vaccine Initiative (IAVI), New York, NY, USA
| | - Jayanta Bhattacharya
- Antibody Translational Research Program, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Jeanette Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Thomas Valerius
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sanjay Ram
- Department of Medicine, Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Suzan H M Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Park SA, Lee Y, Hwang H, Lee JH, Kang YJ, Kim Y, Jin C, An HJ, Oh YJ, Hinterdorfer P, Kim E, Choi S, Ko K. Fc engineered anti-virus therapeutic human IgG 1 expressed in plants with altered binding to the neonatal Fc receptor. Biotechnol J 2024; 19:e2300552. [PMID: 38528347 DOI: 10.1002/biot.202300552] [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: 10/11/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 03/27/2024]
Abstract
Production of therapeutic monoclonal antibody (mAb) in transgenic plants has several advantages such as large-scale production and the absence of pathogenic animal contaminants. However, mAb with high mannose (HM) type glycans has shown a faster clearance compared to antibodies produced in animal cells. The neonatal Fc receptor (FcRn) regulates the persistence of immunoglobulin G (IgG) by the FcRn-mediated recycling pathway, which salvages IgG from lysosomal degradation within cells. In this study, Fc-engineering of antirabies virus therapeutic mAb SO57 with the endoplasmic reticulum (ER)-retention peptide signal (Lys-Asp-Glu-Leu; KDEL) (mAbpK SO57) in plant cell was conducted to enhance its binding activity to human neonatal Fc receptor (hFcRn), consequently improve its serum half-life. Enzyme-linked immunosorbent assay (ELISA) and Surface plasmon resonance assay showed altered binding affinity of the Fc region of three different mAbpK SO57 variants [M252Y/S254T/T256E (MST), M428L/N434S (MN), H433K/N434F (HN)] to hFcRn compared to wild type (WT) of mAbpK SO57. Molecular modeling data visualized the structural alterations in these mAbpK SO57. All of the mAbpK SO57 variants had HM type glycan structures similar to the WT mAbpK SO57. In addition, the neutralizing activity of the three variants against the rabies virus CVS-11 was effective as the WT mAbpK SO57. These results indicate that the binding affinity of mAbpK SO57 variants to hFcRn can be modified without alteration of N-glycan structure and neutralization activity. Taken together, this study suggests that Fc-engineering of antirabies virus mAb can be applied to enhance the efficacy of therapeutic mAbs in plant expression systems.
Collapse
Affiliation(s)
- Sol-Ah Park
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyunjoo Hwang
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Jeong Hwan Lee
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Yang Joo Kang
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Yerin Kim
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Caiquan Jin
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Yoo Jin Oh
- Department of Applied Experimental Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Peter Hinterdorfer
- Department of Applied Experimental Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Eunhye Kim
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sun Choi
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Kisung Ko
- Department of Medicine, BioSystems Design Lab, College of Medicine, Chung-Ang University, Dongjak-gu, Seoul, Republic of Korea
| |
Collapse
|
3
|
Damelang T, Brinkhaus M, van Osch TLJ, Schuurman J, Labrijn AF, Rispens T, Vidarsson G. Impact of structural modifications of IgG antibodies on effector functions. Front Immunol 2024; 14:1304365. [PMID: 38259472 PMCID: PMC10800522 DOI: 10.3389/fimmu.2023.1304365] [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: 09/29/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Immunoglobulin G (IgG) antibodies are a critical component of the adaptive immune system, binding to and neutralizing pathogens and other foreign substances. Recent advances in molecular antibody biology and structural protein engineering enabled the modification of IgG antibodies to enhance their therapeutic potential. This review summarizes recent progress in both natural and engineered structural modifications of IgG antibodies, including allotypic variation, glycosylation, Fc engineering, and Fc gamma receptor binding optimization. We discuss the functional consequences of these modifications to highlight their potential for therapeutical applications.
Collapse
Affiliation(s)
- Timon Damelang
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Maximilian Brinkhaus
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Thijs L. J. van Osch
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Janine Schuurman
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Aran F. Labrijn
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Theo Rispens
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
4
|
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: 59] [Impact Index Per Article: 59.0] [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
|
5
|
Rosenberg YJ, Ordonez T, Khanwalkar US, Barnette P, Pandey S, Backes IM, Otero CE, Goldberg BS, Crowley AR, Leib DA, Shapiro MB, Jiang X, Urban LA, Lees J, Hessell AJ, Permar S, Haigwood NL, Ackerman ME. Evidence for the Role of a Second Fc-Binding Receptor in Placental IgG Transfer in Nonhuman Primates. mBio 2023; 14:e0034123. [PMID: 36946726 PMCID: PMC10127586 DOI: 10.1128/mbio.00341-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.
Collapse
Affiliation(s)
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Iara M. Backes
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Claire E. Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | | | - Andrew R. Crowley
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - David A. Leib
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mariya B. Shapiro
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | | | | | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
6
|
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: 7] [Impact Index Per Article: 3.5] [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
|
7
|
Sand KMK, Gruber MM, Sandlie I, Mathiesen L, Andersen JT, Wadsack C. Contribution of the ex vivo placental perfusion model in understanding transplacental immunoglobulin G transfer. Placenta 2022; 127:77-87. [PMID: 35981406 DOI: 10.1016/j.placenta.2022.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/07/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The acquisition of humoral immunity in utero is essential for the fetus. The crucial protein, which is responsible for this part of immunity, is immunoglobulin-G (IgG). Immune functions of IgGs are mediated via the interaction of the crystallizable fragment (Fc) region of IgG with specific Fc γ receptors (FcγRs). However, an atypical FcγR, the neonatal Fc receptor (FcRn), is a key regulator of IgG transfer across the human placenta. During the last four decades ex vivo placental perfusion studies have contributed significantly to the study of mechanisms of IgG transfer across the multicellular placental barrier. METHOD A PubMed search was conducted by using specific keywords: placenta, perfusion and IgG to review manuscripts using human placental perfusion to study the transplacental transfer of IgG. Relevant studies found in reference lists of these manuscripts were also added to the review, and references were included that supported or gave nuance to the discussion of the mechanisms of IgG kinetics in the placenta. RESULTS AND DISCUSSION We found twenty publications on the study of transplacental transfer of IgG using human ex vivo placental perfusion, by research groups with partly different settings. This review summarizes knowledge about placental IgG transfer, with a strong focus on the contributions from ex vivo placental perfusion studies.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- Department of Biosciences, University of Oslo, 0371, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0424, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, 0424, Oslo, Norway
| | - Michael M Gruber
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036, Graz, Austria
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, 0371, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0424, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, 0424, Oslo, Norway
| | - Line Mathiesen
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0424, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, 0424, Oslo, Norway
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, 8036, Graz, Austria; BioTechMed-Graz, Austria
| |
Collapse
|
8
|
Molitoris BA, Sandoval RM, Yadav SPS, Wagner MC. Albumin Uptake and Processing by the Proximal Tubule: Physiologic, Pathologic and Therapeutic Implications. Physiol Rev 2022; 102:1625-1667. [PMID: 35378997 PMCID: PMC9255719 DOI: 10.1152/physrev.00014.2021] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
For nearly 50 years the proximal tubule (PT) has been known to reabsorb, process, and either catabolize or transcytose albumin from the glomerular filtrate. Innovative techniques and approaches have provided insights into these processes. Several genetic diseases, nonselective PT cell defects, chronic kidney disease (CKD), and acute PT injury lead to significant albuminuria, reaching nephrotic range. Albumin is also known to stimulate PT injury cascades. Thus, the mechanisms of albumin reabsorption, catabolism, and transcytosis are being reexamined with the use of techniques that allow for novel molecular and cellular discoveries. Megalin, a scavenger receptor, cubilin, amnionless, and Dab2 form a nonselective multireceptor complex that mediates albumin binding and uptake and directs proteins for lysosomal degradation after endocytosis. Albumin transcytosis is mediated by a pH-dependent binding affinity to the neonatal Fc receptor (FcRn) in the endosomal compartments. This reclamation pathway rescues albumin from urinary losses and cellular catabolism, extending its serum half-life. Albumin that has been altered by oxidation, glycation, or carbamylation or because of other bound ligands that do not bind to FcRn traffics to the lysosome. This molecular sorting mechanism reclaims physiological albumin and eliminates potentially toxic albumin. The clinical importance of PT albumin metabolism has also increased as albumin is now being used to bind therapeutic agents to extend their half-life and minimize filtration and kidney injury. The purpose of this review is to update and integrate evolving information regarding the reabsorption and processing of albumin by proximal tubule cells including discussion of genetic disorders and therapeutic considerations.
Collapse
Affiliation(s)
- Bruce A. Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Dept.of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Ruben M. Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Shiv Pratap S. Yadav
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Mark C. Wagner
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
9
|
Akbar R, Bashour H, Rawat P, Robert PA, Smorodina E, Cotet TS, Flem-Karlsen K, Frank R, Mehta BB, Vu MH, Zengin T, Gutierrez-Marcos J, Lund-Johansen F, Andersen JT, Greiff V. Progress and challenges for the machine learning-based design of fit-for-purpose monoclonal antibodies. MAbs 2022; 14:2008790. [PMID: 35293269 PMCID: PMC8928824 DOI: 10.1080/19420862.2021.2008790] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Although the therapeutic efficacy and commercial success of monoclonal antibodies (mAbs) are tremendous, the design and discovery of new candidates remain a time and cost-intensive endeavor. In this regard, progress in the generation of data describing antigen binding and developability, computational methodology, and artificial intelligence may pave the way for a new era of in silico on-demand immunotherapeutics design and discovery. Here, we argue that the main necessary machine learning (ML) components for an in silico mAb sequence generator are: understanding of the rules of mAb-antigen binding, capacity to modularly combine mAb design parameters, and algorithms for unconstrained parameter-driven in silico mAb sequence synthesis. We review the current progress toward the realization of these necessary components and discuss the challenges that must be overcome to allow the on-demand ML-based discovery and design of fit-for-purpose mAb therapeutic candidates.
Collapse
Affiliation(s)
- Rahmad Akbar
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Habib Bashour
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Puneet Rawat
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Philippe A. Robert
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Eva Smorodina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Russia
| | | | - Karine Flem-Karlsen
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Norway
| | - Robert Frank
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Brij Bhushan Mehta
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Mai Ha Vu
- Department of Linguistics and Scandinavian Studies, University of Oslo, Norway
| | - Talip Zengin
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Bioinformatics, Mugla Sitki Kocman University, Turkey
| | | | | | - Jan Terje Andersen
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo and Oslo University Hospital, Norway
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
10
|
Valente D, Mauriac C, Schmidt T, Focken I, Beninga J, Mackness B, Qiu H, Vicat P, Kandira A, Radošević K, Rao S, Darbyshire J, Kabiri M. Pharmacokinetics of novel Fc-engineered monoclonal and multispecific antibodies in cynomolgus monkeys and humanized FcRn transgenic mouse models. MAbs 2021; 12:1829337. [PMID: 33079615 PMCID: PMC7587234 DOI: 10.1080/19420862.2020.1829337] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Monoclonal antibodies (mAbs) are among the fastest growing and most effective therapies for myriad diseases. Multispecific antibodies are an emerging class of novel therapeutics that can target more than one tumor- or immune-associated modulators per molecule. The combination of different binding affinities and target classes, such as soluble or membrane-bound antigens, within multispecific antibodies confers unique pharmacokinetic (PK) properties. Numerous factors affect an antibody’s PK, with affinity to the neonatal Fc receptor (FcRn) a key determinant of half-life. Recent work has demonstrated the potential for humanized FcRn transgenic mice to predict the PK of mAbs in humans. However, such work has not been extended to multispecific antibodies. We engineered mAbs and multispecific antibodies with various Fc modifications to enhance antibody performance. PK analyses in humanized FcRn transgenic mouse (homozygous Tg32 and Tg276) and non-human primate (NHP) models showed that FcRn-binding mutations improved the plasma half-lives of the engineered mAbs and multispecific antibodies, while glycan engineering to eliminate effector function did not affect the PK compared with wild-type controls. Furthermore, results suggest that the homozygous Tg32 mouse model can replace NHP models to differentiate PK of variants during lead optimization, not only for wild-type mAbs but also for Fc-engineered mAbs and multispecific antibodies. This Tg32-mouse model would enable prediction of half-life and linear clearance of mAbs and multispecific antibodies in NHPs to guide the design of further pharmacology/safety studies in this species. The allometric exponent for clearance scaling from Tg32 mice to NHPs was estimated to be 0.91 for all antibodies.
Collapse
Affiliation(s)
| | | | | | - Ingo Focken
- Biologics Research, Sanofi , Frankfurt, Germany
| | | | - Brian Mackness
- Translational In vivo Models (TIM), Sanofi, Frankfurt, Germany
| | - Huawei Qiu
- Biologics, Xilio Therapeutics, Inc. Cambridge, US
| | | | | | | | - Srini Rao
- Translational In vivo Models (TIM), Sanofi, Cambridge, US
| | | | - Mostafa Kabiri
- Translational In vivo Models (TIM), Sanofi, Frankfurt, Germany
| |
Collapse
|
11
|
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
|
12
|
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:ijms22126475. [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] [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
- Correspondence: ; Tel.: +33-4-7266-6415
| | - 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
|
13
|
Aftimos P, Rolfo C, Rottey S, Barthélémy P, Borg C, Park K, Oh DY, Kim SW, De Jonge N, Hanssens V, Zwanenpoel K, Molthoff C, Vugts D, Dreier T, Verheesen P, van Dongen GA, Jacobs J, Van Rompaey L, Hultberg A, Michieli P, Pauwels P, Fung S, Thibault A, de Haard H, Leupin N, Awada A. The NHance ® Mutation-Equipped Anti-MET Antibody ARGX-111 Displays Increased Tissue Penetration and Anti-Tumor Activity in Advanced Cancer Patients. Biomedicines 2021; 9:biomedicines9060665. [PMID: 34200749 PMCID: PMC8229762 DOI: 10.3390/biomedicines9060665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of MET signaling has been implicated in tumorigenesis and metastasis. ARGX-111 combines complete blockade of this pathway with enhanced tumor cell killing and was investigated in 24 patients with MET-positive advanced cancers in a phase 1b study at four dose levels (0.3–10 mg/kg). ARGX-111 was well tolerated up to 3 mg/kg (MTD). Anti-tumor activity was observed in nearly half of the patients (46%) with a mean duration of treatment of 12 weeks. NHance® mutations in the Fc of ARGX-111 increased affinity for the neonatal Fc receptor (FcRn) at acidic pH, stimulating transcytosis across FcRn-expressing cells and radiolabeled ARGX-111 accumulated in lymphoid tissues, bone and liver, organs expressing FcRn at high levels in a biodistribution study using human FcRn transgenic mice. In line with this, we observed, in a patient with MET-amplified (>10 copies) gastric cancer, diminished metabolic activity in multiple metastatic lesions in lymphoid and bone tissues by 18F-FDG-PET/CT after two infusions with 0.3 mg/kg ARGX-111. When escalated to 1 mg/kg, a partial response was reached. Furthermore, decreased numbers of CTC (75%) possibly by the enhanced tumor cell killing witnessed the modes of action of the drug, warranting further clinical investigation of ARGX-111.
Collapse
Affiliation(s)
- Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (A.A.)
| | - Christian Rolfo
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | | | - Philippe Barthélémy
- Medical Oncology Unit, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
| | - Christophe Borg
- Medical Oncology Department, University Hospital of Besançon, CEDEX, 25000 Besançon, France;
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Do-Youn Oh
- Seoul National University Hospital, Seoul 03080, Korea;
| | - Sang-We Kim
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Natalie De Jonge
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Valérie Hanssens
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Karen Zwanenpoel
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | - Carla Molthoff
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Daniëlle Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Torsten Dreier
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
- AgomAb Therapeutics NV, 9000 Ghent, Belgium;
| | - Peter Verheesen
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Guus A.M.S. van Dongen
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Julie Jacobs
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Luc Van Rompaey
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Anna Hultberg
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Paolo Michieli
- AgomAb Therapeutics NV, 9000 Ghent, Belgium;
- Department of Oncology, University of Torino Medical School, 10124 Turin, Italy
| | - Patrick Pauwels
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | - Samson Fung
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Alain Thibault
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Hans de Haard
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Nicolas Leupin
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
- Correspondence: ; Tel.: +41-79-293-18-14
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (A.A.)
| |
Collapse
|
14
|
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
|
15
|
Kang SH, Lee CH. Development of Therapeutic Antibodies and Modulating the Characteristics of Therapeutic Antibodies to Maximize the Therapeutic Efficacy. BIOTECHNOL BIOPROC E 2021; 26:295-311. [PMID: 34220207 PMCID: PMC8236339 DOI: 10.1007/s12257-020-0181-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Monoclonal antibodies (mAb) have been used as therapeutic agents for various diseases, and immunoglobulin G (IgG) is mainly used among antibody isotypes due to its structural and functional properties. So far, regardless of the purpose of the therapeutic antibody, wildtype IgG has been mainly used, but recently, the engineered antibodies with various strategies according to the role of the therapeutic antibody have been used to maximize the therapeutic efficacy. In this review paper, first, the overall structural features and functional characteristics of antibody IgG, second, the old and new techniques for antibody discovery, and finally, several antibody engineering strategies for maximizing therapeutic efficacy according to the role of a therapeutic antibody will be introduced.
Collapse
Affiliation(s)
- Seung Hyun Kang
- grid.31501.360000 0004 0470 5905Department of Pharmacology, Seoul National University College of Medicine, Seoul, 03080 Korea
| | - Chang-Han Lee
- grid.31501.360000 0004 0470 5905Department of Pharmacology, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080 Korea ,Hongcheon, 25159 Korea ,grid.31501.360000 0004 0470 5905SNU Dementia Research Center, Seoul National University College of Medicine, Seoul, 03080 Korea
| |
Collapse
|
16
|
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
|
17
|
Van de Walle I, Silence K, Budding K, Van de Ven L, Dijkxhoorn K, de Zeeuw E, Yildiz C, Gabriels S, Percier JM, Wildemann J, Meeldijk J, Simons PJ, Boon L, Cox L, Holgate R, Urbanus R, Otten HG, Leusen JHW, Blanchetot C, de Haard H, Hack CE, Boross P. ARGX-117, a therapeutic complement inhibiting antibody targeting C2. J Allergy Clin Immunol 2020; 147:1420-1429.e7. [PMID: 32926878 PMCID: PMC7485568 DOI: 10.1016/j.jaci.2020.08.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
Abstract
Background Activation of the classical and lectin pathway of complement may contribute to tissue damage and organ dysfunction of antibody-mediated diseases and ischemia-reperfusion conditions. Complement factors are being considered as targets for therapeutic intervention. Objective We sought to characterize ARGX-117, a humanized inhibitory monoclonal antibody against complement C2. Methods The mode-of-action and binding characteristics of ARGX-117 were investigated in detail. Furthermore, its efficacy was analyzed in in vitro complement cytotoxicity assays. Finally, a pharmacokinetic/pharmacodynamic study was conducted in cynomolgus monkeys. Results Through binding to the Sushi-2 domain of C2, ARGX-117 prevents the formation of the C3 proconvertase and inhibits classical and lectin pathway activation upstream of C3 activation. As ARGX-117 does not inhibit the alternative pathway, it is expected not to affect the antimicrobial activity of this complement pathway. ARGX-117 prevents complement-mediated cytotoxicity in in vitro models for autoimmune hemolytic anemia and antibody-mediated rejection of organ transplants. ARGX-117 exhibits pH- and calcium-dependent target binding and is Fc-engineered to increase affinity at acidic pH to the neonatal Fc receptor, and to reduce effector functions. In cynomolgus monkeys, ARGX-117 dose-dependently reduces free C2 levels and classical pathway activity. A 2-dose regimen of 80 and 20 mg/kg separated by a week, resulted in profound reduction of classical pathway activity lasting for at least 7 weeks. Conclusions ARGX-117 is a promising new complement inhibitor that is uniquely positioned to target both the classical and lectin pathways while leaving the alternative pathway intact.
Collapse
Affiliation(s)
| | | | - Kevin Budding
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Kim Dijkxhoorn
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elisabeth de Zeeuw
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Cafer Yildiz
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | | | - Johanna Wildemann
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan Meeldijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | | | - Linda Cox
- Bioceros BV, Utrecht, The Netherlands
| | | | - Rolf Urbanus
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Henny G Otten
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | | | - C Erik Hack
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Prothix BV, Leiden, The Netherlands
| | - Peter Boross
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Prothix BV, Leiden, The Netherlands.
| |
Collapse
|
18
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020; 11:1986. [PMID: 32983137 PMCID: PMC7485114 DOI: 10.3389/fimmu.2020.01986] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage–derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
19
|
FcRn, but not FcγRs, drives maternal-fetal transplacental transport of human IgG antibodies. Proc Natl Acad Sci U S A 2020; 117:12943-12951. [PMID: 32461366 DOI: 10.1073/pnas.2004325117] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The IgG Fc domain has the capacity to interact with diverse types of receptors, including the neonatal Fc receptor (FcRn) and Fcγ receptors (FcγRs), which confer pleiotropic biological activities. Whereas FcRn regulates IgG epithelial transport and recycling, Fc effector activities, such as antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis, are mediated by FcγRs, which upon cross-linking transduce signals that modulate the function of effector leukocytes. Despite the well-defined and nonoverlapping functional properties of FcRn and FcγRs, recent studies have suggested that FcγRs mediate transplacental IgG transport, as certain Fc glycoforms were reported to be enriched in fetal circulation. To determine the contribution of FcγRs and FcRn to the maternal-fetal transport of IgG, we characterized the IgG Fc glycosylation in paired maternal-fetal samples from patient cohorts from Uganda and Nicaragua. No differences in IgG1 Fc glycan profiles and minimal differences in IgG2 Fc glycans were noted, whereas the presence or absence of galactose on the Fc glycan of IgG1 did not alter FcγRIIIa or FcRn binding, half-life, or their ability to deplete target cells in FcγR/FcRn humanized mice. Modeling maternal-fetal transport in FcγR/FcRn humanized mice confirmed that only FcRn contributed to transplacental transport of IgG; IgG selectively enhanced for FcRn binding resulted in enhanced accumulation of maternal antibody in the fetus. In contrast, enhancing FcγRIIIa binding did not result in enhanced maternal-fetal transport. These results argue against a role for FcγRs in IgG transplacental transport, suggesting Fc engineering of maternally administered antibody to enhance only FcRn binding as a means to improve maternal-fetal transport of IgG.
Collapse
|
20
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020. [PMID: 32983137 DOI: 10.3389/fimmu.2020.01986/bibtex] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage-derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
21
|
Lee CH, Kang TH, Godon O, Watanabe M, Delidakis G, Gillis CM, Sterlin D, Hardy D, Cogné M, Macdonald LE, Murphy AJ, Tu N, Lee J, McDaniel JR, Makowski E, Tessier PM, Meyer AS, Bruhns P, Georgiou G. An engineered human Fc domain that behaves like a pH-toggle switch for ultra-long circulation persistence. Nat Commun 2019; 10:5031. [PMID: 31695028 PMCID: PMC6834678 DOI: 10.1038/s41467-019-13108-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
The pharmacokinetic properties of antibodies are largely dictated by the pH-dependent binding of the IgG fragment crystallizable (Fc) domain to the human neonatal Fc receptor (hFcRn). Engineered Fc domains that confer a longer circulation half-life by virtue of more favorable pH-dependent binding to hFcRn are of great therapeutic interest. Here we developed a pH Toggle switch Fc variant containing the L309D/Q311H/N434S (DHS) substitutions, which exhibits markedly improved pharmacokinetics relative to both native IgG1 and widely used half-life extension variants, both in conventional hFcRn transgenic mice and in new knock-in mouse strains. engineered specifically to recapitulate all the key processes relevant to human antibody persistence in circulation, namely: (i) physiological expression of hFcRn, (ii) the impact of hFcγRs on antibody clearance and (iii) the role of competing endogenous IgG. DHS-IgG retains intact effector functions, which are important for the clearance of target pathogenic cells and also has favorable developability.
Collapse
Affiliation(s)
- Chang-Han Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Tae Hyun Kang
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Department of Applied Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ophélie Godon
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France
| | - Makiko Watanabe
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Caitlin M Gillis
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France
| | - Delphine Sterlin
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France
| | - David Hardy
- Experimental Neuropathology Unit, Infection and Epidemiology Department, Institut Pasteur, 25, rue du Docteur Roux, 75015, Paris, France
| | | | | | | | - Naxin Tu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Jonathan R McDaniel
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Emily Makowski
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, USA
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France.
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA.
- Department of Molecular Bioscience, University of Texas at Austin, Austin, TX, USA.
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
22
|
Mackness BC, Jaworski JA, Boudanova E, Park A, Valente D, Mauriac C, Pasquier O, Schmidt T, Kabiri M, Kandira A, Radošević K, Qiu H. Antibody Fc engineering for enhanced neonatal Fc receptor binding and prolonged circulation half-life. MAbs 2019; 11:1276-1288. [PMID: 31216930 PMCID: PMC6748615 DOI: 10.1080/19420862.2019.1633883] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The neonatal Fc receptor (FcRn) promotes antibody recycling through rescue from normal lysosomal degradation. The binding interaction is pH-dependent with high affinity at low pH, but not under physiological pH conditions. Here, we combined rational design and saturation mutagenesis to generate novel antibody variants with prolonged half-life and acceptable development profiles. First, a panel of saturation point mutations was created at 11 key FcRn-interacting sites on the Fc region of an antibody. Multiple variants with slower FcRn dissociation kinetics than the wildtype (WT) antibody at pH 6.0 were successfully identified. The mutations were further combined and characterized for pH-dependent FcRn binding properties, thermal stability and the FcγRIIIa and rheumatoid factor binding. The most promising variants, YD (M252Y/T256D), DQ (T256D/T307Q) and DW (T256D/T307W), exhibited significantly improved binding to FcRn at pH 6.0 and retained similar binding properties as WT at pH 7.4. The pharmacokinetics in human FcRn transgenic mice and cynomolgus monkeys demonstrated that these properties translated to significantly prolonged plasma elimination half-life compared to the WT control. The novel variants exhibited thermal stability and binding to FcγRIIIa in the range comparable to clinically validated YTE and LS variants, and showed no enhanced binding to rheumatoid factor compared to the WT control. These engineered Fc mutants are promising new variants that are widely applicable to therapeutic antibodies, to extend their circulation half-life with obvious benefits of increased efficacy, and reduced dose and administration frequency.
Collapse
Affiliation(s)
| | | | | | - Anna Park
- Biologics Research, Sanofi , Framingham , MA , USA
| | | | | | | | | | | | | | | | - Huawei Qiu
- Biologics Research, Sanofi , Framingham , MA , USA
| |
Collapse
|
23
|
Pyzik M, Sand KMK, Hubbard JJ, Andersen JT, Sandlie I, Blumberg RS. The Neonatal Fc Receptor (FcRn): A Misnomer? Front Immunol 2019; 10:1540. [PMID: 31354709 PMCID: PMC6636548 DOI: 10.3389/fimmu.2019.01540] [Citation(s) in RCA: 271] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antibodies are essential components of an adaptive immune response. Immunoglobulin G (IgG) is the most common type of antibody found in circulation and extracellular fluids. Although IgG alone can directly protect the body from infection through the activities of its antigen binding region, the majority of IgG immune functions are mediated via proteins and receptors expressed by specialized cell subsets that bind to the fragment crystallizable (Fc) region of IgG. Fc gamma (γ) receptors (FcγR) belong to a broad family of proteins that presently include classical membrane-bound surface receptors as well as atypical intracellular receptors and cytoplasmic glycoproteins. Among the atypical FcγRs, the neonatal Fc receptor (FcRn) has increasingly gained notoriety given its intimate influence on IgG biology and its ability to also bind to albumin. FcRn functions as a recycling or transcytosis receptor that is responsible for maintaining IgG and albumin in the circulation, and bidirectionally transporting these two ligands across polarized cellular barriers. More recently, it has been appreciated that FcRn acts as an immune receptor by interacting with and facilitating antigen presentation of peptides derived from IgG immune complexes (IC). Here we review FcRn biology and focus on newer advances including how emerging FcRn-targeted therapies may affect the immune responses to IgG and IgG IC.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Kine M K Sand
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jonathan J Hubbard
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Digestive Diseases Center, Boston, MA, United States
| |
Collapse
|
24
|
Buse E, Markert UR. The immunology of the macaque placenta: A detailed analysis and critical comparison with the human placenta. Crit Rev Clin Lab Sci 2019; 56:118-145. [PMID: 30632863 DOI: 10.1080/10408363.2018.1538200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The cynomolgus monkey is increasingly considered in toxicological research as the most appropriate model for humans due to the species' close physiological contiguity, including reproductive physiology. Here, literature on the cynomolgus monkey placenta is reviewed in regards to its similarity to the human placenta and particularly for its immunological role, which is not entirely mirrored in humans. Pertinent original data are included in this article. The cynomolgus monkey placenta is evaluated based on three aspects: first, morphological development; second, the spatial and temporal appearance of maternal and fetal immune cells and certain immune cell products of the innate and adaptive immune systems; and third, the expression of relevant immune tolerance-related molecules including the homologs of anti-human leucocyte antigen, indoleamine 2,3-dioxygenase, FAS/FAS-L, annexin II, and progesterone. Parameters relevant to the immunological role of the placenta are evaluated from the immunologically immature stage of gestational day (GD) 50 until more mature stages close to birth. Selected comparisons are drawn with human and other laboratory animal placentas. In conclusion, the cynomolgus monkey placenta has a high degree of morphological and physiological similarity to the human placenta. However, there are differences in the topographical distribution of cell types and immune tolerance-related molecules. Three basic features are recognized: (1) the immunological capacity of the placenta changes throughout the lifetime of the organ; (2) these immunological changes include multiple parameters such as morphological adaptations, cell type involvement, and changes in immune-relevant molecule expression; and (3) the immune systems of two genetically disparate individuals (mother and child) are functionally intertwined at the maternal-fetal interface.
Collapse
Affiliation(s)
| | - Udo R Markert
- b Placenta Lab, Department of Obstetrics , University Hospital Jena , Jena , Germany
| |
Collapse
|
25
|
Velmurugan R, Ramakrishnan S, Kim M, Ober RJ, Ward ES. Phagocytosis of antibody-opsonized tumor cells leads to the formation of a discrete vacuolar compartment in macrophages. Traffic 2019; 19:273-284. [PMID: 29437282 PMCID: PMC5869154 DOI: 10.1111/tra.12552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 01/26/2018] [Accepted: 01/26/2018] [Indexed: 12/17/2022]
Abstract
Despite the rapidly expanding use of antibody‐based therapeutics to treat cancer, knowledge of the cellular processes following phagocytosis of antibody‐opsonized tumor cells is limited. Here we report the formation of a phagosome‐associated vacuole that is observed in macrophages as these degradative compartments mature following phagocytosis of HER2‐positive cancer cells in the presence of the HER2‐specific antibody, trastuzumab. We demonstrate that this vacuole is a distinct organelle that is closely apposed to the phagosome. Furthermore, the size of the phagosome‐associated vacuole is increased by inhibition of the mTOR pathway. Collectively, the identification of this vacuolar compartment has implications for understanding the subcellular trafficking processes leading to the destruction of phagocytosed, antibody‐opsonized cancer cells by macrophages.
Collapse
Affiliation(s)
- Ramraj Velmurugan
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas.,Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sreevidhya Ramakrishnan
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Mingin Kim
- Medical Science Graduate Program, Texas A&M University Health Science Center, College Station, Texas
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
26
|
Improvement of pharmacokinetic properties of therapeutic antibodies by antibody engineering. Drug Metab Pharmacokinet 2018; 34:25-41. [PMID: 30472066 DOI: 10.1016/j.dmpk.2018.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023]
Abstract
Monoclonal antibodies (mAbs) have become an important therapeutic option for several diseases. Since several mAbs have shown promising efficacy in clinic, the competition to develop mAbs has become severe. In efforts to gain a competitive advantage over other mAbs and provide significant benefits to patients, innovations in antibody engineering have aimed at improving the pharmacokinetic properties of mAbs. Because engineering can provide therapeutics that are more convenient, safer, and more efficacious for patients in several disease areas, it is an attractive approach to provide significant benefits to patients. Further advances in engineering mAbs to modulate their pharmacokinetics were driven by the increase of total soluble target antigen concentration that is often observed after injecting a mAb, which then requires a high dosage to antagonize. To decrease the required dosage, several antibody engineering techniques have been invented that reduce the total concentration of soluble target antigen. Here, we review the various ways that antibody engineering can improve the pharmacokinetic properties of mAbs.
Collapse
|
27
|
Targeting FcRn to Generate Antibody-Based Therapeutics. Trends Pharmacol Sci 2018; 39:892-904. [PMID: 30143244 DOI: 10.1016/j.tips.2018.07.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 01/01/2023]
Abstract
The MHC class I-related receptor FcRn serves multiple roles ranging from the regulation of levels of IgG isotype antibodies and albumin throughout the body to the delivery of antigen into antigen loading compartments in specialized antigen-presenting cells. In parallel with studies directed towards understanding FcRn at the molecular and cellular levels, there has been an enormous expansion in the development of engineering strategies involving FcRn to modulate the dynamic behavior of antibodies, antigens, and albumin. In this review article, we focus on a discussion of FcRn-targeted approaches that have resulted in the production of novel antibody-based platforms with considerable potential for use in the clinic.
Collapse
|
28
|
Ulrichts P, Guglietta A, Dreier T, van Bragt T, Hanssens V, Hofman E, Vankerckhoven B, Verheesen P, Ongenae N, Lykhopiy V, Enriquez FJ, Cho J, Ober RJ, Ward ES, de Haard H, Leupin N. Neonatal Fc receptor antagonist efgartigimod safely and sustainably reduces IgGs in humans. J Clin Invest 2018; 128:4372-4386. [PMID: 30040076 DOI: 10.1172/jci97911] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/03/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Intravenous Ig (IVIg), plasma exchange, and immunoadsorption are frequently used in the management of severe autoimmune diseases mediated by pathogenic IgG autoantibodies. These approaches modulating IgG levels can, however, be associated with some severe adverse reactions and a substantial burden to patients. Targeting the neonatal Fc receptor (FcRn) presents an innovative and potentially more effective, safer, and more convenient alternative for clearing pathogenic IgGs. METHODS A randomized, double-blind, placebo-controlled first-in-human study was conducted in 62 healthy volunteers to explore single and multiple ascending intravenous doses of the FcRn antagonist efgartigimod. The study objectives were to assess safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity. The findings of this study were compared with the pharmacodynamics profile elicited by efgartigimod in cynomolgus monkeys. RESULTS Efgartigimod treatment resulted in a rapid and specific clearance of serum IgG levels in both cynomolgus monkeys and healthy volunteers. In humans, single administration of efgartigimod reduced IgG levels up to 50%, while multiple dosing further lowered IgGs on average by 75% of baseline levels. Approximately 8 weeks following the last administration, IgG levels returned to baseline. Efgartigimod did not alter the homeostasis of albumin or Igs other than IgG, and no serious adverse events related to efgartigimod infusion were observed. CONCLUSION Antagonizing FcRn using efgartigimod is safe and results in a specific, profound, and sustained reduction of serum IgG levels. These results warrant further evaluation of this therapeutic approach in IgG-driven autoimmune diseases. TRIAL REGISTRATION Clinicaltrials.gov NCT03457649. FUNDING argenx BVBA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - JunHaeng Cho
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | | | | |
Collapse
|
29
|
Grevys A, Nilsen J, Sand KMK, Daba MB, Øynebråten I, Bern M, McAdam MB, Foss S, Schlothauer T, Michaelsen TE, Christianson GJ, Roopenian DC, Blumberg RS, Sandlie I, Andersen JT. A human endothelial cell-based recycling assay for screening of FcRn targeted molecules. Nat Commun 2018; 9:621. [PMID: 29434196 PMCID: PMC5809500 DOI: 10.1038/s41467-018-03061-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Albumin and IgG have remarkably long serum half-lives due to pH-dependent FcRn-mediated cellular recycling that rescues both ligands from intracellular degradation. Furthermore, increase in half-lives of IgG and albumin-based therapeutics has the potential to improve their efficacies, but there is a great need for robust methods for screening of relative FcRn-dependent recycling ability. Here, we report on a novel human endothelial cell-based recycling assay (HERA) that can be used for such pre-clinical screening. In HERA, rescue from degradation depends on FcRn, and engineered ligands are recycled in a manner that correlates with their half-lives in human FcRn transgenic mice. Thus, HERA is a novel cellular assay that can be used to predict how FcRn-binding proteins are rescued from intracellular degradation.
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, N-0450 Oslo, Norway
| | - Kine M K Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Muluneh B Daba
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Inger Øynebråten
- Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, N-0424, Oslo, Norway
| | - Malin Bern
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Martin B McAdam
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Stian Foss
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Tilman Schlothauer
- Biochemical and Analytical Research, Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center, DE-82377 Munich, Germany
| | - Terje E Michaelsen
- School of Pharmacy, University of Oslo, N-0371, Oslo, Norway.,Norwegian Institute of Public Health, Infection Immunology, N-0403, Oslo, Norway
| | | | | | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway.,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway
| | - Jan Terje Andersen
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316, Oslo, Norway. .,CIR and Department of Immunology, Rikshospitalet, Oslo University Hospital and University of Oslo, PO Box 4950, N-0424, Oslo, Norway. .,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, N-0424, Oslo, Norway.
| |
Collapse
|
30
|
Yang D, Kroe-Barrett R, Singh S, Roberts CJ, Laue TM. IgG cooperativity - Is there allostery? Implications for antibody functions and therapeutic antibody development. MAbs 2017; 9:1231-1252. [PMID: 28812955 PMCID: PMC5680800 DOI: 10.1080/19420862.2017.1367074] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A central dogma in immunology is that an antibody's in vivo functionality is mediated by 2 independent events: antigen binding by the variable (V) region, followed by effector activation by the constant (C) region. However, this view has recently been challenged by reports suggesting allostery exists between the 2 regions, triggered by conformational changes or configurational differences. The possibility of allosteric signals propagating through the IgG domains complicates our understanding of the antibody structure-function relationship, and challenges the current subclass selection process in therapeutic antibody design. Here we review the types of cooperativity in IgG molecules by examining evidence for and against allosteric cooperativity in both Fab and Fc domains and the characteristics of associative cooperativity in effector system activation. We investigate the origin and the mechanism of allostery with an emphasis on the C-region-mediated effects on both V and C region interactions, and discuss its implications in biological functions. While available research does not support the existence of antigen-induced conformational allosteric cooperativity in IgGs, there is substantial evidence for configurational allostery due to glycosylation and sequence variations.
Collapse
Affiliation(s)
- Danlin Yang
- a Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Rachel Kroe-Barrett
- a Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield , Connecticut , USA
| | - Sanjaya Singh
- b Janssen BioTherapeutics, Janssen Research & Development, LLC, Spring House , Pennsylvania , USA
| | - Christopher J Roberts
- c Department of Chemical and Biomolecular Engineering , University of Delaware , Newark , Delaware , USA
| | - Thomas M Laue
- d Department of Molecular, Cellular, and Biomedical Sciences , University of New Hampshire , Durham , New Hampshire , USA
| |
Collapse
|
31
|
Devanaboyina SC, Khare P, Challa DK, Ober RJ, Ward ES. Engineered clearing agents for the selective depletion of antigen-specific antibodies. Nat Commun 2017; 8:15314. [PMID: 28561044 PMCID: PMC5460014 DOI: 10.1038/ncomms15314] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/20/2017] [Indexed: 12/03/2022] Open
Abstract
Here we have designed a novel class of engineered antibody-based reagents (‘Seldegs') that induce the selective degradation of antigen-specific antibodies. We demonstrate the rapid and specific clearance of antibodies recognizing the autoantigen, myelin oligodendrocyte glycoprotein and tumour target, HER2. Seldegs have considerable potential in multiple areas, including the treatment of antibody-mediated autoimmunity and diagnostic imaging. The depletion of antigen-specific, deleterious antibodies during therapy and diagnosis remains an unsolved challenge. Here the authors develop ‘Seldegs', antigens linked to human Fc fragments with high affinity for the neonatal Fc receptor FcRn, to deplete MOG- and HER2-specific antibodies in mice.
Collapse
Affiliation(s)
- Siva Charan Devanaboyina
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, 469 Joe H. Reynolds Medical Sciences Building, 1114 TAMU, College Station, Texas 77843, USA.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 3107 Medical Research &Education Building, 8447 State Highway 47, Bryan, Texas 77807, USA
| | - Priyanka Khare
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, 469 Joe H. Reynolds Medical Sciences Building, 1114 TAMU, College Station, Texas 77843, USA.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 3107 Medical Research &Education Building, 8447 State Highway 47, Bryan, Texas 77807, USA
| | - Dilip K Challa
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, 469 Joe H. Reynolds Medical Sciences Building, 1114 TAMU, College Station, Texas 77843, USA.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 3107 Medical Research &Education Building, 8447 State Highway 47, Bryan, Texas 77807, USA
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, 469 Joe H. Reynolds Medical Sciences Building, 1114 TAMU, College Station, Texas 77843, USA.,Department of Biomedical Engineering, Texas A&M University, 5045 Emerging Technologies Building, 3120 TAMU, College Station, Texas 77843, USA
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, 469 Joe H. Reynolds Medical Sciences Building, 1114 TAMU, College Station, Texas 77843, USA.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 3107 Medical Research &Education Building, 8447 State Highway 47, Bryan, Texas 77807, USA
| |
Collapse
|
32
|
Jaramillo CAC, Belli S, Cascais AC, Dudal S, Edelmann MR, Haak M, Brun ME, Otteneder MB, Ullah M, Funk C, Schuler F, Simon S. Toward in vitro-to-in vivo translation of monoclonal antibody pharmacokinetics: Application of a neonatal Fc receptor-mediated transcytosis assay to understand the interplaying clearance mechanisms. MAbs 2017; 9:781-791. [PMID: 28440708 DOI: 10.1080/19420862.2017.1320008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) are a rapidly growing drug class for which great efforts have been made to optimize certain molecular features to achieve the desired pharmacokinetic (PK) properties. One approach is to engineer the interactions of the mAb with the neonatal Fc receptor (FcRn) by introducing specific amino acid sequence mutations, and to assess their effect on the PK profile with in vivo studies. Indeed, FcRn protects mAbs from intracellular degradation, thereby prolongs antibody circulation time in plasma and modulates its systemic clearance. To allow more efficient and focused mAb optimization, in vitro input that helps to identify and quantitatively predict the contribution of different processes driving non-target mediated mAb clearance in vivo and supporting translational PK modeling activities is essential. With this aim, we evaluated the applicability and in vivo-relevance of an in vitro cellular FcRn-mediated transcytosis assay to explain the PK behavior of 25 mAbs in rat or monkey. The assay was able to capture species-specific differences in IgG-FcRn interactions and overall correctly ranked Fc mutants according to their in vivo clearance. However, it could not explain the PK behavior of all tested IgGs, indicating that mAb disposition in vivo is a complex interplay of additional processes besides the FcRn interaction. Overall, the transcytosis assay was considered suitable to rank mAb candidates for their FcRn-mediated clearance component before extensive in vivo testing, and represents a first step toward a multi-factorial in vivo clearance prediction approach based on in vitro data.
Collapse
Affiliation(s)
| | - Sara Belli
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Anne-Christine Cascais
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | | | - Martin R Edelmann
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Markus Haak
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Marie-Elise Brun
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Michael B Otteneder
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Mohammed Ullah
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Christoph Funk
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Franz Schuler
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| | - Silke Simon
- a Roche Pharmaceutical Research and Early Development, Roche Innovation Center , Basel , Switzerland
| |
Collapse
|
33
|
Saxena A, Wu D. Advances in Therapeutic Fc Engineering - Modulation of IgG-Associated Effector Functions and Serum Half-life. Front Immunol 2016; 7:580. [PMID: 28018347 PMCID: PMC5149539 DOI: 10.3389/fimmu.2016.00580] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/24/2016] [Indexed: 12/20/2022] Open
Abstract
Today, monoclonal immunoglobulin gamma (IgG) antibodies have become a major option in cancer therapy especially for the patients with advanced or metastatic cancers. Efficacy of monoclonal antibodies (mAbs) is achieved through both its antigen-binding fragment (Fab) and crystallizable fragment (Fc). Fab can specifically recognize tumor-associated antigen (TAA) and thus modulate TAA-linked downstream signaling pathways that may lead to the inhibition of tumor growth, induction of tumor apoptosis, and differentiation. The Fc region can further improve mAbs’ efficacy by mediating effector functions such as antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cell-mediated phagocytosis. Moreover, Fc is the region interacting with the neonatal Fc receptor in a pH-dependent manner that can slow down IgG’s degradation and extend its serum half-life. Loss of the antibody Fc region dramatically shortens its serum half-life and weakens its anticancer effects. Given the essential roles that the Fc region plays in the modulation of the efficacy of mAb in cancer treatment, Fc engineering has been extensively studied in the past years. This review focuses on the recent advances in therapeutic Fc engineering that modulates its related effector functions and serum half-life. We also discuss the progress made in aglycosylated mAb development that may substantially reduce the cost of manufacture but maintain similar efficacies as conventional glycosylated mAb. Finally, we highlight several Fc engineering-based mAbs under clinical trials.
Collapse
Affiliation(s)
- Abhishek Saxena
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai , China
| | - Donghui Wu
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai , China
| |
Collapse
|
34
|
Challa DK, Mi W, Lo ST, Ober RJ, Ward ES. Antigen dynamics govern the induction of CD4 + T cell tolerance during autoimmunity. J Autoimmun 2016; 72:84-94. [DOI: 10.1016/j.jaut.2016.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 11/16/2022]
|
35
|
Souders CA, Nelson SC, Wang Y, Crowley AR, Klempner MS, Thomas W. A novel in vitro assay to predict neonatal Fc receptor-mediated human IgG half-life. MAbs 2016; 7:912-21. [PMID: 26018774 PMCID: PMC4622054 DOI: 10.1080/19420862.2015.1054585] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Immunoglobulin G (IgG) has an unusually long serum half-life in comparison to proteins of a similar size. It is well-known that this phenomenon is due to IgG's ability to bind the neonatal Fc receptor (FcRn) in a pH-dependent manner. FcRn binding properties can vary among IgGs, resulting in altered in vivo half-lives, and therefore it would be beneficial to accurately predict the FcRn binding properties of therapeutic IgG monoclonal antibodies (mAbs). Here we describe the development of an in vitro model capable of predicting the in vivo half-life of human IgG. Using a high-throughput biolayer interferometry (BLI) platform, the human FcRn association rate at acidic pH and subsequent dissociation rate at physiological pH was determined for 5 human IgG1 mAbs. Comparing the combined FcRn association and dissociation rates to the Phase 1 clinical study half-lives of the mAbs resulted in a strong correlation. The correlation was also verified in vivo using mice transgenic for human FcRn. The model was used to characterize various factors that may influence FcRn-mAb binding, including mAb variable region sequence differences and constant region glycosylation patterns. Results indicated that the complementarity-determining regions of the heavy chain significantly influence the mAb's FcRn binding properties, while the absence of glycosylation does not alter mAb-FcRn binding. Development of this high-throughput FcRn binding model could potentially predict the half-life of therapeutic IgGs and aid in selection of lead candidates while also serving as a screening tool for the development of mAbs with desired pharmacokinetic properties.
Collapse
Affiliation(s)
- Colby A Souders
- a MassBiologics of the University of Massachusetts Medical School ; Boston , MA USA
| | | | | | | | | | | |
Collapse
|
36
|
Martins JP, Kennedy PJ, Santos HA, Barrias C, Sarmento B. A comprehensive review of the neonatal Fc receptor and its application in drug delivery. Pharmacol Ther 2016; 161:22-39. [PMID: 27016466 DOI: 10.1016/j.pharmthera.2016.03.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Advances in the understanding of neonatal Fc receptor (FcRn) biology and function have demonstrated that this receptor, primarily identified for the transfer of passive immunity from mother infant, is involved in several biological and immunological processes. In fact, FcRn is responsible for the long half-life of IgG and albumin in the serum, by creating an intracellular protein reservoir, which is protected from lysosomal degradation and, importantly, trafficked across the cell. Such discovery has led researchers to hypothesize the role for this unique receptor in the controlled delivery of therapeutic agents. A great amount of FcRn-based strategies are already under extensive investigation, in which FcRn reveals to have profound impact on the biodistribution and half-life extension of therapeutic agents. This review summarizes the main findings on FcRn biology, function and distribution throughout different tissues, together with the main advances on the FcRn-based therapeutic opportunities and model systems, which indicate that this receptor is a potential target for therapeutic regimen modification.
Collapse
Affiliation(s)
- João Pedro Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal
| | - Patrick J Kennedy
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo 228, 4150-180 Porto, Portugal; Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI -00014 Helsinki, Finland
| | - Cristina Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde and Instituto Universitário de Ciências da Saúde, 4585-116 Gandra, Portugal.
| |
Collapse
|
37
|
Foss S, Grevys A, Sand KMK, Bern M, Blundell P, Michaelsen TE, Pleass RJ, Sandlie I, Andersen JT. Enhanced FcRn-dependent transepithelial delivery of IgG by Fc-engineering and polymerization. J Control Release 2015; 223:42-52. [PMID: 26718855 DOI: 10.1016/j.jconrel.2015.12.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/14/2015] [Accepted: 12/19/2015] [Indexed: 01/28/2023]
Abstract
Monoclonal IgG antibodies (Abs) are used extensively in the clinic to treat cancer and autoimmune diseases. In addition, therapeutic proteins are genetically fused to the constant Fc part of IgG. In both cases, the Fc secures a long serum half-life and favourable pharmacokinetics due to its pH-dependent interaction with the neonatal Fc receptor (FcRn). FcRn also mediates transport of intact IgG across polarized epithelial barriers, a pathway that is attractive for delivery of Fc-containing therapeutics. So far, no study has thoroughly compared side-by-side how IgG and different Fc-fusion formats are transported across human polarizing epithelial cells. Here, we used an in vitro cellular transport assay based on the human polarizing epithelial cell line (T84) in which both IgG1 and Fc-fusions were transported in an FcRn-dependent manner. Furthermore, we found that the efficacy of transport was dependent on the format. We demonstrate that transepithelial delivery could be enhanced by Fc-engineering for improved FcRn binding as well as by Fc-polymerization. In both cases, transport was driven by pH-dependent binding kinetics and the pH at the luminal side. Hence, efficient transcellular delivery of IgG-based drugs across human epithelial cells requires optimal pH-dependent FcRn binding that can be manipulated by avidity and Fc-engineering, factors that should inspire the design of future therapeutics targeted for transmucosal delivery.
Collapse
Affiliation(s)
- Stian Foss
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Algirdas Grevys
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Malin Bern
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Pat Blundell
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Terje E Michaelsen
- Department of Bacteriology and Immunology, Norwegian Institute of Public Health, Oslo, Norway; Department of Chemical Pharmacy, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Richard J Pleass
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Inger Sandlie
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Jan Terje Andersen
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway.
| |
Collapse
|
38
|
The neonatal Fc receptor, FcRn, as a target for drug delivery and therapy. Adv Drug Deliv Rev 2015; 91:109-24. [PMID: 25703189 DOI: 10.1016/j.addr.2015.02.005] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/22/2022]
Abstract
Immunoglobulin G (IgG)-based drugs are arguably the most successful class of protein therapeutics due in part to their remarkably long blood circulation. This arises from IgG interaction with the neonatal Fc receptor, FcRn. FcRn is the central regulator of IgG and albumin homeostasis throughout life and is increasingly being recognized as an important player in autoimmune disease, mucosal immunity, and tumor immune surveillance. Various engineering approaches that hijack or disrupt the FcRn-mediated transport pathway have been devised to develop long-lasting and non-invasive protein therapeutics, protein subunit vaccines, and therapeutics for treatment of autoimmune and infectious disease. In this review, we highlight the diverse biological functions of FcRn, emerging therapeutic opportunities, as well as the associated challenges of targeting FcRn for drug delivery and disease therapy.
Collapse
|
39
|
Ward ES, Ober RJ. Commentary: "There's been a Flaw in Our Thinking". Front Immunol 2015; 6:351. [PMID: 26236309 PMCID: PMC4503918 DOI: 10.3389/fimmu.2015.00351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/28/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center , College Station, TX , USA ; Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center , Bryan, TX , USA ; Department of Immunology, University of Texas Southwestern Medical Center , Dallas, TX , USA
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center , College Station, TX , USA ; Department of Biomedical Engineering, Texas A&M University , College Station, TX , USA
| |
Collapse
|
40
|
Hultberg A, Morello V, Huyghe L, De Jonge N, Blanchetot C, Hanssens V, De Boeck G, Silence K, Festjens E, Heukers R, Roux B, Lamballe F, Ginestier C, Charafe-Jauffret E, Maina F, Brouckaert P, Saunders M, Thibault A, Dreier T, de Haard H, Michieli P. Depleting MET-Expressing Tumor Cells by ADCC Provides a Therapeutic Advantage over Inhibiting HGF/MET Signaling. Cancer Res 2015; 75:3373-83. [DOI: 10.1158/0008-5472.can-15-0356] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/29/2015] [Indexed: 11/16/2022]
|
41
|
Rath T, Baker K, Dumont JA, Peters RT, Jiang H, Qiao SW, Lencer WI, Pierce GF, Blumberg RS. Fc-fusion proteins and FcRn: structural insights for longer-lasting and more effective therapeutics. Crit Rev Biotechnol 2015; 35:235-54. [PMID: 24156398 PMCID: PMC4876602 DOI: 10.3109/07388551.2013.834293] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Nearly 350 IgG-based therapeutics are approved for clinical use or are under development for many diseases lacking adequate treatment options. These include molecularly engineered biologicals comprising the IgG Fc-domain fused to various effector molecules (so-called Fc-fusion proteins) that confer the advantages of IgG, including binding to the neonatal Fc receptor (FcRn) to facilitate in vivo stability, and the therapeutic benefit of the specific effector functions. Advances in IgG structure-function relationships and an understanding of FcRn biology have provided therapeutic opportunities for previously unapproachable diseases. This article discusses approved Fc-fusion therapeutics, novel Fc-fusion proteins and FcRn-dependent delivery approaches in development, and how engineering of the FcRn-Fc interaction can generate longer-lasting and more effective therapeutics.
Collapse
Affiliation(s)
- Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristi Baker
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Shuo-Wang Qiao
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Wayne I. Lencer
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Hanson QM, Barb AW. A perspective on the structure and receptor binding properties of immunoglobulin G Fc. Biochemistry 2015; 54:2931-42. [PMID: 25926001 DOI: 10.1021/acs.biochem.5b00299] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recombinant antibodies spurred a revolution in medicine that saw the introduction of powerful therapeutics for treating a wide range of diseases, from cancers to autoimmune disorders and transplant rejection, with more applications looming on the horizon. Many of these therapeutic monoclonal antibodies (mAbs) are based on human immunoglobulin G1 (IgG1) or contain at least a portion of the molecule. Most mAbs require interactions with cell surface receptors for efficacy, including the Fc γ receptors. High-resolution structural models of antibodies and antibody fragments have been available for nearly 40 years; however, a thorough description of the structural features that determine the affinity with which antibodies interact with human receptors has not been published. In this review, we will cover the relevant history of IgG-related literature and how recent developments have changed our view of critical antibody-cell interactions at the atomic level with a nod to outstanding questions in the field and future prospects.
Collapse
Affiliation(s)
- Quinlin M Hanson
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, 2214 Molecular Biology Building, Ames, Iowa 50011, United States
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, 2214 Molecular Biology Building, Ames, Iowa 50011, United States
| |
Collapse
|
43
|
Grevys A, Bern M, Foss S, Bratlie DB, Moen A, Gunnarsen KS, Aase A, Michaelsen TE, Sandlie I, Andersen JT. Fc Engineering of Human IgG1 for Altered Binding to the Neonatal Fc Receptor Affects Fc Effector Functions. THE JOURNAL OF IMMUNOLOGY 2015; 194:5497-508. [PMID: 25904551 DOI: 10.4049/jimmunol.1401218] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 03/23/2015] [Indexed: 12/18/2022]
Abstract
Engineering of the constant Fc part of monoclonal human IgG1 (hIgG1) Abs is an approach to improve effector functions and clinical efficacy of next-generation IgG1-based therapeutics. A main focus in such development is tailoring of in vivo half-life and transport properties by engineering the pH-dependent interaction between IgG and the neonatal Fc receptor (FcRn), as FcRn is the main homeostatic regulator of hIgG1 half-life. However, whether such engineering affects binding to other Fc-binding molecules, such as the classical FcγRs and complement factor C1q, has not been studied in detail. These effector molecules bind to IgG1 in the lower hinge-CH2 region, structurally distant from the binding site for FcRn at the CH2-CH3 elbow region. However, alterations of the structural composition of the Fc may have long-distance effects. Indeed, in this study we show that Fc engineering of hIgG1 for altered binding to FcRn also influences binding to both the classical FcγRs and complement factor C1q, which ultimately results in alterations of cellular mechanisms such as Ab-dependent cell-mediated cytotoxicity, Ab-dependent cellular phagocytosis, and Ab-dependent complement-mediated cell lysis. Thus, engineering of the FcRn-IgG1 interaction may greatly influence effector functions, which has implications for the therapeutic efficacy and use of Fc-engineered hIgG1 variants.
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Centre for Immune Regulation and Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, 0372 Oslo, Norway
| | - Malin Bern
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Centre for Immune Regulation and Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, 0372 Oslo, Norway
| | - Stian Foss
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Centre for Immune Regulation and Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, 0372 Oslo, Norway
| | - Diane Bryant Bratlie
- Department of Bacteriology and Immunology, Norwegian Institute of Public Health, 0403 Oslo, Norway
| | - Anders Moen
- Department of Biosciences and the Mass Spectrometry and Proteomics Unit, University of Oslo, 0371 Oslo, Norway; and
| | - Kristin Støen Gunnarsen
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Centre for Immune Regulation and Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, 0372 Oslo, Norway
| | - Audun Aase
- Department of Bacteriology and Immunology, Norwegian Institute of Public Health, 0403 Oslo, Norway
| | - Terje Einar Michaelsen
- Department of Bacteriology and Immunology, Norwegian Institute of Public Health, 0403 Oslo, Norway; Department of Chemical Pharmacy, School of Pharmacy, University of Oslo, 0316 Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation and Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Centre for Immune Regulation and Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, 0372 Oslo, Norway
| | - Jan Terje Andersen
- Centre for Immune Regulation and Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, 0372 Oslo, Norway;
| |
Collapse
|
44
|
Targeting FcRn for the modulation of antibody dynamics. Mol Immunol 2015; 67:131-41. [PMID: 25766596 DOI: 10.1016/j.molimm.2015.02.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/08/2023]
Abstract
The MHC class I-related receptor, FcRn, is a multitasking protein that transports its IgG ligand within and across cells of diverse origins. The role of this receptor as a global regulator of IgG homeostasis and transport, combined with knowledge of the molecular details of FcRn-IgG interactions, has led to opportunities to modulate the in vivo dynamics of antibodies and their antigens through protein engineering. Consequently, the generation of half-life extended antibodies has shown a rapid expansion over the past decade. Further, FcRn itself can be targeted by inhibitors to induce decreased levels of circulating IgGs, which could have applications in multiple clinical settings. The engineering of antibody-antigen interactions to reduce antibody-mediated buffering of soluble ligand has also developed into an active area of investigation, leading to novel antibody platforms designed to result in more effective antigen clearance. Similarly, the target-mediated elimination of antibodies by internalizing, membrane bound antigens (receptors) can be decreased using novel engineering approaches. These strategies, combined with subcellular trafficking analyses of antibody/antigen/FcRn behavior in cells to predict in vivo behavior, have considerable promise for the production of next generation therapeutics and diagnostics.
Collapse
|
45
|
Monnet C, Jorieux S, Urbain R, Fournier N, Bouayadi K, De Romeuf C, Behrens CK, Fontayne A, Mondon P. Selection of IgG Variants with Increased FcRn Binding Using Random and Directed Mutagenesis: Impact on Effector Functions. Front Immunol 2015; 6:39. [PMID: 25699055 PMCID: PMC4316771 DOI: 10.3389/fimmu.2015.00039] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/21/2015] [Indexed: 12/26/2022] Open
Abstract
Despite the reasonably long half-life of immunoglogulin G (IgGs), market pressure for higher patient convenience while conserving efficacy continues to drive IgG half-life improvement. IgG half-life is dependent on the neonatal Fc receptor (FcRn), which among other functions, protects IgG from catabolism. FcRn binds the Fc domain of IgG at an acidic pH ensuring that endocytosed IgG will not be degraded in lysosomal compartments and will then be released into the bloodstream. Consistent with this mechanism of action, several Fc-engineered IgG with increased FcRn affinity and conserved pH dependency were designed and resulted in longer half-life in vivo in human FcRn-transgenic mice (hFcRn), cynomolgus monkeys, and recently in healthy humans. These IgG variants were usually obtained by in silico approaches or directed mutagenesis in the FcRn-binding site. Using random mutagenesis, combined with a pH-dependent phage display selection process, we isolated IgG variants with improved FcRn-binding, which exhibited longer in vivo half-life in hFcRn mice. Interestingly, many mutations enhancing Fc/FcRn interaction were located at a distance from the FcRn-binding site validating our random molecular approach. Directed mutagenesis was then applied to generate new variants to further characterize our IgG variants and the effect of the mutations selected. Since these mutations are distributed over the whole Fc sequence, binding to other Fc effectors, such as complement C1q and FcγRs, was dramatically modified, even by mutations distant from these effectors’ binding sites. Hence, we obtained numerous IgG variants with increased FcRn-binding and different binding patterns to other Fc effectors, including variants without any effector function, providing distinct “fit-for-purpose” Fc molecules. We therefore provide evidence that half-life and effector functions should be optimized simultaneously as mutations can have unexpected effects on all Fc receptors that are critical for IgG therapeutic efficacy.
Collapse
|
46
|
Deyev SM, Lebedenko EN, Petrovskaya LE, Dolgikh DA, Gabibov AG, Kirpichnikov MP. Man-made antibodies and immunoconjugates with desired properties: function optimization using structural engineering. RUSSIAN CHEMICAL REVIEWS 2015. [DOI: 10.1070/rcr4459] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
47
|
Sand KMK, Bern M, Nilsen J, Noordzij HT, Sandlie I, Andersen JT. Unraveling the Interaction between FcRn and Albumin: Opportunities for Design of Albumin-Based Therapeutics. Front Immunol 2015; 5:682. [PMID: 25674083 PMCID: PMC4306297 DOI: 10.3389/fimmu.2014.00682] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/17/2014] [Indexed: 01/08/2023] Open
Abstract
The neonatal Fc receptor (FcRn) was first found to be responsible for transporting antibodies of the immunoglobulin G (IgG) class from the mother to the fetus or neonate as well as for protecting IgG from intracellular catabolism. However, it has now become apparent that the same receptor also binds albumin and plays a fundamental role in homeostatic regulation of both IgG and albumin, as FcRn is expressed in many different cell types and organs at diverse body sites. Thus, to gain a complete understanding of the biological function of each ligand, and also their distribution in the body, an in-depth characterization of how FcRn binds and regulates the transport of both ligands is necessary. Importantly, such knowledge is also relevant when developing new drugs, as IgG and albumin are increasingly utilized in therapy. This review discusses our current structural and biological understanding of the relationship between FcRn and its ligands, with a particular focus on albumin and design of albumin-based therapeutics.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Malin Bern
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jeannette Nilsen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway ; Institute of Clinical Medicine, University of Oslo , Oslo , Norway
| | - Hanna Theodora Noordzij
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jan Terje Andersen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| |
Collapse
|
48
|
Devanaboyina SC, Lynch SM, Ober RJ, Ram S, Kim D, Puig-Canto A, Breen S, Kasturirangan S, Fowler S, Peng L, Zhong H, Jermutus L, Wu H, Webster C, Ward ES, Gao C. The effect of pH dependence of antibody-antigen interactions on subcellular trafficking dynamics. MAbs 2015; 5:851-9. [PMID: 24492341 DOI: 10.4161/mabs.26389] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A drawback of targeting soluble antigens such as cytokines or toxins with long-lived antibodies is that such antibodies can prolong the half-life of the target antigen by a "buffering" effect. This has motivated the design of antibodies that bind to target with higher affinity at near neutral pH relative to acidic endosomal pH (~pH 6.0). Such antibodies are expected to release antigen within endosomes following uptake into cells, whereas antibody will be recycled and exocytosed in FcRn-expressing cells. To understand how the pH dependence of antibody-antigen interactions affects intracellular trafficking, we generated three antibodies that bind IL-6 with different pH dependencies in the range pH 6.0-7.4. The behavior of antigen in the presence of these antibodies has been characterized using a combination of fixed and live cell fluorescence microscopy. As the affinity of the antibody:IL-6 interaction at pH 6.0 decreases, an increasing amount of antigen dissociates from FcRn-bound antibody in early and late endosomes, and then enters lysosomes. Segregation of antibody and FcRn from endosomes in tubulovesicular transport carriers (TCs) into the recycling pathway can also be observed in live cells, and the extent of IL-6 association with TCs correlates with increasing affinity of the antibody:IL-6 interaction at acidic pH. These analyses result in an understanding, in spatiotemporal terms, of the effect of pH dependence of antibody-antigen interactions on subcellular trafficking and inform the design of antibodies with optimized binding properties for antigen elimination.
Collapse
Affiliation(s)
| | - Sandra M Lynch
- Department of Immunology; University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Raimund J Ober
- Department of Immunology; University of Texas Southwestern Medical Center; Dallas, TX, USA; Department of Electrical Engineering; University of Texas at Dallas; Richardson, TX, USA
| | - Sripad Ram
- Department of Immunology; University of Texas Southwestern Medical Center; Dallas, TX, USA; Department of Electrical Engineering; University of Texas at Dallas; Richardson, TX, USA
| | - Dongyoung Kim
- Department of Immunology; University of Texas Southwestern Medical Center; Dallas, TX, USA; Department of Electrical Engineering; University of Texas at Dallas; Richardson, TX, USA
| | - Alberto Puig-Canto
- Department of Immunology; University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Shannon Breen
- Department of Oncology; Medimmune; Gaithersburg, MD, USA
| | - Srinath Kasturirangan
- Department of Antibody Discovery & Protein Engineering; Medimmune; Gaithersburg, MD, USA
| | - Susan Fowler
- Department of Antibody Discovery & Protein Engineering; Medimmune; Granta Park, Cambridge, UK
| | - Li Peng
- Department of Antibody Discovery & Protein Engineering; Medimmune; Gaithersburg, MD, USA
| | - Haihong Zhong
- Department of Oncology; Medimmune; Gaithersburg, MD, USA
| | - Lutz Jermutus
- Department of Antibody Discovery & Protein Engineering; Medimmune; Granta Park, Cambridge, UK
| | - Herren Wu
- Department of Antibody Discovery & Protein Engineering; Medimmune; Gaithersburg, MD, USA
| | - Carl Webster
- Department of Antibody Discovery & Protein Engineering; Medimmune; Granta Park, Cambridge, UK
| | - E Sally Ward
- Department of Immunology; University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Changshou Gao
- Department of Antibody Discovery & Protein Engineering; Medimmune; Gaithersburg, MD, USA
| |
Collapse
|
49
|
Wu YY, V. Nguyen A, Wu XX, Loh M, Vu M, Zou Y, Liu Q, Guo P, Wang Y, Montgomery LL, Orlofsky A, Rand JH, Lin EY. Antiphospholipid Antibodies Promote Tissue Factor–Dependent Angiogenic Switch and Tumor Progression. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3359-75. [DOI: 10.1016/j.ajpath.2014.07.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 07/22/2014] [Accepted: 07/29/2014] [Indexed: 12/30/2022]
|
50
|
Abstract
INTRODUCTION The Fc receptors (FcRs) and their interactions with immunoglobulin and innate immune opsonins, such as C-reactive protein, are key players in humoral and cellular immune responses. As the effector mechanism for some therapeutic monoclonal antibodies, and often a contributor to the pathogenesis and progression of autoimmunity, FcRs are promising targets for treating autoimmune diseases. AREAS COVERED This review discusses the nature of different FcRs and the various mechanisms of their involvement in initiating and modulating immunocyte functions and their biological consequences. It describes a range of current strategies in targeting FcRs and manipulating their interaction with specific ligands, while presenting the pros and cons of these approaches. This review also discusses potential new strategies including regulation of FcR expression and receptor crosstalk. EXPERT OPINION FcRs are appealing targets in the treatment of inflammatory autoimmune diseases. However, there are still knowledge limitations and technical challenges, the most important being a better understanding of the individual roles of each of the FcRs and enhancement of the specificity in targeting particular cell types and specific FcRs.
Collapse
Affiliation(s)
- Xinrui Li
- The University of Alabama , SHEL 272, 1825 University Blvd, Birmingham, AL 35294 , USA
| | | |
Collapse
|