1
|
Grevys A, Frick R, Mester S, Flem-Karlsen K, Nilsen J, Foss S, Sand KMK, Emrich T, Fischer JAA, Greiff V, Sandlie I, Schlothauer T, Andersen JT. Antibody variable sequences have a pronounced effect on cellular transport and plasma half-life. iScience 2022; 25:103746. [PMID: 35118359 PMCID: PMC8800109 DOI: 10.1016/j.isci.2022.103746] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 01/05/2022] [Indexed: 11/15/2022] Open
Abstract
Monoclonal IgG antibodies are the fastest growing class of biologics, but large differences exist in their plasma half-life in humans. Thus, to design IgG antibodies with favorable pharmacokinetics, it is crucial to identify the determinants of such differences. Here, we demonstrate that the variable region sequences of IgG antibodies greatly affect cellular uptake and subsequent recycling and rescue from intracellular degradation by endothelial cells. When the variable sequences are masked by the cognate antigen, it influences both their transport behavior and binding to the neonatal Fc receptor (FcRn), a key regulator of IgG plasma half-life. Furthermore, we show how charge patch differences in the variable domains modulate both binding and transport properties and that a short plasma half-life, due to unfavorable charge patches, may partly be overcome by Fc-engineering for improved FcRn binding. IgG variable region sequences greatly affect cellular uptake and recycling Variable region charge patches affect FcRn binding and transport The presence of cognate antigen modulates cellular transport and FcRn binding Fc-engineering for improved FcRn binding can overcome unfavorable charge patches
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
- Corresponding author
| | - Rahel Frick
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Simone Mester
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Karine Flem-Karlsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Stian Foss
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Thomas Emrich
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | | | - Victor Greiff
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Corresponding author
| |
Collapse
|
2
|
van Bussel MTJ, Beijnen JH, Brandsma D. Intracranial antitumor responses of nivolumab and ipilimumab: a pharmacodynamic and pharmacokinetic perspective, a scoping systematic review. BMC Cancer 2019; 19:519. [PMID: 31146733 PMCID: PMC6543612 DOI: 10.1186/s12885-019-5741-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/22/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Recently, two phase II trials showed intracranial activity of the immune checkpoint inhibitors nivolumab and ipilimumab in patients with melanoma brain metastases. However, it is generally assumed that large molecules like monoclonal antibodies nivolumab and ipilimumab cannot penetrate and pass an intact blood brain barrier (BBB). In this systematic review we provide a pharmacodynamic and pharmacokinetic consideration of the clinical activity of the immune checkpoint inhibitors nivolumab and ipilimumab in melanoma brain metastases. METHODS Pubmed was systematically searched for prospective phase II and III studies on nivolumab and ipilimumab in melanoma brain metastases and cerebrospinal fluid (CSF) levels of nivolumab and ipilimumab. Results were discussed and a perspective on the pharmacodynamics and pharmacokinetics for the intracranial activity of these agents was given. RESULTS Two phase II studies with the combination nivolumab and ipilimumab and one phase II study with ipilimumab monotherapy in melanoma brain metastases were included in this review. One article reported drug levels of nivolumab in CSF. Intracranial responses were achieved in 16 of 35 patients (46%; 95% confidence interval (CI) 29-63) in a phase II study cohort treated with nivolumab and ipilimumab. In a second phase II study in 94 patients, the rate of intracranial clinical benefit was 57% (95% CI 47-68). The CSF/serum ratio of nivolumab was 0.88-1.9% in a cohort of metastatic melanoma patients treated with nivolumab 1-3 mg/kg. Nivolumab concentrations ranged from 35 to 150 ng/ml in CSF of these patients, which is in the range of the half maximal effective concentration (EC50) of 0.64 nM. CONCLUSIONS Ipilimumab and nivolumab are active in melanoma brain metastases. Nivolumab penetrates into the CSF. Based on the described findings the general consensus that monoclonal antibodies do not penetrate into the central nervous system (CNS) and cannot have a direct intracranial effect needs to be reconsidered.
Collapse
Affiliation(s)
- Mark T J van Bussel
- Division of Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Department of Medical Oncology & Clinical Pharmacology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Dieta Brandsma
- Department of Neuro-oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| |
Collapse
|
3
|
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: 9.3] [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
|
4
|
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: 2.0] [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
|
5
|
Sand KMK, Dalhus B, Christianson GJ, Bern M, Foss S, Cameron J, Sleep D, Bjørås M, Roopenian DC, Sandlie I, Andersen JT. Dissection of the neonatal Fc receptor (FcRn)-albumin interface using mutagenesis and anti-FcRn albumin-blocking antibodies. J Biol Chem 2014; 289:17228-39. [PMID: 24764301 DOI: 10.1074/jbc.m113.522565] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Albumin is the most abundant protein in blood and plays a pivotal role as a multitransporter of a wide range of molecules such as fatty acids, metabolites, hormones, and toxins. In addition, it binds a variety of drugs. Its role as distributor is supported by its extraordinary serum half-life of 3 weeks. This is related to its size and binding to the cellular receptor FcRn, which rescues albumin from intracellular degradation. Furthermore, the long half-life has fostered a great and increasing interest in utilization of albumin as a carrier of protein therapeutics and chemical drugs. However, to fully understand how FcRn acts as a regulator of albumin homeostasis and to take advantage of the FcRn-albumin interaction in drug design, the interaction interface needs to be dissected. Here, we used a panel of monoclonal antibodies directed towards human FcRn in combination with site-directed mutagenesis and structural modeling to unmask the binding sites for albumin blocking antibodies and albumin on the receptor, which revealed that the interaction is not only strictly pH-dependent, but predominantly hydrophobic in nature. Specifically, we provide mechanistic evidence for a crucial role of a cluster of conserved tryptophan residues that expose a pH-sensitive loop of FcRn, and identify structural differences in proximity to these hot spot residues that explain divergent cross-species binding properties of FcRn. Our findings expand our knowledge of how FcRn is controlling albumin homeostasis at a molecular level, which will guide design and engineering of novel albumin variants with altered transport properties.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- From the Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316 Oslo, Norway, CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Norway, N-0424 Oslo, Norway
| | - Bjørn Dalhus
- the Department for Microbiology, Oslo University Hospital Rikshospitalet and University of Oslo, Nydalen, N-0424 Oslo, Norway, the Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet and University of Oslo, Nydalen, N-0424 Oslo, Norway
| | | | - Malin Bern
- From the Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316 Oslo, Norway, CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Norway, N-0424 Oslo, Norway
| | - Stian Foss
- From the Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316 Oslo, Norway, CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Norway, N-0424 Oslo, Norway
| | - Jason Cameron
- Novozymes Biopharma UK, Ltd., Castle Court, 59 Castle Boulevard, NG7 1FD Nottingham, United Kingdom
| | - Darrell Sleep
- Novozymes Biopharma UK, Ltd., Castle Court, 59 Castle Boulevard, NG7 1FD Nottingham, United Kingdom
| | - Magnar Bjørås
- the Department for Microbiology, Oslo University Hospital Rikshospitalet and University of Oslo, Nydalen, N-0424 Oslo, Norway, the Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet and University of Oslo, Nydalen, N-0424 Oslo, Norway
| | | | - Inger Sandlie
- From the Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0316 Oslo, Norway, CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Norway, N-0424 Oslo, Norway
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Norway, N-0424 Oslo, Norway,
| |
Collapse
|
6
|
FcRn: from molecular interactions to regulation of IgG pharmacokinetics and functions. Curr Top Microbiol Immunol 2014; 382:249-72. [PMID: 25116104 DOI: 10.1007/978-3-319-07911-0_12] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neonatal Fc receptor, FcRn, is related to MHC class I with respect to its structure and association with β2microglobulin (β2m). However, by contrast with MHC class I molecules, FcRn does not bind to peptides, but interacts with the Fc portion of IgGs and belongs to the Fc receptor family. Unlike the 'classical' Fc receptors, however, the primary functions of FcRn include salvage of IgG (and albumin) from lysosomal degradation through the recycling and transcytosis of IgG within cells. The characteristic feature of FcRn is pH-dependent binding to IgG, with relatively strong binding at acidic pH (<6.5) and negligible binding at physiological pH (7.3-7.4). FcRn is expressed in many different cell types, and endothelial and hematopoietic cells are the dominant cell types involved in IgG homeostasis in vivo. FcRn also delivers IgG across cellular barriers to sites of pathogen encounter and consequently plays a role in protection against infections, in addition to regulating renal filtration and immune complex-mediated antigen presentation. Further, FcRn has been targeted to develop both IgGs with extended half-lives and FcRn inhibitors that can lower endogenous antibody levels. These approaches have implications for the development of longer lived therapeutics and the removal of pathogenic or deleterious antibodies.
Collapse
|
7
|
Receptor-mediated endocytosis and brain delivery of therapeutic biologics. Int J Cell Biol 2013; 2013:703545. [PMID: 23840214 PMCID: PMC3693099 DOI: 10.1155/2013/703545] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/13/2013] [Indexed: 11/18/2022] Open
Abstract
Transport of macromolecules across the blood-brain-barrier (BBB) requires both specific and nonspecific interactions between macromolecules and proteins/receptors expressed on the luminal and/or the abluminal surfaces of the brain capillary endothelial cells. Endocytosis and transcytosis play important roles in the distribution of macromolecules. Due to the tight junction of BBB, brain delivery of traditional therapeutic proteins with large molecular weight is generally not possible. There are multiple pathways through which macromolecules can be taken up into cells through both specific and nonspecific interactions with proteins/receptors on the cell surface. This review is focused on the current knowledge of receptor-mediated endocytosis/transcytosis and brain delivery using the Angiopep-2-conjugated system and the molecular Trojan horses. In addition, the role of neonatal Fc receptor (FcRn) in regulating the efflux of Immunoglobulin G (IgG) from brain to blood, and approaches to improve the pharmacokinetics of therapeutic biologics by generating Fc fusion proteins, and increasing the pH dependent binding affinity between Fc and FcRn, are discussed.
Collapse
|
8
|
Murai A, Murota R, Doi K, Yoshida T, Aoyama H, Kobayashi M, Horio F. Avian IgY is selectively incorporated into the egg yolks of oocytes by discriminating Fc amino acid residues located on the Cυ3/Cυ4 interface. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 39:378-387. [PMID: 23276880 DOI: 10.1016/j.dci.2012.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 12/03/2012] [Accepted: 12/03/2012] [Indexed: 06/01/2023]
Abstract
In avian species, maternal IgY is selectively incorporated into the egg yolks of maturing oocytes, but the relevance of receptor-mediated uptake is unclear. Here we investigated the critical amino acid residues of IgY required for egg yolk transport by conducting mutational analyses of selected residues located along the Cυ3 and Cυ4 domains of chicken IgY. Recombinant wild-type IgY-Fc (WT) and its mutants were synthesized, and their uptakes into the egg yolks of quail were determined. Among the 17 amino acid residues located on the Cυ3/Cυ4 interface, the substitution of Y363 at the Cυ3 domain to alanine abolished the IgY-Fc uptake into egg yolks. The comprehensive substitution of Y363 with other amino acids revealed that the residue at 363 needs to be allocated with aromatic amino acids to maintain the high transport ability. The deglycosylation of the N-linked carbohydrate chain by substituting N407 at the Cυ3 domain with alanine also caused a marked reduction of IgY-Fc uptake. The microscopic detection of the injected WT and Y363A mutant in ovarian follicles showed that the WT was concentrically accumulated in yolk granules, whereas the Y363A mutant was hardly accumulated in yolk granules, but it had infiltrated into the granulosa cell layer, suggesting that a major hurdle disturbing the infiltration of the Y363A mutant lies on the inside of the granulosa cell layer. The identification of important amino acid residues required for efficient IgY transport enhances our understanding of the molecular mechanisms underlying IgY transport through a specific IgY receptor in ovarian follicles.
Collapse
Affiliation(s)
- Atsushi Murai
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
9
|
|
10
|
Sesarman A, Vidarsson G, Sitaru C. The neonatal Fc receptor as therapeutic target in IgG-mediated autoimmune diseases. Cell Mol Life Sci 2010; 67:2533-50. [PMID: 20217455 PMCID: PMC11115620 DOI: 10.1007/s00018-010-0318-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 01/29/2010] [Accepted: 02/12/2010] [Indexed: 01/01/2023]
Abstract
Therapy approaches based on lowering levels of pathogenic autoantibodies represent rational, effective, and safe treatment modalities of autoimmune diseases. The neonatal Fc receptor (FcRn) is a major factor regulating the serum levels of IgG antibodies. While FcRn-mediated half-life extension is beneficial for IgG antibody responses against pathogens, it also prolongs the serum half-life of IgG autoantibodies and thus promotes tissue damage in autoimmune diseases. In the present review article, we examine current evidence on the relevance of FcRn in maintaining high autoantibody levels and discuss FcRn-targeted therapeutic approaches. Further investigation of the FcRn-IgG interaction will not only provide mechanistic insights into the receptor function, but should also greatly facilitate the design of therapeutics combining optimal pharmacokinetic properties with the appropriate antibody effector functions in autoimmune diseases.
Collapse
Affiliation(s)
- Alina Sesarman
- Department of Dermatology, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Cassian Sitaru
- Department of Dermatology, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
- Centre for Biological Signalling Studies (bioss), University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Masuda A, Yoshida M, Shiomi H, Morita Y, Kutsumi H, Inokuchi H, Mizuno S, Nakamura A, Takai T, Blumberg RS, Azuma T. Role of Fc Receptors as a therapeutic target. ACTA ACUST UNITED AC 2009; 8:80-6. [PMID: 19275696 DOI: 10.2174/187152809787582525] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has been forty years since the discovery of Fc Receptors and their function. Fc Receptors include the IgG receptors (FcgammaR), high-affinity IgE receptor (FcepsilonRI), IgA and IgA/IgM receptors, and neonatal Fc receptor for IgG (FcRn). In particular, the FcgammaRs have been well known to play an important role in many biologic processes including those associated with the response to infection and cancer as well as in the pathogenesis of immune-mediated diseases. Both positive and negative regulatory function has ascribed to Fc receptors and FcgammaRs in particular which serve to establish a threshold for immune cell activation. In other cases, Fc receptors such as FcRn possess a novel structure and function by playing a major role in the transport of IgG across polarized epithelial barriers at mucosal surfaces and in the regulation of IgG half-life. These diverse functions highlight the potential effectiveness of targeting Fc receptors for therapeutic purposes. This review summarizes new information available in the therapeutic applications of this biology.
Collapse
Affiliation(s)
- Atsuhiro Masuda
- Department of Gastroenterology, Kobe University School of Medicine, Kobe, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ward ES, Ober RJ. Chapter 4: Multitasking by exploitation of intracellular transport functions the many faces of FcRn. Adv Immunol 2009; 103:77-115. [PMID: 19755184 DOI: 10.1016/s0065-2776(09)03004-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The MHC Class I-related receptor, FcRn, transports antibodies of the immunoglobulin G (IgG) class within and across a diverse array of different cell types. Through this transport, FcRn serves multiple roles throughout adult life that extend well beyond its earlier defined function of transcytosing IgGs from mother to offspring. These roles include the maintenance of IgG levels and the delivery of antigen in the form of immune complexes to degradative compartments within cells. Recent studies have led to significant advances in knowledge of the intracellular trafficking of FcRn and (engineered) IgGs at both the molecular and cellular levels. The engineering of FcRn-IgG (or Fc) interactions to generate antibodies of increased longevity represents an area of active interest, particularly in the light of the expanding use of antibodies in therapy. The strict pH dependence of FcRn-IgG interactions, with binding at pH 6 that becomes essentially undetectable as near neutral pH is approached, is essential for efficient transport. The requirement for retention of low affinity at near neutral pH increases the complexity of engineering antibodies for increased half-life. Conversely, engineered IgGs that have gained significant binding for FcRn at this pH can be potent inhibitors of FcRn that lower endogenous IgG levels and have multiple potential uses as therapeutics. In addition, molecular studies of FcRn-IgG interactions indicate that mice have limitations as preclinical models for FcRn function, primarily due to cross-species differences in FcRn-binding specificity.
Collapse
Affiliation(s)
- E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
13
|
Andersen JT, Sandlie I. The Versatile MHC Class I-related FcRn Protects IgG and Albumin from Degradation: Implications for Development of New Diagnostics and Therapeutics. Drug Metab Pharmacokinet 2009; 24:318-32. [DOI: 10.2133/dmpk.24.318] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Polte T, Hennig C, Hansen G. Allergy prevention starts before conception: maternofetal transfer of tolerance protects against the development of asthma. J Allergy Clin Immunol 2008; 122:1022-1030.e5. [PMID: 19000583 DOI: 10.1016/j.jaci.2008.09.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 09/05/2008] [Accepted: 09/08/2008] [Indexed: 11/13/2022]
Abstract
BACKGROUND Allergy prevention must start early because environmental exposures during pregnancy and young life determine disease risk. OBJECTIVE In this study we analyzed whether prevention can start even earlier before conception by transfer of immunologic tolerance from the mother to the offspring preventing the offspring from having asthma. METHODS BALB/c mice were orally tolerized with ovalbumin before conception by means of oral application of antigen. The offspring of tolerized and naive mothers were immunized with ovalbumin at 6 weeks and 4 months of age and analyzed in our murine asthma model. RESULTS Although the offspring of naive mothers had an asthma-like phenotype, the offspring of tolerized mice were completely protected, even when immunized as late as 8 months after birth. Critically involved in the tolerance transfer was allergen-specific IgG, levels of which were increased in the sera of the mother, fetus, and pup and breast milk. FcRn(-/-) mice, which cannot transport IgG through the placenta, transferred tolerance to the offspring only when the missing diaplacental IgG transfer was compensated by IgG transfer through breast milk from tolerant mothers but not when weaned by naive wet nurses. Inhibition of IFN-gamma, produced by memory T cells in the offspring, abrogated the protective effect of maternal tolerance, demonstrating a crucial role for IFN-gamma in the maintenance of allergen-specific tolerance. CONCLUSION Our data show that maternal immunologic memory has a significant and persistent effect on the immune response of the offspring.
Collapse
Affiliation(s)
- Tobias Polte
- Department of Pediatrics, Division of Pediatric Pulmonology and Neonatology, Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
15
|
Datta-Mannan A, Witcher DR, Tang Y, Watkins J, Jiang W, Wroblewski VJ. Humanized IgG1 variants with differential binding properties to the neonatal Fc receptor: relationship to pharmacokinetics in mice and primates. Drug Metab Dispos 2007; 35:86-94. [PMID: 17050651 DOI: 10.1124/dmd.106.011734] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is well established that the neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. As such, modification of the interaction of IgG with FcRn has been the focus of protein-engineering strategies designed to generate therapeutic antibodies with improved pharmacokinetic properties. In the current work, we characterized differences in interaction of IgG between mouse and primate receptors using three humanized anti-tumor necrosis factor alpha antibodies with variant IgG(1) Fc regions. The wild-type and variant IgG showed a differential combination of improved affinity, modified dissociation kinetics, and altered pH-dependent complex dissociation when evaluated on the primate and murine receptors. The observed in vitro binding differences within and between species allowed us to more completely relate these parameters to their influence on the in vivo pharmacokinetics in mice and cynomolgus monkeys. The variant antibodies have different pharmacokinetic behavior in cynomolgus monkeys and mice, which appears to be related to the unique binding characteristics observed with the murine receptor. However, we did not observe a direct relationship between increased binding affinity to the receptor and improved pharmacokinetic properties for these molecules in either species. This work provides further insights into how the FcRn/IgG interaction may be modulated to develop monoclonal antibodies with improved therapeutic properties.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Lilly Research Laboratories, Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | | | | | | | | | | |
Collapse
|
16
|
Datta-Mannan A, Witcher DR, Tang Y, Watkins J, Wroblewski VJ. Monoclonal Antibody Clearance. J Biol Chem 2007; 282:1709-17. [PMID: 17135257 DOI: 10.1074/jbc.m607161200] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. There are mixed reports on whether modification of the interaction with FcRn can be used as an engineering strategy to improve the pharmacokinetic and pharmacodynamic properties of monoclonal antibodies. We tested whether the T250Q/M428L mutations, which improved the pharmacokinetics of humanized IgGs in the rhesus monkey, would translate to a pharmacokinetic benefit in both cynomolgus monkeys and mice when constructed on a different humanized IgG framework (anti-tumor necrosis factor-alpha (TNFalpha)). The T250Q/M428L anti-TNFalpha variant displayed an approximately 40-fold increase in binding affinity to cynomolgus monkey FcRn (C-FcRn) at pH 6.0, with maintenance of the pH binding dependence. We also constructed another anti-TNFalpha variant (P257I/Q311I) whose binding kinetics with the C-FcRn was similar to that of the T250Q/M428L variant. The binding affinity of the T250Q/M428L variant for murine FcRn was increased approximately 500-fold, with maintenance of pH dependence. In contrast to the interaction with C-FcRn, this interaction was driven mainly by a decrease in the rate of dissociation. Despite the improved in vitro binding properties of the anti-TNFalpha T250Q/M428L and P257I/Q311I variants to C-FcRn, the pharmacokinetic profiles of these molecules were not differentiated from the wild-type antibody in cynomolgus monkeys after intravenous administration. When administered intravenously to mice, the T250Q/M428L anti-TNFalpha variant displayed improved pharmacokinetics, characterized by an approximately 2-fold slower clearance than the wild-type antibody. The discrepancy between these data and previously reported benefits in rhesus monkeys and the inability of these mutations to translate to improved kinetics across species may be related to a number of factors. We propose extending consideration to differences in the absolute IgG-FcRn affinity, the kinetics of the IgG/FcRn interaction, and differences in the relative involvement of this pathway in the context of other factors influencing the disposition or elimination of monoclonal antibodies.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Department of Drug Disposition Development/Commercialization and Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285, USA
| | | | | | | | | |
Collapse
|
17
|
Yoshida M, Masuda A, Kuo TT, Kobayashi K, Claypool SM, Takagawa T, Kutsumi H, Azuma T, Lencer WI, Blumberg RS. IgG transport across mucosal barriers by neonatal Fc receptor for IgG and mucosal immunity. ACTA ACUST UNITED AC 2006; 28:397-403. [PMID: 17051393 DOI: 10.1007/s00281-006-0054-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Accepted: 10/03/2006] [Indexed: 12/21/2022]
Abstract
Mucosal secretions of the human gastrointestinal, respiratory, and genital tracts contain significant quantities of IgG. The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. The FcRn can then recycle the IgG/antigen complex back across the intestinal barrier into the lamina propria for processing by dendritic cells and presentation to CD4(+) T cells in regional organized lymphoid structures. FcRn, through its ability to secrete and absorb IgG, thus integrates luminal antigen encounters with systemic immune compartments and, as such, provides essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Frontier Medical Science in Gastroenterology, ICMRT, Kobe University School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dall'Acqua WF, Kiener PA, Wu H. Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn). J Biol Chem 2006; 281:23514-24. [PMID: 16793771 DOI: 10.1074/jbc.m604292200] [Citation(s) in RCA: 461] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe here the functional implications of an increase in IgG binding to the neonatal Fc receptor. We have defined in a systematic fashion the relationship between enhanced FcRn binding of a humanized anti-respiratory syncytial virus (RSV) monoclonal antibody (MEDI-524) and the corresponding biological consequences in cynomolgus monkeys. The triple mutation M252Y/S254T/T256E (YTE) was introduced into the Fc portion of MEDI-524. Whereas these substitutions did not affect the ability of MEDI-524 to bind to its cognate antigen and inhibit RSV replication, they resulted in a 10-fold increase in its binding to both cynomolgus monkey and human FcRn at pH 6.0. MEDI-524-YTE was efficiently released from FcRn at pH 7.4 in both cases. We show that MEDI-524-YTE consistently exhibited a nearly 4-fold increase in serum half-life in cynomolgus monkeys when compared with MEDI-524. This constituted the largest half-life improvement described to date for an IgG in a primate. For the first time, we demonstrate that these sustained serum levels resulted in an up to 4-fold increase in lung bioavailability. Importantly, we also establish that our non-human primate model is relevant to human. Finally, we report that the YTE triple substitution provided a means to modulate the antibody-dependent cell-mediated cytotoxicity (ADCC) activity of a humanized IgG1 directed against the human integrin alpha(v)beta3. Therefore, the YTE substitutions allow the simultaneous modulation of serum half-life, tissue distribution and activity of a given human IgG1.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized
- Antibody Diversity
- Antibody-Dependent Cell Cytotoxicity
- Binding Sites, Antibody
- Half-Life
- Histocompatibility Antigens Class I/blood
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/chemistry
- Immunoglobulin G/metabolism
- Macaca fascicularis
- Organ Specificity
- Protein Binding
- Protein Engineering/methods
- Receptors, Fc/blood
- Receptors, Fc/metabolism
- Receptors, IgG/metabolism
Collapse
Affiliation(s)
- William F Dall'Acqua
- Department of Antibody Discovery and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland 20878, USA. dall'
| | | | | |
Collapse
|
19
|
Li N, Zhao M, Hilario-Vargas J, Prisayanh P, Warren S, Diaz LA, Roopenian DC, Liu Z. Complete FcRn dependence for intravenous Ig therapy in autoimmune skin blistering diseases. J Clin Invest 2005; 115:3440-50. [PMID: 16284651 PMCID: PMC1280965 DOI: 10.1172/jci24394] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 09/13/2005] [Indexed: 01/16/2023] Open
Abstract
Numerous mechanisms of action have been proposed for intravenous Ig (IVIG). In this study, we used IgG passive transfer murine models of bullous pemphigoid (BP), pemphigus foliaceus (PF), and pemphigus vulgaris (PV) to test the hypothesis that the effect of IVIG in autoantibody-mediated cutaneous bullous diseases is to accelerate the degradation of pathogenic IgG by saturation of the MHC-like Fc receptor neonatal Fc receptor (FcRn). BP, PF, and PV are organ-specific antibody-mediated diseases in which autoantibodies target the hemidesmosomal antigen BP180 and desmosomal antigens Dsg1 and Dsg3, respectively. Antibodies against BP180, Dsg1, and Dsg3, when injected into neonatal mice, induce the BP, PF, and PV disease phenotypes, respectively. We found that FcRn-deficient mice were resistant to experimental BP, PF, and PV. Circulating levels of pathogenic IgG in FcRn-deficient mice were significantly reduced compared with those in WT mice. Administration of high-dose human IgG (HDIG) to WT mice also drastically reduced circulating pathogenic IgG levels and prevented blistering. In FcRn-deficient mice, no additional protective effect with HDIG was realized. These data demonstrate that the therapeutic efficacy of HDIG treatment in the pemphigus and pemphigoid models is dependent on FcRn. Thus, FcRn is a promising therapeutic target for treating such IgG-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Ning Li
- Department of Dermatology, University of North Carolina at Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Claypool SM, Dickinson BL, Wagner JS, Johansen FE, Venu N, Borawski JA, Lencer WI, Blumberg RS. Bidirectional transepithelial IgG transport by a strongly polarized basolateral membrane Fcgamma-receptor. Mol Biol Cell 2004; 15:1746-59. [PMID: 14767057 PMCID: PMC379272 DOI: 10.1091/mbc.e03-11-0832] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Revised: 01/05/2004] [Accepted: 01/18/2004] [Indexed: 12/26/2022] Open
Abstract
The human MHC class I-related neonatal Fc receptor, hFcRn, mediates bidirectional transport of IgG across mucosal barriers. Here, we find that at steady state hFcRn distributes predominantly to an apical intracellular compartment and almost exclusively to the basolateral cell surface of polarized epithelial cells. It moves only transiently to the apical membrane. Ligand binding does not redistribute the steady state location of the receptor. Removal of the cytoplasmic tail that contains di-leucine and tryptophan-based endocytosis motifs or incubation at low temperature (18 degrees C) redistributes the receptor apically. The rates of endocytosis of the full-length hFcRn from the apical or basolateral membrane domains, however, are equal. Thus, the strong cell surface polarity displayed by hFcRn results from dominant basolateral sorting by motifs in the cytoplasmic tail that nonetheless allows for a cycle of bidirectional transcytosis.
Collapse
Affiliation(s)
- Steven M Claypool
- Harvard Medical School, Program in Immunology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Affiliation(s)
- Roald Nezlin
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
22
|
Dall'Acqua WF, Woods RM, Ward ES, Palaszynski SR, Patel NK, Brewah YA, Wu H, Kiener PA, Langermann S. Increasing the affinity of a human IgG1 for the neonatal Fc receptor: biological consequences. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5171-80. [PMID: 12391234 DOI: 10.4049/jimmunol.169.9.5171] [Citation(s) in RCA: 264] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Many biological functions, including control of the homeostasis and maternofetal transfer of serum gamma-globulins, are mediated by the MHC class I-related neonatal FcR (FcRn). A correlation exists in mice between the binding affinity of IgG1/Fc fragments to FcRn at pH 6.0 and their serum t(1/2). To expand this observation, phage display of mutagenized Fc fragments derived from a human IgG1 was used to increase their affinity to both murine and human FcRn. Ten variants were identified that have a higher affinity toward murine and human FcRn at pH 6.0, with DeltaDeltaG (DeltaG(wild type) - DeltaG(mutant)) from 1.0 to 2.0 kcal/mol and from 0.6 to 2.4 kcal/mol, respectively. Those variants exhibit a parallel increase in binding at pH 7.4 to murine, but not human, FcRn. Although not degraded in blood in vitro, accumulated in tissues, nor excreted in urine, their serum concentration in mice is decreased. We propose that higher affinity to FcRn at pH 7.4 adversely affects release into the serum and offsets the benefit of the enhanced binding at pH 6.0.
Collapse
MESH Headings
- Animals
- Animals, Newborn/genetics
- Animals, Newborn/immunology
- Bacteriophage M13/genetics
- Bacteriophage M13/immunology
- Binding Sites, Antibody/genetics
- Histocompatibility Antigens Class I
- Humans
- Hydrogen-Ion Concentration
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/genetics
- Immunoglobulin G/metabolism
- Infant, Newborn
- Injections, Intramuscular
- Injections, Intravenous
- Mice
- Mice, Inbred BALB C
- Mutagenesis, Site-Directed
- Organ Specificity/genetics
- Organ Specificity/immunology
- Peptide Library
- Precipitin Tests
- Receptors, Fc/administration & dosage
- Receptors, Fc/blood
- Receptors, Fc/genetics
- Receptors, Fc/metabolism
- Receptors, IgG/administration & dosage
- Receptors, IgG/blood
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Surface Plasmon Resonance
Collapse
|
23
|
Abstract
This review describes the evolution of our knowledge of the transmission of immunoglobulin G (IgG) from mother to infant and the factors which regulate the persistence of IgG in the circulation. These apparently unrelated processes involve the same Fc receptor, FcRn (n = neonatal). FcRn appears to carry out these diverse roles by binding to IgG and then either transporting the bound IgG across cells (transcytosis) or recycling its cargo back to the cell surface (control of catabolism). IgG that is taken up by cells in the absence of binding to FcRn undergoes degradation. Thus, FcRn is the "protective" receptor that serves to maintain IgG homeostasis and deliver IgGs across cellular barriers.
Collapse
Affiliation(s)
- Victor Ghetie
- Cancer Immunobiology Center, University of Texas Southwestern Medical Center, Dallas 75390-8576, USA.
| | | |
Collapse
|
24
|
Ober RJ, Radu CG, Ghetie V, Ward ES. Differences in promiscuity for antibody-FcRn interactions across species: implications for therapeutic antibodies. Int Immunol 2001; 13:1551-9. [PMID: 11717196 DOI: 10.1093/intimm/13.12.1551] [Citation(s) in RCA: 374] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Preclinical tests of therapeutic antibodies are frequently carried out in mice to evaluate pharmacokinetics and efficacy. However, the observation that mouse IgG are cleared rapidly from the human circulation suggests that mice may not always be an ideal model. The Fc receptor, FcRn, regulates the serum half-lives of IgG in mice and most likely has a similar function in humans. In the current study we have carried out an extensive analysis of the interaction of the human or mouse forms of FcRn with IgG from various species using surface plasmon resonance. We show that in contrast to mouse FcRn, human FcRn is surprisingly stringent in its binding specificity for IgG derived from different species. Human FcRn binds to human, rabbit and guinea pig IgG, but not significantly to rat, bovine, sheep or mouse IgG (with the exception of weak binding to mouse IgG2b). In contrast, mouse FcRn binds to all IgG analyzed. The lack of binding of human FcRn to mouse IgG1 has been confirmed using transfectants that have been engineered to express human FcRn on the cell surface. Our results provide a molecular explanation for the enigmatic observation that mouse IgG behave anomalously in humans. These studies have implications for the successful application of therapeutic antibodies.
Collapse
Affiliation(s)
- R J Ober
- Cancer Immunobiology Center and Center for Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-8576, USA
| | | | | | | |
Collapse
|
25
|
Firan M, Bawdon R, Radu C, Ober RJ, Eaken D, Antohe F, Ghetie V, Ward ES. The MHC class I-related receptor, FcRn, plays an essential role in the maternofetal transfer of gamma-globulin in humans. Int Immunol 2001; 13:993-1002. [PMID: 11470769 DOI: 10.1093/intimm/13.8.993] [Citation(s) in RCA: 241] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The transfer of maternal gamma-globulin (IgG) provides the neonate with humoral immunity during early life. In humans, maternal IgG is transported across the placenta during the third trimester of pregnancy. The expression of the MHC class I-related receptor, FcRn, in the human placenta suggests that this Fc receptor might be involved in the delivery of maternal IgG, but direct evidence to support this is lacking. In the current study an ex vivo placental model has been used to analyze the maternofetal transfer of a recombinant, humanized (IgG1) antibody in which His435 has been mutated to alanine (H435A). In vitro binding studies using surface plasmon resonance indicate that the mutation ablates binding of the antibody to recombinant mouse and human FcRn. Relative to the wild-type antibody, the H435A mutant is deficient in transfer across the placenta. Significantly, the mutation does not affect binding to Fc gamma RIII, an FcR that has been suggested in earlier studies to mediate the transfer of maternal IgG. The analyses demonstrate that binding of an IgG to FcRn is a prerequisite for transport across the perfused placenta. FcRn therefore plays a central role in the maternofetal delivery of IgG and this has implications for the use of protein engineering to improve the properties of therapeutic antibodies.
Collapse
Affiliation(s)
- M Firan
- Center for Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Tizard I. The protective properties of milk and colostrum in non-human species. ADVANCES IN NUTRITIONAL RESEARCH 2001; 10:139-66. [PMID: 11795038 DOI: 10.1007/978-1-4615-0661-4_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Affiliation(s)
- I Tizard
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
27
|
Ghetie V, Ward ES. Multiple roles for the major histocompatibility complex class I- related receptor FcRn. Annu Rev Immunol 2000; 18:739-66. [PMID: 10837074 DOI: 10.1146/annurev.immunol.18.1.739] [Citation(s) in RCA: 361] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Multiple functions have recently been identified for the neonatal Fc receptor FcRn. In addition, a human homolog of the rodent forms of FcRn has been identified and characterized. This major histocompatibility complex class I-related receptor plays a role in the passive delivery of immunoglobulin (Ig)Gs from mother to young and the regulation of serum IgG levels. In addition, FcRn expression in tissues such as liver, mammary gland, and adult intestine suggests that it may modulate IgG transport at these sites. These diverse functions are apparently brought about by the ability of FcRn to bind IgGs and transport them within and across cells. However, the molecular details as to how FcRn traffics within cells have yet to be fully understood, although in vitro systems have been developed for this purpose. The molecular nature of the FcRn-IgG interaction has been studied extensively and encompasses residues located at the CH2-CH3 domain interface of the Fc region of IgG. These Fc amino acids are highly conserved in rodents and man and interact with residues primarily located on the alpha2 domain of FcRn. Thus, it is now possible to engineer IgGs with altered affinities for FcRn, and this has relevance to the modulation of IgG serum half-life and maternofetal IgG transport for therapeutic applications.
Collapse
Affiliation(s)
- V Ghetie
- Center for Immunology and Cancer Immunobiology Center, University of Texas Southwestern Medical Center, Dallas, Texas 75235-8576, USA
| | | |
Collapse
|
28
|
Affiliation(s)
- B K Flesch
- Institute of Transfusion Medicine, Christian Albrechts University of Kiel, Germany.
| | | |
Collapse
|
29
|
Abstract
The analysis of the pharmacokinetics of wild-type and mutated Fc fragments derived from human IgG1 indicates that Ile253, His310 and His435 play a central role in regulating serum half-life in mice. Reduced serum half-life of the recombinant, mutated fragments correlates with decreased binding to the MHC class I-related neonatal Fc receptor, FcRn. In addition, the analysis of an Fc fragment in which His435 is mutated to Arg435 demonstrates that the sequence difference at this position between human IgG1 (His435) and IgG3 (Arg435) most likely accounts for the shorter serum half-life of IgG3 relative to IgG1. In contrast to His310 and His435, the data indicate that His433 does not play a role in regulating the serum half-life of human IgG1. Thus, the interaction site of mouse FcRn on human and mouse IgG1 involves the same conserved amino acids located at the CH2-CH3 domain interface of the IgG molecule. The sequence similarities between mouse and human FcRn suggest that these studies have direct relevance to understanding the factors that govern the pharmacokinetics of therapeutic IgG.
Collapse
Affiliation(s)
- J K Kim
- Department of Microbiology, Changwon National University, Changwon, Kyungnam, South Korea
| | | | | | | | | | | |
Collapse
|
30
|
Praetor A, Ellinger I, Hunziker W. Intracellular traffic of the MHC class I-like IgG Fc receptor, FcRn, expressed in epithelial MDCK cells. J Cell Sci 1999; 112 ( Pt 14):2291-9. [PMID: 10381385 DOI: 10.1242/jcs.112.14.2291] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transfer of passive immunity from mother to the fetus or newborn involves the transport of IgG across several epithelia. Depending on the species, IgG is transported prenatally across the placenta and yolk sac or is absorbed from colostrum and milk by the small intestine of the suckling newborn. In both cases apical to basolateral transepithelial transport of IgG is thought to be mediated by FcRn, an IgG Fc receptor with homology to MHC class I antigens. We have now expressed the human FcRn in polarized MDCK cells and analyzed the intracellular routing of the receptor. FcRn showed a predominant intracellular localization at steady state. Newly synthesized FcRn was delivered in a non-vectorial fashion to both the apical and basolateral surfaces of MDCK cell monolayers. Following internalization from the apical or basolateral domain, the receptor transcytosed to the opposite surface. These findings provide direct evidence for the transepithelial transport function of FcRn and indicate that the receptor undergoes multiple rounds of transcytosis.
Collapse
Affiliation(s)
- A Praetor
- Institute of Biochemistry, University of Lausanne, BIL Biomedical Research Center, 1066 Epalinges, Switzerland
| | | | | |
Collapse
|
31
|
Ellinger I, Schwab M, Stefanescu A, Hunziker W, Fuchs R. IgG transport across trophoblast-derived BeWo cells: a model system to study IgG transport in the placenta. Eur J Immunol 1999; 29:733-44. [PMID: 10092075 DOI: 10.1002/(sici)1521-4141(199903)29:03<733::aid-immu733>3.0.co;2-c] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In primates, prenatal transfer of IgG from mother to offspring occurs predominantly across the placenta. Although a number of Fcgamma-receptors and IgG binding proteins have been detected in human placental tissue, an involvement of any of these receptors in IgG transport across the syncytiotrophoblast remains to be demonstrated. Therefore, we investigated the mechanism of IgG transcytosis in trophoblast-derived BeWo cells. BeWo cells were not only found to express the MHC class I-related IgG Fc receptor, human FcRn, but also specifically bound fluorescein isothiocyanate (FITC)-labeled human IgG (FITC-hIgG) at the apical surface at mildly acidic pH. The cells preferentially transcytosed FITC-hIgG from the apical to the basolateral side when compared to the fluid-phase marker FITC-dextran and to FITC-hIgG transcytosis in the opposite direction. However, endocytosis of FITC-hIgG at the apical plasma membrane at physiological pH required the continuous presence of FITC-hIgG at concentrations similar to those present in the maternal circulation. These results suggest a mechanism by which IgG is internalized by BeWo cells via fluid-phase endocytosis. Tight binding of IgG to hFcRn may then occur in acidic endosomes, followed by selective sorting into the transcytotic pathway. Thus, the main function of this receptor is to prevent entry of IgG into the degradative pathway in lysosomes.
Collapse
Affiliation(s)
- I Ellinger
- Department of General and Experimental Pathology, University of Vienna, Austria
| | | | | | | | | |
Collapse
|
32
|
Abstract
Transcytosis plays a central role in the immunological functions of epithelia, including the sampling of antigens that enter the body via the digestive, respiratory and urogenital tracts and their presentation to underlying lymphoid tissues, the secretion of specific immunoglobulins required for the immune protection of mucosal surfaces and the transfer of maternal immunoglobulins to the fetus or newborn, providing the latter with passive immunity for the first weeks of independent life.
Collapse
Affiliation(s)
- W Hunziker
- Institute of Biochemistry, University of Lausanne, BIL Research Center, Epalinges, Switzerland.
| | | |
Collapse
|
33
|
Medesan C, Cianga P, Mummert M, Stanescu D, Ghetie V, Ward ES. Comparative studies of rat IgG to further delineate the Fc:FcRn interaction site. Eur J Immunol 1998; 28:2092-100. [PMID: 9692877 DOI: 10.1002/(sici)1521-4141(199807)28:07<2092::aid-immu2092>3.0.co;2-e] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent data have indicated that the MHC class I-related receptor, FcRn, regulates the half-lives of serum IgG in addition to its known role in transferring IgG from mother to young. In the current study, the activity of rat IgG (rIgG) isotypes in FcRn-mediated functions has been analyzed. The serum half-life and maternofetal transfer in mice decreased in the order rIgG2a > rIgG1 > rIgG2c > rIgG2b. This decrease in activity correlates well with reduced binding affinity for soluble mouse FcRn, and site-directed mutagenesis of a recombinant Fc-hinge fragment has been used to investigate the molecular basis for the differences in activities of the rIgG. Analysis of the serum half-lives of the mutated Fc-hinge fragments demonstrated that, in addition to Ile253, His310, His435 and His436 that were identified in earlier studies, amino acids at positions 257, 307 and 309 play a role in building the FcRn interaction site of IgG. The study also excludes the involvement of amino acids in a fourth loop located at the CH2-CH3 domain interface that encompasses residues 386-387 in FcRn binding. Sequence differences at positions 257, 307 and 309 between rIgG most likely account for the reduced affinity of rIgG2b and IgG2c relative to rIgG1 and rIgG2a for binding to FcRn.
Collapse
Affiliation(s)
- C Medesan
- Department of Microbiology and Cancer Immunobiology Center, University of Texas Southwestern Medical Center, Dallas 75235-8576, USA
| | | | | | | | | | | |
Collapse
|
34
|
Swaan PW. Recent advances in intestinal macromolecular drug delivery via receptor-mediated transport pathways. Pharm Res 1998; 15:826-34. [PMID: 9647346 DOI: 10.1023/a:1011908128045] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Receptor-mediated transport mechanisms provide a pathway for the trafficking of extracellular macromolecules into (endocytosis) and across (transcytosis) the cell. This comprises the binding of a ligand to a specific cell-surface receptor, clustering of the ligand-receptor complexes in endocytotic vesicles and vesicular sorting. This review focuses on recent advances in cellular and molecular biology pertaining to receptor-mediated endocytosis. A concise overview is presented of current and potential future applications of targeting to RME mechanisms to improve oral macromolecular drug delivery.
Collapse
Affiliation(s)
- P W Swaan
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus 43210, USA.
| |
Collapse
|
35
|
Ghetie V, Ward ES. FcRn: the MHC class I-related receptor that is more than an IgG transporter. IMMUNOLOGY TODAY 1997; 18:592-8. [PMID: 9425738 DOI: 10.1016/s0167-5699(97)01172-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recent data have indicated that the major histocompatibility complex (MHC) class I-related receptor FcRn plays a role in regulating serum IgG levels, in addition to transferring maternal IgGs across the rodent neonatal gut. The isolation of a human homolog of FcRn from placenta suggests that the studies in rodents have relevance to understanding similar processes in humans. This has implications for the engineering of improved antibodies for therapy.
Collapse
Affiliation(s)
- V Ghetie
- Cancer Immunobiology Center, University of Texas Southwestern Medical Center, Dallas 75235-8576, USA
| | | |
Collapse
|
36
|
Major AS, Cuff CF. Enhanced mucosal and systemic immune responses to intestinal reovirus infection in beta2-microglobulin-deficient mice. J Virol 1997; 71:5782-9. [PMID: 9223466 PMCID: PMC191832 DOI: 10.1128/jvi.71.8.5782-5789.1997] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Enteric infection of mice with respiratory enteric orphan virus (reovirus) type 1, strain Lang elicits both humoral and cellular immune responses. To investigate the role of CD8+, alpha/beta T-cell receptor (TCR)+ T cells in mucosal immunity to an enteric pathogen, we examined immune responses and viral clearance following enteric reovirus infection in C57BL/6, B6129F2, and beta2-microglobulin-deficient (beta2m-/-) mice. Analysis of Peyer's patch and lamina propria culture supernatants revealed a two- to threefold increase in levels of reovirus-specific immunoglobulin A in beta2m-/- mice compared to normal controls. These data corresponded to a similar increase in the frequency of virus-specific immunoglobulin A-producing cells in Peyer's patches and lamina propria and an increase in immunoglobulin G-producing cells in spleens from beta2m-/- mice compared to controls. These increased humoral immune responses were not due to a difference in B-cell populations because cell counts and flow cytometric analyses showed that beta2m-/- and control mice had similar numbers and percentages of B cells in mucosal and systemic tissues. Analysis of cytokine message by reverse transcriptase-PCR 5 and 10 days after infection revealed no difference in message level for transforming growth factor beta, gamma interferon, interleukin-4, interleukin-5, or interleukin-6 for all mouse strains. Virus tissue titers determined by plaque assay at 5 and 10 days after infection demonstrated that beta2m-/- mice cleared reovirus from the small intestines with the same efficiency as control mice. Collectively, these data suggest that CD8+, alpha/beta TCR+ T cells may regulate mucosal and systemic humoral immune responses to oral infection with reovirus.
Collapse
Affiliation(s)
- A S Major
- Department of Microbiology and Immunology, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown 26506, USA
| | | |
Collapse
|
37
|
Ghetie V, Popov S, Borvak J, Radu C, Matesoi D, Medesan C, Ober RJ, Ward ES. Increasing the serum persistence of an IgG fragment by random mutagenesis. Nat Biotechnol 1997; 15:637-40. [PMID: 9219265 DOI: 10.1038/nbt0797-637] [Citation(s) in RCA: 184] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The major histocompatibility complex (MHC) class I-related receptor FcRn is involved in regulating serum gammaglobulin (IgG) levels in mice. With the aim of increasing the serum half-life of a recombinant murine Fc gamma 1 fragment, the affinity for binding to FcRn at pH 6.0 has been increased by random mutagenesis of Thr252, Thr254, and Thr256 followed by selection using bacteriophage display. These residues were chosen as they are in proximity to the FcRn-IgG (Fc) interaction site. Two mutants with higher affinity (due to lower off-rates) than the wild-type Fc have been isolated and analyzed in pharmacokinetic studies in mice. The mutant with the highest affinity has a significantly longer serum half-life than the wild type fragment, despite its lower off-rate from FcRn at pH 7.4. The results provide support for the involvement of FcRn in the homeostasis of serum IgGs in mice. The indications that a homologous FcRn regulates IgG levels in humans suggest that this approach has implications for increasing the serum persistence of therapeutic antibodies.
Collapse
Affiliation(s)
- V Ghetie
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas 75235-8576, USA
| | | | | | | | | | | | | | | |
Collapse
|