201
|
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: 48] [Impact Index Per Article: 4.8] [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
|
202
|
Cui D, Zhang L, Li J, Zhao Y, Hu X, Dai Y, Zhang R, Li N. Bovine FcRn-mediated human immunoglobulin G transfer across the milk-blood barrier in transgenic mice. PLoS One 2014; 9:e115972. [PMID: 25546424 PMCID: PMC4278800 DOI: 10.1371/journal.pone.0115972] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/28/2014] [Indexed: 11/18/2022] Open
Abstract
Maternal-fetal IgGs transport occurs either prenatally or postnatally, which confers the newborns with passive immunity before their own immune system has matured. However, little is known about the mechanisms of postnatal IgGs passage in the mammary gland. To investigate how FcRn mediates the IgGs transport in the mammary gland, we first generated bFcRn and anti-HAV mAb transgenic mice, and then obtained HF transgenic mice expressing both transgenes by mating the above two strains. Transgene expression of bFcRn in the four lines was determined by qRT-PCR and western blot. We then localized the expression of bFcRn to the acinar epithelial cells in the mammary gland, and anti-HAV mAb was mainly detected in the acini with weak staining in the acinar epithelial cells. Human IgGs could be detected in both milk and serum of HF transgenic mice by western blot and ELISA. A significantly lower milk to serum ratio of human IgGs in HF mice compared with that of anti-HAV mAb mice, indicating that bFcRn could transport human IgGs across the milk-blood barrier from milk to serum during lactation in HF mice. While, there were no transport of murine IgGs, IgAs, or IgMs. These results provide understandings about the mechanisms of maternal-fetal immunity transfer in the mammary gland.
Collapse
Affiliation(s)
- Dan Cui
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
| | - Linlin Zhang
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
| | - Jia Li
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
| | - Yaofeng Zhao
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
| | - Xiaoxiang Hu
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
| | | | - Ran Zhang
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
- * E-mail: (RZ); (NL)
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, China
- * E-mail: (RZ); (NL)
| |
Collapse
|
203
|
Pike AF, Kramer NI, Blaauboer BJ, Seinen W, Brands R. An alkaline phosphatase transport mechanism in the pathogenesis of Alzheimer's disease and neurodegeneration. Chem Biol Interact 2014; 226:30-9. [PMID: 25500268 DOI: 10.1016/j.cbi.2014.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/23/2014] [Accepted: 12/03/2014] [Indexed: 12/19/2022]
Abstract
Systemic inflammation is associated with loss of blood-brain barrier integrity and neuroinflammation that lead to the exacerbation of neurodegenerative diseases. It is also associated specifically with the characteristic amyloid-β and tau pathologies of Alzheimer's disease. We have previously proposed an immunosurveillance mechanism for epithelial barriers involving negative feedback-regulated alkaline phosphatase transcytosis as an acute phase anti-inflammatory response that hangs in the balance between the resolution and the progression of inflammation. We now extend this model to endothelial barriers, particularly the blood-brain barrier, and present a literature-supported mechanistic explanation for Alzheimer's disease pathology with this system at its foundation. In this mechanism, a switch in the role of alkaline phosphatase from its baseline duties to a stopgap anti-inflammatory function results in the loss of alkaline phosphatase from cell membranes into circulation, thereby decreasing blood-brain barrier integrity and functionality. This occurs with impairment of both amyloid-β efflux and tau dephosphorylating activity in the brain as alkaline phosphatase is replenished at the barrier by receptor-mediated transport. We suggest systemic alkaline phosphatase administration as a potential therapy for the resolution of inflammation and the prevention of Alzheimer's disease pathology as well as that of other inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Adrianne F Pike
- AMRIF B.V., Agro Business Park 10, 6708PW Wageningen, The Netherlands.
| | - Nynke I Kramer
- Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80177, 3508TD Utrecht, The Netherlands
| | - Bas J Blaauboer
- Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80177, 3508TD Utrecht, The Netherlands
| | - Willem Seinen
- AMRIF B.V., Agro Business Park 10, 6708PW Wageningen, The Netherlands; Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80177, 3508TD Utrecht, The Netherlands
| | - Ruud Brands
- AMRIF B.V., Agro Business Park 10, 6708PW Wageningen, The Netherlands; Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80177, 3508TD Utrecht, The Netherlands
| |
Collapse
|
204
|
Berger M, McCallus DE, Lin CSY. Rapid and reversible responses to IVIG in autoimmune neuromuscular diseases suggest mechanisms of action involving competition with functionally important autoantibodies. J Peripher Nerv Syst 2014; 18:275-96. [PMID: 24200120 PMCID: PMC4285221 DOI: 10.1111/jns5.12048] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intravenous immunoglobulin (IVIG) is widely used in autoimmune neuromuscular diseases whose pathogenesis is undefined. Many different effects of IVIG have been demonstrated in vitro, but few studies actually identify the mechanism(s) most important in vivo. Doses and treatment intervals are generally chosen empirically. Recent studies in Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy show that some effects of IVIG are readily reversible and highly dependent on the serum IgG level. This suggests that in some autoantibody-mediated neuromuscular diseases, IVIG directly competes with autoantibodies that reversibly interfere with nerve conduction. Mechanisms of action of IVIG which most likely involve direct competition with autoantibodies include: neutralization of autoantibodies by anti-idiotypes, inhibition of complement deposition, and increasing catabolism of pathologic antibodies by saturating FcRn. Indirect immunomodulatory effects are not as likely to involve competition and may not have the same reversibility and dose-dependency. Pharmacodynamic analyses should be informative regarding most relevant mechanism(s) of action of IVIG as well as the role of autoantibodies in the immunopathogenesis of each disease. Better understanding of the role of autoantibodies and of the target(s) of IVIG could lead to more efficient use of this therapy and better patient outcomes.
Collapse
Affiliation(s)
- Melvin Berger
- Departments of Pediatrics and Pathology, Case Western Reserve University, Cleveland, OH, USA; Immunology Research and Development, CSL Behring, LLC, King of Prussia, PA, USA
| | | | | |
Collapse
|
205
|
Buyue Y, Liu T, Kulman JD, Toby GG, Kamphaus GD, Patarroyo-White S, Lu Q, Reidy TJ, Mei B, Jiang H, Pierce GF, Sommer JM, Peters RT. A single chain variant of factor VIII Fc fusion protein retains normal in vivo efficacy but exhibits altered in vitro activity. PLoS One 2014; 9:e113600. [PMID: 25415306 PMCID: PMC4240654 DOI: 10.1371/journal.pone.0113600] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/25/2014] [Indexed: 11/19/2022] Open
Abstract
Recombinant factor VIII Fc (rFVIIIFc) is a fusion protein consisting of a single B-domain-deleted (BDD) FVIII linked recombinantly to the Fc domain of human IgG1 to extend half-life. To determine if rFVIIIFc could be further improved by maintaining the heavy and light chains within a contiguous single chain (SC), we evaluated the activity and function of SC rFVIIIFc, an isoform that is not processed at residue R1648. SC rFVIIIFc showed equivalent activity in a chromogenic assay compared to rFVIIIFc, but approximately 40% activity by the one-stage clotting assay in the presence of von Willebrand Factor (VWF), with full activity in the absence of VWF. Moreover, SC rFVIIIFc demonstrated markedly delayed thrombin-mediated release from VWF, but an activity similar to that of rFVIIIFc upon activation in FXa generation assays. Therefore, the apparent reduction in specific activity in the aPTT assay appears to be primarily due to delayed release of FVIII from VWF. To assess whether stability and activity of SC rFVIIIFc were affected in vivo, a tail vein transection model in Hemophilia A mice was utilized. The results demonstrated similar pharmacokinetic profiles and comparable efficacy for SC rFVIIIFc and rFVIIIFc. Thus, while the single chain configuration did not promote enhanced half-life, it reduced the rate of release of FVIII from VWF required for activation. This impaired release may underlie the observed reduction in the one-stage clotting assay, but does not appear to affect the physiological activity of SC rFVIIIFc.
Collapse
Affiliation(s)
- Yang Buyue
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Tongyao Liu
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - John D. Kulman
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Garabet G. Toby
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - George D. Kamphaus
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | | | - Qi Lu
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Thomas J. Reidy
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Baisong Mei
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Haiyan Jiang
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Glenn F. Pierce
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Jurg M. Sommer
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Robert T. Peters
- Hematology Research, Biogen Idec, Cambridge, Massachusetts, United States of America
| |
Collapse
|
206
|
Fathallah AM, Balu-Iyer SV. Anatomical, physiological, and experimental factors affecting the bioavailability of sc-administered large biotherapeutics. J Pharm Sci 2014; 104:301-6. [PMID: 25411114 DOI: 10.1002/jps.24277] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 01/06/2023]
Abstract
Subcutaneous route of administration is highly desirable for protein therapeutics. It improves patient compliance and quality of life (McDonald TA, Zepeda ML, Tomlinson MJ, Bee WH, Ivens IA. 2010. Curr Opin Mol Ther 12(4):461-470; Dychter SS, Gold DA, Haller MF. 2012. J Infus Nurs 35(3):154-160), while reducing healthcare cost (Dychter SS, Gold DA, Haller MF. 2012. J Infus Nurs 35(3):154-160). Recent evidence also suggests that sc administration of protein therapeutics can increase tolerability to some treatments such as intravenous immunoglobulin therapy by administering it subcutaneously (subcutaneous immunoglobulin therapy SCIG), which will reduce fluctuation in plasma drug concentration (Kobrynski L. 2012. Biologics 6:277-287). Furthermore, sc administration may reduce the risk of systemic infections associated with i.v. infusion (McDonald TA, Zepeda ML, Tomlinson MJ, Bee WH, Ivens IA. 2010. Curr Opin Mol Ther 12(4):461-470; Dychter SS, Gold DA, Haller MF. 2012. J Infus Nurs 35(3):154-160). This route, however, has its challenges, especially for large multidomain proteins. Poor bioavailability and poor scalability from preclinical models are often cited. This commentary will discuss barriers to sc absorption as well as physiological and experimental factors that could affect pharmacokinetics of subcutaneously administered large protein therapeutics in preclinical models. A mechanistic pharmacokinetic model is proposed as a potential tool to address the issue of scalability of sc pharmacokinetic from preclinical models to humans.
Collapse
Affiliation(s)
- Anas M Fathallah
- Department of Pharmaceutical Sciences, University of Buffalo, Buffalo, New York
| | | |
Collapse
|
207
|
An engineered affibody molecule with pH-dependent binding to FcRn mediates extended circulatory half-life of a fusion protein. Proc Natl Acad Sci U S A 2014; 111:17110-5. [PMID: 25406323 DOI: 10.1073/pnas.1417717111] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Proteins endocytosed from serum are degraded in the lysosomes. However, serum albumin (SA) and IgG, through its Fc part, bind to the neonatal Fc receptor (FcRn) at low pH in the endosome after endocytosis, and are transported back to the cellular surface, where they are released into the bloodstream, resulting in an extended serum circulation time. Association with Fc or SA has been used to prolong the in vivo half-life of biopharmaceuticals, using the interaction with FcRn to improve treatment regimens. This has been achieved either directly, by fusion or conjugation to Fc or SA, or indirectly, using SA-binding proteins. The present work takes this principle one step further, presenting small affinity proteins that bind directly to FcRn, mediating extension of the serum half-life of fused biomolecules. Phage display technology was used to select affibody molecules that can bind to FcRn in the pH-dependent manner required for rescue by FcRn. The biophysical and binding properties were characterized in vitro, and the affibody molecules were found to bind to FcRn more strongly at low pH than at neutral pH. Attachment of the affibody molecules to a recombinant protein, already engineered for increased half-life, resulted in a nearly threefold longer half-life in mice. These tags should have general use as fusion partners to biopharmaceuticals to extend their half-lives in vivo.
Collapse
|
208
|
Kagan L. Pharmacokinetic modeling of the subcutaneous absorption of therapeutic proteins. Drug Metab Dispos 2014; 42:1890-905. [PMID: 25122564 DOI: 10.1124/dmd.114.059121] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Subcutaneous injection is an important route of administration for therapeutic proteins that provides several advantages over other modes of parenteral delivery. Despite extensive clinical use, the exact mechanism underlying subcutaneous absorption of proteins is not completely understood, and the accuracy of prediction of absorption of biotherapeutics in humans remains unsatisfactory. This review summarizes a variety of models that have been developed for describing the pharmacokinetics of therapeutic proteins administered by subcutaneous injection, including single- and dual-pathway absorption models. Modeling of the lymphatic uptake and redistribution, absorption of monoclonal antibodies and insulin, and population analysis of protein absorption are discussed. The review also addresses interspecies modeling and prediction of absorption in humans, highlights important factors affecting the absorption processes, and suggests approaches for future development of mechanism-based absorption models.
Collapse
Affiliation(s)
- Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
209
|
Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol 2014; 5:520. [PMID: 25368619 PMCID: PMC4202688 DOI: 10.3389/fimmu.2014.00520] [Citation(s) in RCA: 1799] [Impact Index Per Article: 163.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/06/2014] [Indexed: 12/21/2022] Open
Abstract
Of the five immunoglobulin isotypes, immunoglobulin G (IgG) is most abundant in human serum. The four subclasses, IgG1, IgG2, IgG3, and IgG4, which are highly conserved, differ in their constant region, particularly in their hinges and upper CH2 domains. These regions are involved in binding to both IgG-Fc receptors (FcγR) and C1q. As a result, the different subclasses have different effector functions, both in terms of triggering FcγR-expressing cells, resulting in phagocytosis or antibody-dependent cell-mediated cytotoxicity, and activating complement. The Fc-regions also contain a binding epitope for the neonatal Fc receptor (FcRn), responsible for the extended half-life, placental transport, and bidirectional transport of IgG to mucosal surfaces. However, FcRn is also expressed in myeloid cells, where it participates in both phagocytosis and antigen presentation together with classical FcγR and complement. How these properties, IgG-polymorphisms and post-translational modification of the antibodies in the form of glycosylation, affect IgG-function will be the focus of the current review.
Collapse
Affiliation(s)
- Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Gillian Dekkers
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| |
Collapse
|
210
|
Schweizer D, Serno T, Goepferich A. Controlled release of therapeutic antibody formats. Eur J Pharm Biopharm 2014; 88:291-309. [DOI: 10.1016/j.ejpb.2014.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 06/30/2014] [Accepted: 08/03/2014] [Indexed: 10/24/2022]
|
211
|
Gao X, Ji JA, Veeravalli K, Wang YJ, Zhang T, Mcgreevy W, Zheng K, Kelley RF, Laird MW, Liu J, Cromwell M. Effect of individual Fc methionine oxidation on FcRn binding: Met252 oxidation impairs FcRn binding more profoundly than Met428 oxidation. J Pharm Sci 2014; 104:368-77. [PMID: 25175600 DOI: 10.1002/jps.24136] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/07/2014] [Accepted: 07/29/2014] [Indexed: 01/17/2023]
Abstract
The long serum half-lives of mAbs are conferred by pH-dependent binding of IgG-Fc to the neonatal Fc receptor (FcRn). The Fc region of human IgG1 has three conserved methionine residues, Met252, Met358, and Met428. Recent studies showed oxidation of these Met residues impairs FcRn binding and consequently affects pharmacokinetics of therapeutic antibodies. However, the quantitative effect of individual Met oxidation on Fc-FcRn binding has not been addressed. This information is valuable for defining critical quality attributes. In the present study, two sets of homodimeric site-directed IgG1 mutations were generated to understand how individual Fc Met oxidation affects FcRn binding. The first approach used Met to Leu mutants to block site-specific Met oxidation. In the other approach, Met to Gln mutants were designed to mimic site-specific Met oxidation. Both mutagenesis approaches show that either Met252 or Met428 oxidation alone significantly impairs Fc-FcRn binding. Met252 oxidation has a more deleterious effect on FcRn binding than M428 oxidation, whereas Met428 oxidation has a bigger destabilization effect on the thermal stability. Our results also show that Met358 oxidation does not affect FcRn binding. In addition, our study suggests that Met to Gln mutation may serve as an important tool to understand Met oxidation.
Collapse
Affiliation(s)
- Xuan Gao
- Late Stage Pharmaceutical Development, Genentech, South San Francisco, California, 94080
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Abstract
The main purpose of treating diabetes is to prevent the onset and the progression of diabetic chronic complications. Since the mechanism of onset of chronic complications is still not well understood, the main strategy to achieve this purpose is to bring the plasma glucose level in diabetic patients as close as possible to that in healthy subjects and try to maintain good glycemic control over the long term. Glycated hemoglobin (HbA1c), glycated albumin (GA), fructosamine, and 1,5-anhydroglucitol (1,5 AG) are used for evaluating glycemic control. At present, HbA1c is widely used as a gold standard index for glycemic control in clinical practice. While HbA1c reflects the long-term glycemic control state (for the past 1-2 months), it does not accurately reflect glycemic control in the clinical state in which glycemic control improves or deteriorates in the short-term. It is also known that HbA1c in patients with hematological disorders such as anemia and variant hemoglobin shows an abnormal value. In addition, HbA1c mainly reflects the mean plasma glucose but does not reflect the postprandial plasma glucose. On the other hand, GA and 1,5-AG reflect intermediate- or short-term glycemic control and are not influenced by hemoglobin metabolism. While 1,5-AG is known to reflect the postprandial plasma glucose, it was shown recently that GA also reflects the postprandial plasma glucose. This chapter summarizes the measurement methods, usage methods, evidence, and problems concerning such indices for glycemic control.
Collapse
|
213
|
Haraya K, Tachibana T, Nanami M, Ishigai M. Application of human FcRn transgenic mice as a pharmacokinetic screening tool of monoclonal antibody. Xenobiotica 2014; 44:1127-34. [PMID: 25030041 DOI: 10.3109/00498254.2014.941963] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
1. For drug discovery, useful screening tools are essential to select superior candidates. Here, we evaluated the applicability of transgenic mice expressing human neonatal Fc receptor (FcRn) (hFcRn Tgm) as a pharmacokinetic screening tool of therapeutic monoclonal antibodies (mAbs) and Fc-fusion proteins that overcomes the species difference in FcRn binding. 2. Marketed 11 mAbs and 2 Fc-fusion proteins were intravenously administered to hFcRn Tgm and WT mice. The half-lives in hFcRn Tgm and WT mice were compared with those in human obtained from literature. The linear half-lives in human and monkey were also calculated by nonlinear pharmacokinetic analysis. For comparison, correlations of half-lives between monkey and human were also evaluated. 3. The half-lives of mAbs and Fc-fusion proteins after intravenous administration ranged from 1.1 to 13.2 days in hFcRn Tgm and from 1.2 to 30.3 days in WT mice. The half-lives in human correlated more closely with those in hFcRn Tgm than in WT mice and monkey. 4. Our results suggest that hFcRn Tgm are a valuable and useful tool for pharmacokinetic screening of mAbs and Fc-fusion proteins in the preclinical stage. Furthermore, we believe that hFcRn Tgm are broadly applicable to preclinical pharmacokinetic screening of mAbs-based therapeutics.
Collapse
Affiliation(s)
- Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd. , Singapore and
| | | | | | | |
Collapse
|
214
|
Faucette AN, Unger BL, Gonik B, Chen K. Maternal vaccination: moving the science forward. Hum Reprod Update 2014; 21:119-35. [PMID: 25015234 DOI: 10.1093/humupd/dmu041] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Infections remain one of the leading causes of morbidity in pregnant women and newborns, with vaccine-preventable infections contributing significantly to the burden of disease. In the past decade, maternal vaccination has emerged as a promising public health strategy to prevent and combat maternal, fetal and neonatal infections. Despite a number of universally recommended maternal vaccines, the development and evaluation of safe and effective maternal vaccines and their wide acceptance are hampered by the lack of thorough understanding of the efficacy and safety in the pregnant women and the offspring. METHODS An outline was synthesized based on the current status and major gaps in the knowledge of maternal vaccination. A systematic literature search in PUBMED was undertaken using the key words in each section title of the outline to retrieve articles relevant to pregnancy. Articles cited were selected based on relevance and quality. On the basis of the reviewed information, a perspective on the future directions of maternal vaccination research was formulated. RESULTS Maternal vaccination can generate active immune protection in the mother and elicit systemic immunoglobulin G (IgG) and mucosal IgG, IgA and IgM responses to confer neonatal protection. The maternal immune system undergoes significant modulation during pregnancy, which influences responsiveness to vaccines. Significant gaps exist in our knowledge of the efficacy and safety of maternal vaccines, and no maternal vaccines against a large number of old and emerging pathogens are available. Public acceptance of maternal vaccination has been low. CONCLUSIONS To tackle the scientific challenges of maternal vaccination and to provide the public with informed vaccination choices, scientists and clinicians in different disciplines must work closely and have a mechanistic understanding of the systemic, reproductive and mammary mucosal immune responses to vaccines. The use of animal models should be coupled with human studies in an iterative manner for maternal vaccine experimentation, evaluation and optimization. Systems biology approaches should be adopted to improve the speed, accuracy and safety of maternal vaccine targeting.
Collapse
Affiliation(s)
- Azure N Faucette
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI 48201, USA
| | - Benjamin L Unger
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI 48201, USA
| | - Bernard Gonik
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Kang Chen
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI 48201, USA Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA Department of Immunology and Microbiology, Wayne State University, Detroit, MI 48201, USA Department of Oncology, Wayne State University, Detroit, MI 48201, USA Mucosal Immunology Studies Team, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
215
|
Feng Z, Hirai-Yuki A, McKnight KL, Lemon SM. Naked Viruses That Aren't Always Naked: Quasi-Enveloped Agents of Acute Hepatitis. Annu Rev Virol 2014; 1:539-60. [PMID: 26958733 DOI: 10.1146/annurev-virology-031413-085359] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Historically, viruses were considered to be either enveloped or nonenveloped. However, recent work on hepatitis A virus and hepatitis E virus challenges this long-held tenet. Whereas these human pathogens are shed in feces as naked nonenveloped virions, recent studies indicate that both circulate in the blood completely masked in membranes during acute infection. These membrane-wrapped virions are as infectious as their naked counterparts, although they do not express a virally encoded protein on their surface, thus distinguishing them from conventional enveloped viruses. The absence of a viral fusion protein implies that these quasi-enveloped virions have unique mechanisms for entry into cells. Like true enveloped viruses, however, these phylogenetically distinct viruses usurp components of the host ESCRT system to hijack host cell membranes and noncytolytically exit infected cells. The membrane protects these viruses from neutralizing antibodies, facilitating dissemination within the host, whereas nonenveloped virions shed in feces are stable in the environment, allowing for epidemic transmission.
Collapse
Affiliation(s)
- Zongdi Feng
- Lineberger Comprehensive Cancer Center, Inflammatory Diseases Institute, and Departments of Medicine and Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7292;
| | - Asuka Hirai-Yuki
- Lineberger Comprehensive Cancer Center, Inflammatory Diseases Institute, and Departments of Medicine and Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7292;
| | - Kevin L McKnight
- Lineberger Comprehensive Cancer Center, Inflammatory Diseases Institute, and Departments of Medicine and Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7292;
| | - Stanley M Lemon
- Lineberger Comprehensive Cancer Center, Inflammatory Diseases Institute, and Departments of Medicine and Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7292;
| |
Collapse
|
216
|
Abstract
FcRn, resembling a major histocompatibility complex class I molecule with a closed peptide cleft, is an intracellular molecule that binds endocytosed albumin and IgG by a pH-dependent mechanism, diverting them from degradative fates and moving them out of the cell. The turnover of both of these important plasma proteins is thus regulated, as discussed by Schmidt and colleagues in this issue of Structure.
Collapse
Affiliation(s)
- Clark L Anderson
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | | |
Collapse
|
217
|
Azanza JR, Sádaba B, Gómez-Guiu A. Monoclonal antibodies: Pharmacokinetics as a basis for new dosage regimens? J Oncol Pharm Pract 2014; 21:370-6. [DOI: 10.1177/1078155214538085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Complete monoclonal IgG antibodies which are in use in clinical practice share some pharmacological properties resulting in high concentrations in plasma. This fact is reflected in their low volumes of distribution, which can also be correlated with a high molecular weight and water solubility. This feature allows a novel approach to be applied to the dosing schedule for this group of drugs with fixed doses being used instead of the initially developed weight- or body surface-adjusted dosing schedules. In addition, the development of a new formulation containing hyaluronidase allows a subcutaneous route of administration to be used, because hyaluronidase creates a space in the subcutaneous tissue that helps antibody absorption. This method requires higher doses, but has allowed testing the feasibility of administering a fixed dose, with no individual dose adjustments based on weight or body surface. Moreover, loading doses are not needed, because the first dose results, within 3 weeks, in minimum concentrations that are higher than effective concentrations.
Collapse
Affiliation(s)
- J-R Azanza
- Department of Clinical Pharmacology, Clínica Universidad de Navarra, Pamplona, Spain
| | - B Sádaba
- Department of Clinical Pharmacology, Clínica Universidad de Navarra, Pamplona, Spain
| | - A Gómez-Guiu
- Department of Clinical Pharmacology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
218
|
Pang G, Qiao D, Chen Q, Hu Z, Xie J. Heterogeneous IgG interacts with FcRn and its transport across gastrointestinal barrier. FOOD AGR IMMUNOL 2014. [DOI: 10.1080/09540105.2014.918588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
219
|
In Vitro functional characterization of feline IgGs. Vet Immunol Immunopathol 2014; 158:214-23. [DOI: 10.1016/j.vetimm.2014.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/19/2013] [Accepted: 01/28/2014] [Indexed: 11/20/2022]
|
220
|
Precise and in situ genetic humanization of 6 Mb of mouse immunoglobulin genes. Proc Natl Acad Sci U S A 2014; 111:5147-52. [PMID: 24706858 DOI: 10.1073/pnas.1323896111] [Citation(s) in RCA: 318] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Genetic humanization, which involves replacing mouse genes with their human counterparts, can create powerful animal models for the study of human genes and diseases. One important example of genetic humanization involves mice humanized for their Ig genes, allowing for human antibody responses within a mouse background (HumAb mice) and also providing a valuable platform for the generation of fully human antibodies as therapeutics. However, existing HumAb mice do not have fully functional immune systems, perhaps because of the manner in which they were genetically humanized. Heretofore, most genetic humanizations have involved disruption of the endogenous mouse gene with simultaneous introduction of a human transgene at a new and random location (so-called KO-plus-transgenic humanization). More recent efforts have attempted to replace mouse genes with their human counterparts at the same genetic location (in situ humanization), but such efforts involved laborious procedures and were limited in size and precision. We describe a general and efficient method for very large, in situ, and precise genetic humanization using large compound bacterial artificial chromosome-based targeting vectors introduced into mouse ES cells. We applied this method to genetically humanize 3-Mb segments of both the mouse heavy and κ light chain Ig loci, by far the largest genetic humanizations ever described. This paper provides a detailed description of our genetic humanization approach, and the companion paper reports that the humoral immune systems of mice bearing these genetically humanized loci function as efficiently as those of WT mice.
Collapse
|
221
|
Baumrucker CR, Bruckmaier RM. Colostrogenesis: IgG1 transcytosis mechanisms. J Mammary Gland Biol Neoplasia 2014; 19:103-17. [PMID: 24474529 DOI: 10.1007/s10911-013-9313-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/28/2013] [Indexed: 01/14/2023] Open
Abstract
Biological transport of intact proteins across epithelial cells has been documented for many absorptive and secretory tissues. Immunoglobulins were some of the earliest studied proteins in this category. The transcellular transport (transcytosis) of immunoglobulins in neonatal health and development has been recognized; the process is especially significant with ungulates because they do not transcytose immunoglobulins across the placenta to the neonate. Rather, they depend upon mammary secretion of colostrum and intestinal absorption of immunoglobulins in order to provide intestinal and systemic defense until the young ungulate develops its own humoral defense mechanisms. The neonatal dairy calf's ability to absorb immunoglobulins from colostrum is assisted by a ~24 h "open gut" phenomenon where large proteins pass the intestinal epithelial cells and enter the systemic system. However, a critical problem recognized for newborn dairy calves is that an optimum mass of colostrum Immunoglobulin G (IgG) needs to be absorbed within that 24 h window in order to provide maximal resistance to disease. Many calves do not achieve the optimum because of poor quality colostrum. While many studies have focused on calf absorption, the principal cause of the problem resides with the extreme variation (g to kg) in the mammary gland's capacity to transfer blood IgG1 into colostrum. Colostrum is a unique mammary secretory product that is formed during late pregnancy when mammary cells are proliferating and differentiating in preparation for lactation. In addition to the transcytosis of immunoglobulins, the mammary gland also concentrates a number of circulating hormones into colostrum. Remarkably, the mechanisms in the formation of colostrum in ungulates have been rather modestly studied. The mechanisms and causes of this variation in mammary gland transcytosis of IgG1 are examined, evaluated, and in some cases, explained.
Collapse
Affiliation(s)
- Craig R Baumrucker
- Department of Animal Science, The Pennsylvania State University, 302 Henning Bldg, University Park, PA, 16802, USA,
| | | |
Collapse
|
222
|
Ward ES, Velmurugan R, Ober RJ. Targeting FcRn for therapy: from live cell imaging to in vivo studies in mice. Immunol Lett 2014; 160:158-62. [PMID: 24572175 DOI: 10.1016/j.imlet.2014.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/18/2022]
Abstract
The role of FcRn in regulating antibody levels and transport in the body is well documented. The use of fluorescence microscopy to investigate the subcellular trafficking behavior of FcRn and its IgG ligand has led to insight into the function of this receptor, including the identification of new intracellular pathways. The inhibition of FcRn using engineered antibodies that bind to this receptor with increased affinity through their Fc region can be exploited to treat antibody mediated autoimmunity. The efficacy of this approach in mouse models of arthritis and multiple sclerosis has been demonstrated. Finally, the cross-species difference between mouse and man for FcRn-IgG interactions needs to be considered when engineering antibodies for improved activities in FcRn-mediated functions.
Collapse
Affiliation(s)
- E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| | - Ramraj Velmurugan
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas TX 75390, USA.
| | - Raimund J Ober
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
223
|
van Vught R, Pieters RJ, Breukink E. Site-specific functionalization of proteins and their applications to therapeutic antibodies. Comput Struct Biotechnol J 2014; 9:e201402001. [PMID: 24757499 PMCID: PMC3995230 DOI: 10.5936/csbj.201402001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/28/2014] [Accepted: 02/04/2014] [Indexed: 12/19/2022] Open
Abstract
Protein modifications are often required to study structure and function relationships. Instead of the random labeling of lysine residues, methods have been developed to (sequence) specific label proteins. Next to chemical modifications, tools to integrate new chemical groups for bioorthogonal reactions have been applied. Alternatively, proteins can also be selectively modified by enzymes. Herein we review the methods available for site-specific modification of proteins and their applications for therapeutic antibodies.
Collapse
Affiliation(s)
- Remko van Vught
- Department of Membrane Biochemistry and Biophysics, Institute of Biomembranes, Utrecht University, Padualaan 8, 3584CH Utrecht, The Netherlands
| | - Roland J Pieters
- Department of Medicinal Chemistry and Chemical Biology. Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Eefjan Breukink
- Department of Membrane Biochemistry and Biophysics, Institute of Biomembranes, Utrecht University, Padualaan 8, 3584CH Utrecht, The Netherlands
| |
Collapse
|
224
|
Fronton L, Pilari S, Huisinga W. Monoclonal antibody disposition: a simplified PBPK model and its implications for the derivation and interpretation of classical compartment models. J Pharmacokinet Pharmacodyn 2014; 41:87-107. [PMID: 24493102 DOI: 10.1007/s10928-014-9349-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 01/15/2014] [Indexed: 11/24/2022]
Abstract
The structure, interpretation and parameterization of classical compartment models as well as physiologically-based pharmacokinetic (PBPK) models for monoclonal antibody (mAb) disposition are very diverse, with no apparent consensus. In addition, there is a remarkable discrepancy between the simplicity of experimental plasma and tissue profiles and the complexity of published PBPK models. We present a simplified PBPK model based on an extravasation rate-limited tissue model with elimination potentially occurring from various tissues and plasma. Based on model reduction (lumping), we derive several classical compartment model structures that are consistent with the simplified PBPK model and experimental data. We show that a common interpretation of classical two-compartment models for mAb disposition-identifying the central compartment with the total plasma volume and the peripheral compartment with the interstitial space (or part of it)-is not consistent with current knowledge. Results are illustrated for the monoclonal antibodies 7E3 and T84.66 in mice.
Collapse
Affiliation(s)
- Ludivine Fronton
- Institute of Biochemistry and Biology, Universität Potsdam, Potsdam, Germany
| | | | | |
Collapse
|
225
|
Oganesyan V, Damschroder MM, Cook KE, Li Q, Gao C, Wu H, Dall'Acqua WF. Structural insights into neonatal Fc receptor-based recycling mechanisms. J Biol Chem 2014; 289:7812-24. [PMID: 24469444 DOI: 10.1074/jbc.m113.537563] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We report the three-dimensional structure of human neonatal Fc receptor (FcRn) bound concurrently to its two known ligands. More particularly, we solved the crystal structure of the complex between human FcRn, wild-type human serum albumin (HSA), and a human Fc engineered for improved pharmacokinetics properties (Fc-YTE). The crystal structure of human FcRn bound to wild-type HSA alone is also presented. HSA domain III exhibits an extensive interface of contact with FcRn, whereas domain I plays a lesser role. A molecular explanation for the HSA recycling mechanism is provided with the identification of FcRn His(161) as the only potential direct contributor to the corresponding pH-dependent process. At last, this study also allows an accurate structural definition of residues considered for decades as important to the human IgG/FcRn interaction and reveals Fc His(310) as a significant contributor to pH-dependent binding. Finally, we explain various structural mechanisms by which several Fc mutations (including YTE) result in increased human IgG binding to FcRn. Our study provides an unprecedented relevant understanding of the molecular basis of human Fc interaction with human FcRn.
Collapse
Affiliation(s)
- Vaheh Oganesyan
- From the Department of Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, Maryland 20878
| | | | | | | | | | | | | |
Collapse
|
226
|
Divergent outcomes following transcytosis of IgG targeting intracellular and extracellular chlamydial antigens. Immunol Cell Biol 2014; 92:417-26. [PMID: 24445600 DOI: 10.1038/icb.2013.110] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/11/2013] [Accepted: 12/12/2013] [Indexed: 12/12/2022]
Abstract
Antibodies can have a protective but non-essential role in natural chlamydial infections dependent on antigen specificity and antibody isotype. IgG is the dominant antibody in both male and female reproductive tract mucosal secretions, and is bi-directionally trafficked across epithelia by the neonatal Fc receptor (FcRn). Using pH-polarized epididymal epithelia grown on Transwells, IgG specifically targeted at an extracellular chlamydial antigen; the major outer membrane protein (MOMP), enhanced uptake and translocation of infection at pH 6-6.5 but not at neutral pH. This was dependent on FcRn expression. Conversely, FcRn-mediated transport of IgG targeting the intracellular chlamydial inclusion membrane protein A (IncA), induced aberrant inclusion morphology, recruited autophagic proteins independent of lysosomes and significantly reduced infection. Challenge of female mice with MOMP-specific IgG-opsonized Chlamydia muridarum delayed infection clearance but exacerbated oviduct occlusion. In male mice, MOMP-IgG elicited by immunization afforded no protection against testicular chlamydial infection, whereas the transcytosis of IncA-IgG significantly reduced testicular chlamydial burden. Together these data show that the protective and pathological effects of IgG are dependent on FcRn-mediated transport as well as the specificity of IgG for intracellular or extracellular antigens.
Collapse
|
227
|
Dickson LE, Wagner MC, Sandoval RM, Molitoris BA. The proximal tubule and albuminuria: really! J Am Soc Nephrol 2014; 25:443-53. [PMID: 24408874 DOI: 10.1681/asn.2013090950] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recent data highlight the role of the proximal tubule (PT) in reabsorbing, processing, and transcytosing urinary albumin from the glomerular filtrate. Innovative techniques and approaches have provided exciting insights into these processes, and numerous investigators have shown that selective PT cell defects lead to significant albuminuria, even reaching nephrotic range in animal models. Thus, the mechanisms of albumin reabsorption and transcytosis are undergoing intense study. Working in concert with megalin and cubilin, a nonselective multireceptor complex that predominantly directs proteins for lysosomal degradation, the neonatal Fc receptor (FcRn) located at the brush border of the apical membrane has been implicated as the "receptor" mediating albumin transcytosis. The FcRn pathway facilitates reabsorption and mediates transcytosis by its pH-dependent binding affinity in endosomal compartments. This also allows for selective albumin sorting within the PT cell. This reclamation pathway minimizes urinary losses and catabolism of albumin, thus prolonging its serum half-life. It may also serve as a molecular sorter to preserve and reclaim normal albumin while allowing "altered" albumin to be catabolized via lysosomal pathways. Here, we critically review the data supporting this novel mechanism.
Collapse
Affiliation(s)
- Landon E Dickson
- Indiana University School of Medicine, The Roudebush Veterans Affairs Medical Center, Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | | | | | | |
Collapse
|
228
|
Abstract
Subcutaneous administration of IgG (SCIG) has become widely used in primary immune deficiency diseases but it has only recently been studied for maintenance therapy in autoimmune peripheral neuropathies, such as chronic idiopathic demyelinating polyneuropathy and multifocal motor neuropathy. Weekly self-administration of SCIG is safe and well-tolerated, and results in steady-state serum IgG levels, as contrasted with the peaks and troughs of monthly immune globulin (human) for intravenous use. Freedom from the need for venous access or medical personnel for infusions, flexibility in scheduling, convenience of home therapy, and improved clinical stability due to the steady-state IgG levels, lead many patients to prefer SCIG to immune globulin (human) for intravenous use. Long-term studies are needed to determine if the constant IgG levels and clinical stability translate into better long-term outcomes.
Collapse
Affiliation(s)
- Melvin Berger
- Immunology Research & Development, CSL Behring, LLC, 1020 First Avenue, King of Prussia, PA 19406, USA and Pediatrics & Pathology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
229
|
Bergeron LM, McCandless EE, Dunham S, Dunkle B, Zhu Y, Shelly J, Lightle S, Gonzales A, Bainbridge G. Comparative functional characterization of canine IgG subclasses. Vet Immunol Immunopathol 2014; 157:31-41. [DOI: 10.1016/j.vetimm.2013.10.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/26/2013] [Accepted: 10/28/2013] [Indexed: 01/15/2023]
|
230
|
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
|
231
|
Abstract
Antibodies are major molecular effectors of adaptive immune responses. Most, if not all, biological activities of antibodies, however, depend on the functional properties of cells that express receptors for the Fc portion of antibodies (FcR). Most FcR are activating receptors; some are inhibitory. When engaged by antibodies and antigen, the various FcR expressed by a given cell trigger a mixture of positive and negative signals whose integration determines cellular responses. Responses of cell populations can be either protective or pathogenic. As a consequence, FcR are potential target/tools in a variety of diseases including infection, allergy, autoimmune diseases, and cancer.
Collapse
|
232
|
Boross P, Lohse S, Nederend M, Jansen JHM, van Tetering G, Dechant M, Peipp M, Royle L, Liew LP, Boon L, van Rooijen N, Bleeker WK, Parren PWHI, van de Winkel JGJ, Valerius T, Leusen JHW. IgA EGFR antibodies mediate tumour killing in vivo. EMBO Mol Med 2013; 5:1213-26. [PMID: 23918228 PMCID: PMC3944462 DOI: 10.1002/emmm.201201929] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 06/06/2013] [Accepted: 06/08/2013] [Indexed: 01/02/2023] Open
Abstract
Currently all approved anti-cancer therapeutic monoclonal antibodies (mAbs) are of the IgG isotype, which rely on Fcgamma receptors (FcγRs) to recruit cellular effector functions. In vitro studies showed that targeting of FcαRI (CD89) by bispecific antibodies (bsAbs) or recombinant IgA resulted in more effective elimination of tumour cells by myeloid effector cells than targeting of FcγR. Here we studied the in vivo anti-tumour activity of IgA EGFR antibodies generated using the variable sequences of the chimeric EGFR antibody cetuximab. Using FcαRI transgenic mice, we demonstrated significant in vivo anti-tumour activity of IgA2 EGFR against A431 cells in peritoneal and lung xenograft models, as well as against B16F10-EGFR cells in a lung metastasis model in immunocompetent mice. IgA2 EGFR was more effective than cetuximab in a short-term syngeneic peritoneal model using EGFR-transfected Ba/F3 target cells. The in vivo cytotoxic activity of IgA2 EGFR was mediated by macrophages and was significantly decreased in the absence of FcαRI. These results support the potential of targeting FcαRI for effective antibody therapy of cancer. The study reveals that IgA antibodies directed against EGFR and engaging Fcalpha receptor (FcαRI) on effector cells, have in vivo anti-cancer activity. These data support the development of novel immunotherapeutic strategies based on targeting FcαRI.
Collapse
Affiliation(s)
- Peter Boross
- Immunotherapy Laboratory, Laboratory for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Sedykh MA, Buneva VN, Nevinsky GA. Polyreactivity of natural antibodies: Exchange by HL-fragments. BIOCHEMISTRY (MOSCOW) 2013; 78:1305-1320. [DOI: 10.1134/s0006297913120018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
234
|
Engler FA, Zheng B, Balthasar JP. Investigation of the influence of nephropathy on monoclonal antibody disposition: a pharmacokinetic study in a mouse model of diabetic nephropathy. Pharm Res 2013; 31:1185-93. [PMID: 24203494 DOI: 10.1007/s11095-013-1241-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/20/2013] [Indexed: 01/06/2023]
Abstract
PURPOSE This study employed a mouse model to evaluate the effects of diabetic nephropathy on the pharmacokinetics of 8C2, a murine monoclonal antibody (mAb). METHODS Streptozotocin (STZ) was administered to mice to induce diabetic nephropathy (125 mg/kg/day × 2). Mice were grouped (n = 8-10) based on time after STZ-treatment (control, 1, 2, 3, 4, or 6 weeks), and injected intravenously with 10 mg/kg 8C2. Blood samples were collected up to 7 days, and 8C2 plasma concentrations were determined via immunoassay. Inulin clearance and urinary albumin excretion rate (UAE) were determined to assess renal function. RESULTS UAE, inulin clearance, and 8C2 clearance increased significantly following STZ. Comparing control and 6 week STZ-treatment groups, UAE and inulin clearance increased from 25.7 ± 3.3 to 99.3 ± 13.7 μg/day, and from 421 ± 31 to 584 ± 78 μl/min. 8C2 clearance increased from 121 ± 12.5 to 228 ± 61 μl/hr/kg (p < 0.01). 8C2 clearance was highly correlated with UAE (r(2): 0.731). Inclusion of UAE as a covariate in population modeling explained significant residual variability in 8C2 clearance. CONCLUSIONS The clearance of 8C2 increased significantly in STZ-treated mice. Population pharmacokinetic modeling suggests that UAE has potential for use in predicting mAb clearance in subjects with diabetic nephropathy, possibly assisting in the individualization of mAb dosing.
Collapse
Affiliation(s)
- Frank A Engler
- Department of Pharmaceutical Sciences School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 452 Kapoor Hall, Buffalo, New York, 14260, USA
| | | | | |
Collapse
|
235
|
Hristodorov D, Fischer R, Joerissen H, Müller-Tiemann B, Apeler H, Linden L. Generation and comparative characterization of glycosylated and aglycosylated human IgG1 antibodies. Mol Biotechnol 2013; 53:326-35. [PMID: 22427250 DOI: 10.1007/s12033-012-9531-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Monoclonal antibodies (mAbs) are the fastest growing class of biopharmaceuticals reflecting their diverse applications in research and the clinic. The correct glycosylation of mAbs is required to elicit effector functions such as complement-dependent and antibody-dependent cell-mediated cytotoxicity, although these may be undesirable for the treatment of certain chronic diseases. To gain insight into the properties of glycan-deficient mAbs, we generated and characterized six different aglycosylated human IgG1 mAbs (carrying the N297A mutation) and compared them to their glycosylated counterparts. We found no differences in solubility or heterogeneity, and all mAbs the remained stable in stress tests at 4 and 37 °C. Surface plasmon resonance spectroscopy showed no differences in binding affinity, and the in vivo terminal serum half-life and plasma clearance were similar in rats. However, differential scanning calorimetry revealed that the aglycosylated mAbs contained a less stable C(H)2 domain and they were also significantly more susceptible to pH-induced aggregation. We conclude that aglycosylated mAbs are functionally equivalent to their glycosylated counterparts and could be particularly suitable for certain therapeutic applications, such as the treatment of chronic diseases.
Collapse
Affiliation(s)
- Dmitrij Hristodorov
- GDD-GB-BRG-Cell & Protein Science, Purification & Research Analytics, Bayer Healthcare AG, Wuppertal, Germany
| | | | | | | | | | | |
Collapse
|
236
|
Ng CM, Loyet KM, Iyer S, Fielder PJ, Deng R. Modeling approach to investigate the effect of neonatal Fc receptor binding affinity and anti-therapeutic antibody on the pharmacokinetic of humanized monoclonal anti-tumor necrosis factor-α IgG antibody in cynomolgus monkey. Eur J Pharm Sci 2013; 51:51-8. [PMID: 23999033 DOI: 10.1016/j.ejps.2013.08.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 07/08/2013] [Accepted: 08/21/2013] [Indexed: 01/17/2023]
Abstract
PURPOSE Several neonatal Fc receptor (FcRn) variants of an anti-tumor necrosis factor (TNF)-α humanized monoclonal IgG antibodies (mAbs) were developed but the effect of their differential FcRn binding affinities on pharmacokinetic (PK) behavior were difficult to be definitively measured in vivo due to formation of anti-therapeutic antibody (ATA). A semi-mechanistic model was developed to investigate the quantitative relationship between the FcRn binding affinity and PK of mAbs in cynomolgus monkey with the presence of ATA. METHODS PK and ATA data from cynomolgus monkeys which received a single intravenous dose of adalimumab, wild-type or two FcRn variant (N434H and N434A) anti-TNF-α mAbs were included in the analysis. Likelihood-based censored data handling method was used to include many PK observations with BQL values for model development. A fully integrated PK-ATA model was developed and used to fit simultaneously to the PK/ATA data. RESULTS AND CONCLUSIONS The PK and ATA time-profiles and effect of FcRn-binding affinity on PK of mAbs were well described by the model and the parameters were estimated with good precision. The model was used successfully to construct quantitative relationships between FcRn binding affinity and PK of anti-TNF-α mAbs in the presence of the ATA-mediated elimination and interferences.
Collapse
Affiliation(s)
- Chee M Ng
- Clinical Pharmacology and Therapeutic, Children Hospital of Philadelphia, PA, United States; School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | | | | | | | | |
Collapse
|
237
|
Neutralizing IgG at the portal of infection mediates protection against vaginal simian/human immunodeficiency virus challenge. J Virol 2013; 87:11604-16. [PMID: 23966410 DOI: 10.1128/jvi.01361-13] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neutralizing antibodies may have critical importance in immunity against human immunodeficiency virus type 1 (HIV-1) infection. However, the amount of protective antibody needed at mucosal surfaces has not been fully established. Here, we evaluated systemic and mucosal pharmacokinetics (PK) and pharmacodynamics (PD) of 2F5 IgG and 2F5 Fab fragments with respect to protection against vaginal challenge with simian-human immunodeficiency virus-BaL in macaques. Antibody assessment demonstrated that 2F5 IgG was more potent than polymeric forms (IgM and IgA) across a range of cellular and tissue models. Vaginal challenge studies demonstrated a dose-dependent protection for 2F5 IgG and no protection with 2F5 Fab despite higher vaginal Fab levels at the time of challenge. Animals receiving 50 or 25 mg/kg of body weight 2F5 IgG were completely protected, while 3/5 animals receiving 5 mg/kg were protected. In the control animals, infection was established by a minimum of 1 to 4 transmitted/founder (T/F) variants, similar to natural human infection by this mucosal route; in the two infected animals that had received 5 mg 2F5 IgG, infection was established by a single T/F variant. Serum levels of 2F5 IgG were more predictive of sterilizing protection than measured vaginal levels. Fc-mediated antiviral activity did not appear to influence infection of primary target cells in cervical explants. However, PK studies highlighted the importance of the Fc portion in tissue biodistribution. Data presented in this study may be important in modeling serum levels of neutralizing antibodies that need to be achieved by either vaccination or passive infusion to prevent mucosal acquisition of HIV-1 infection in humans.
Collapse
|
238
|
Gurbaxani B, Dostalek M, Gardner I. Are endosomal trafficking parameters better targets for improving mAb pharmacokinetics than FcRn binding affinity? Mol Immunol 2013; 56:660-74. [PMID: 23917469 DOI: 10.1016/j.molimm.2013.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 05/09/2013] [Accepted: 05/14/2013] [Indexed: 12/21/2022]
Abstract
F.W.R. Brambell deduced the existence of a protective receptor for IgG, the neonatal Fc receptor (FcRn), long before its discovery in the early to mid-1990s. With the coincident, explosive development of IgG-based drugs, FcRn became a popular target for tuning the pharmacokinetics of monoclonal antibodies (mAbs). One aspect of Brambell's initial observation, however, that is seldom discussed since the discovery of the receptor, is the compliance in the mechanism that Brambell observed (saturating at 10s-100s of μM concentration), vs. the comparative stiffness of the receptor kinetics (saturating in the nM range for most species). Although some studies reported that increasing the already very high Fc-FcRn affinity at pH 6.0 further improved mAb half-life, in fact the results were mixed, with later studies increasingly implicating non-FcRn-dependent mechanisms as determinants of mAb pharmacokinetics. Mathematical modelling of the FcRn system has also indicated that the processes determining the pharmacokinetics of mAbs have more nuances than had at first been hypothesised. We propose, in keeping with the latest modelling and experimental evidence reviewed here, that the dynamics of endosomal sorting and trafficking have important roles in the compliant salvage mechanism that Brambell first observed nearly 50 years ago, and therefore also in the pharmacokinetics of mAbs. These ideas lead to many open questions regarding the endosomal trafficking of both FcRn and mAbs and also to what properties of a mAb can be altered to achieve an improvement in pharmacokinetics.
Collapse
Affiliation(s)
- Brian Gurbaxani
- Chronic Viral Diseases Branch, Division of High-Consequence Pathogens and Pathology, National Centre for Emerging and Zoonotic Infectious Diseases, Centres for Disease Control and Prevention, Atlanta, GA, USA.
| | | | | |
Collapse
|
239
|
A novel hypothesis for an alkaline phosphatase 'rescue' mechanism in the hepatic acute phase immune response. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2044-56. [PMID: 23899605 DOI: 10.1016/j.bbadis.2013.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/10/2013] [Accepted: 07/22/2013] [Indexed: 12/24/2022]
Abstract
The liver isoform of the enzyme alkaline phosphatase (AP) has been used classically as a serum biomarker for hepatic disease states such as hepatitis, steatosis, cirrhosis, drug-induced liver injury, and hepatocellular carcinoma. Recent studies have demonstrated a more general anti-inflammatory role for AP, as it is capable of dephosphorylating potentially deleterious molecules such as nucleotide phosphates, the pathogenic endotoxin lipopolysaccharide (LPS), and the contact clotting pathway activator polyphosphate (polyP), thereby reducing inflammation and coagulopathy systemically. Yet the mechanism underlying the observed increase in liver AP levels in circulation during inflammatory insults is largely unknown. This paper hypothesizes an immunological role for AP in the liver and the potential of this system for damping generalized inflammation along with a wide range of ancillary pathologies. Based on the provided framework, a mechanism is proposed in which AP undergoes transcytosis in hepatocytes from the canalicular membrane to the sinusoidal membrane during inflammation and the enzyme's expression is upregulated as a result. Through a tightly controlled, nucleotide-stimulated negative feedback process, AP is transported in this model as an immune complex with immunoglobulin G by the asialoglycoprotein receptor through the cell and secreted into the serum, likely using the receptor's State 1 pathway. The subsequent dephosphorylation of inflammatory stimuli by AP and uptake of the circulating immune complex by endothelial cells and macrophages may lead to decreased inflammation and coagulopathy while providing an early upstream signal for the induction of a number of anti-inflammatory gene products, including AP itself.
Collapse
|
240
|
Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A 2013; 110:E2987-96. [PMID: 23882082 DOI: 10.1073/pnas.1302725110] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Binding of hepatocyte growth factor (HGF) to the receptor tyrosine kinase MET is implicated in the malignant process of multiple cancers, making disruption of this interaction a promising therapeutic strategy. However, targeting MET with bivalent antibodies can mimic HGF agonism via receptor dimerization. To address this limitation, we have developed onartuzumab, an Escherichia coli-derived, humanized, and affinity-matured monovalent monoclonal antibody against MET, generated using the knob-into-hole technology that enables the antibody to engage the receptor in a one-to-one fashion. Onartuzumab potently inhibits HGF binding and receptor phosphorylation and signaling and has antibody-like pharmacokinetics and antitumor activity. Biochemical data and a crystal structure of a ternary complex of onartuzumab antigen-binding fragment bound to a MET extracellular domain fragment, consisting of the MET Sema domain fused to the adjacent Plexins, Semaphorins, Integrins domain (MET Sema-PSI), and the HGF β-chain demonstrate that onartuzumab acts specifically by blocking HGF α-chain (but not β-chain) binding to MET. These data suggest a likely binding site of the HGF α-chain on MET, which when dimerized leads to MET signaling. Onartuzumab, therefore, represents the founding member of a class of therapeutic monovalent antibodies that overcomes limitations of antibody bivalency for targets impacted by antibody crosslinking.
Collapse
|
241
|
An inactivated cell culture Japanese encephalitis vaccine (JE-ADVAX) formulated with delta inulin adjuvant provides robust heterologous protection against West Nile encephalitis via cross-protective memory B cells and neutralizing antibody. J Virol 2013; 87:10324-33. [PMID: 23864620 DOI: 10.1128/jvi.00480-13] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
West Nile virus (WNV), currently the cause of a serious U.S. epidemic, is a mosquito-borne flavivirus and member of the Japanese encephalitis (JE) serocomplex. There is currently no approved human WNV vaccine, and treatment options remain limited, resulting in significant mortality and morbidity from human infection. Given the availability of approved human JE vaccines, this study asked whether the JE-ADVAX vaccine, which contains an inactivated cell culture JE virus antigen formulated with Advax delta inulin adjuvant, could provide heterologous protection against WNV infection in wild-type and β2-microglobulin-deficient (β2m(-/-)) murine models. Mice immunized twice or even once with JE-ADVAX were protected against lethal WNV challenge even when mice had low or absent serum cross-neutralizing WNV titers prior to challenge. Similarly, β2m(-/-) mice immunized with JE-ADVAX were protected against lethal WNV challenge in the absence of CD8(+) T cells and prechallenge WNV antibody titers. Protection against WNV could be adoptively transferred to naive mice by memory B cells from JE-ADVAX-immunized animals. Hence, in addition to increasing serum cross-neutralizing antibody titers, JE-ADVAX induced a memory B-cell population able to provide heterologous protection against WNV challenge. Heterologous protection was reduced when JE vaccine antigen was administered alone without Advax, confirming the importance of the adjuvant to induction of cross-protective immunity. In the absence of an approved human WNV vaccine, JE-ADVAX could provide an alternative approach for control of a major human WNV epidemic.
Collapse
|
242
|
Proetzel G, Roopenian DC. Humanized FcRn mouse models for evaluating pharmacokinetics of human IgG antibodies. Methods 2013; 65:148-53. [PMID: 23867339 DOI: 10.1016/j.ymeth.2013.07.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/29/2013] [Accepted: 07/02/2013] [Indexed: 12/22/2022] Open
Abstract
A key element for the successful development of novel therapeutic antibodies is to fully understand their pharmacokinetic and pharmacodynamic behavior before performing clinical trials. While many in vitro modeling approaches exist, these simply cannot substitute for data obtained from appropriate animal models. It was established quite early that the unusual long serum half-life of immunoglobulin G's (IgGs) and Fc domains are due to their rescue and recycling by the neonatal Fc receptor (FcRn). The diverse roles of FcRn became apparent after isolation and cloning. Interesting are the significant species differences between rodent and human FcRn reactivity, rendering wild type rodents an inadequate model for studying IgG serum half-life. With the advance of genetic engineering mouse models have been established expressing human FcRn, and lacking mouse FcRn protein. These models have become highly relevant tools for serum half-life analysis of Fc-containing compounds.
Collapse
|
243
|
Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52:83-124. [PMID: 23299465 DOI: 10.1007/s40262-012-0027-4] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of monoclonal antibodies (mAbs) and their functional derivatives represents a growing segment of the development pipeline in the pharmaceutical industry. More than 25 mAbs and derivatives have been approved for a variety of therapeutic applications. In addition, around 500 mAbs and derivatives are currently in different stages of development. mAbs are considered to be large molecule therapeutics (in general, they are 2-3 orders of magnitude larger than small chemical molecule therapeutics), but they are not just big chemicals. These compounds demonstrate much more complex pharmacokinetic and pharmacodynamic behaviour than small molecules. Because of their large size and relatively poor membrane permeability and instability in the conditions of the gastrointestinal tract, parenteral administration is the most usual route of administration. The rate and extent of mAb distribution is very slow and depends on extravasation in tissue, distribution within the particular tissue, and degradation. Elimination primarily happens via catabolism to peptides and amino acids. Although not definitive, work has been published to define the human tissues mainly involved in the elimination of mAbs, and it seems that many cells throughout the body are involved. mAbs can be targeted against many soluble or membrane-bound targets, thus these compounds may act by a variety of mechanisms to achieve their pharmacological effect. mAbs targeting soluble antigen generally exhibit linear elimination, whereas those targeting membrane-bound antigen often exhibit non-linear elimination, mainly due to target-mediated drug disposition (TMDD). The high-affinity interaction of mAbs and their derivatives with the pharmacological target can often result in non-linear pharmacokinetics. Because of species differences (particularly due to differences in target affinity and abundance) in the pharmacokinetics and pharmacodynamics of mAbs, pharmacokinetic/pharmacodynamic modelling of mAbs has been used routinely to expedite the development of mAbs and their derivatives and has been utilized to help in the selection of appropriate dose regimens. Although modelling approaches have helped to explain variability in both pharmacokinetic and pharmacodynamic properties of these drugs, there is a clear need for more complex models to improve understanding of pharmacokinetic processes and pharmacodynamic interactions of mAbs with the immune system. There are different approaches applied to physiologically based pharmacokinetic (PBPK) modelling of mAbs and important differences between the models developed. Some key additional features that need to be accounted for in PBPK models of mAbs are neonatal Fc receptor (FcRn; an important salvage mechanism for antibodies) binding, TMDD and lymph flow. Several models have been described incorporating some or all of these features and the use of PBPK models are expected to expand over the next few years.
Collapse
|
244
|
Kitamura T, Otsuki M, Tamada D, Tabuchi Y, Mukai K, Morita S, Kasayama S, Shimomura I, Koga M. Glycated albumin is set lower in relation to plasma glucose levels in patients with Cushing's syndrome. Clin Chim Acta 2013; 424:164-7. [PMID: 23792199 DOI: 10.1016/j.cca.2013.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 05/26/2013] [Accepted: 06/07/2013] [Indexed: 11/16/2022]
Abstract
BACKGROUND Glycated albumin (GA) is an indicator of glycemic control, which has some specific characters in comparison with HbA1c. Since glucocorticoids (GC) promote protein catabolism including serum albumin, GC excess state would influence GA levels. We therefore investigated GA levels in patients with Cushing's syndrome. METHODS We studied 16 patients with Cushing's syndrome (8 patients had diabetes mellitus and the remaining 8 patients were non-diabetic). Thirty-two patients with type 2 diabetes mellitus and 32 non-diabetic subjects matched for age, sex and BMI were used as controls. RESULTS In the patients with Cushing's syndrome, GA was significantly correlated with HbA1c, but the regression line shifted downwards as compared with the controls. The GA/HbA1c ratio in the patients with Cushing's syndrome was also significantly lower than the controls. HbA1c in the non-diabetic patients with Cushing's syndrome was not different from the non-diabetic controls, whereas GA was significantly lower. In 7 patients with Cushing's syndrome who performed self-monitoring of blood glucose, the measured HbA1c was matched with HbA1c estimated from mean blood glucose, whereas the measured GA was significantly lower than the estimated GA. CONCLUSIONS We clarified that GA is set lower in relation to plasma glucose levels in patients with Cushing's syndrome.
Collapse
Affiliation(s)
- Tetsuhiro Kitamura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
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
|
246
|
Sleep D, Cameron J, Evans LR. Albumin as a versatile platform for drug half-life extension. Biochim Biophys Acta Gen Subj 2013; 1830:5526-34. [PMID: 23639804 DOI: 10.1016/j.bbagen.2013.04.023] [Citation(s) in RCA: 353] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/16/2013] [Accepted: 04/17/2013] [Indexed: 01/10/2023]
Abstract
BACKGROUND Albumin is the most abundant plasma protein, is highly soluble, very stable and has an extraordinarily long circulatory half-life as a direct result of its size and interaction with the FcRn mediated recycling pathway. In contrast, many therapeutic molecules are smaller than the renal filtration threshold and are rapidly lost from the circulation thereby limiting their therapeutic potential. Albumin can be used in a variety of ways to increase the circulatory half-life of such molecules. SCOPE OF REVIEW This article will review the mechanisms which underpin albumin's extraordinarily long circulatory half-life and how the understanding of these processes are currently being employed to extend the circulatory half-life of drugs which can be engineered to bind to albumin, or are conjugated to, or genetically fused to, albumin. MAJOR CONCLUSIONS The recent and growing understanding of the pivotal role of FcRn in maintaining the extended circulatory half-life of albumin will necessitate a greater and more thorough investigation of suitable pre-clinical model systems for assessing the pharmacokinetic profiles of drugs associated, conjugated or fused to albumin. GENERAL SIGNIFICANCE Association, conjugation or fusion of therapeutic drugs to albumin is a well-accepted and established half-life extension technology. The manipulation of the albumin-FcRn interaction will facilitate the modulation of the circulatory half-life of albumin-enabled drugs, leading to superior pharmacokinetics tailored to the disease state and increased patient compliance. This article is part of a Special Issue entitled Serum Albumin.
Collapse
|
247
|
Wilkinson IC, Fowler SB, Machiesky L, Miller K, Hayes DB, Adib M, Her C, Borrok MJ, Tsui P, Burrell M, Corkill DJ, Witt S, Lowe DC, Webster CI. Monovalent IgG4 molecules: immunoglobulin Fc mutations that result in a monomeric structure. MAbs 2013; 5:406-17. [PMID: 23567207 DOI: 10.4161/mabs.23941] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Antibodies have become the fastest growing class of biological therapeutics, in part due to their exquisite specificity and ability to modulate protein-protein interactions with a high biological potency. The relatively large size and bivalency of antibodies, however, limits their use as therapeutics in certain circumstances. Antibody fragments, such as single-chain variable fragments and antigen binding-fragments, have emerged as viable alternatives, but without further modifications these monovalent formats have reduced terminal serum half-lives because of their small size and lack of an Fc domain, which is required for FcRn-mediated recycling. Using rational engineering of the IgG4 Fc domain to disrupt key interactions at the CH3-CH3 interface, we identified a number of point mutations that abolish Fc dimerization and created half-antibodies, a novel monovalent antibody format that retains a monomeric Fc domain. Introduction of these mutations into an IgG1 framework also led to the creation of half-antibodies. These half-antibodies were shown to be soluble, thermodynamically stable and monomeric, characteristics that are favorable for use as therapeutic proteins. Despite significantly reduced FcRn binding in vitro, which suggests that avidity gains in a dimeric Fc are critical to optimal FcRn binding, this format demonstrated an increased terminal serum half-life compared with that expected for most alternative antibody fragments.
Collapse
Affiliation(s)
- Ian C Wilkinson
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| | - Susan B Fowler
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| | - Leeann Machiesky
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - Kenneth Miller
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - David B Hayes
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - Morshed Adib
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - Cheng Her
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - M Jack Borrok
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - Ping Tsui
- MedImmune LLC.; Departments of Antibody Discovery and Protein Engineering and Analytical Biochemistry; Gaithersburg, MD USA
| | - Matthew Burrell
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| | - Dominic J Corkill
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| | - Susanne Witt
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| | - David C Lowe
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| | - Carl I Webster
- MedImmune Ltd.; Department of Antibody Discovery and Protein Engineering; Cambridge, UK
| |
Collapse
|
248
|
Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol 2013; 13:176-89. [PMID: 23411799 DOI: 10.1038/nri3401] [Citation(s) in RCA: 601] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Intravenous immunoglobulin (IVIG) preparations comprise pooled IgG antibodies from the serum of thousands of donors and were initially used as an IgG replacement therapy in immunocompromised patients. Since the discovery, more than 30 years ago, that IVIG therapy can ameliorate immune thrombocytopenia, the use of IVIG preparations has been extended to a wide range of autoimmune and inflammatory diseases. Despite the broad efficacy of IVIG therapy, its modes of action remain unclear. In this Review, we cover the recent insights into the molecular and cellular pathways that are involved in IVIG-mediated immunosuppression, with a particular focus on IVIG as a therapy for IgG-dependent autoimmune diseases.
Collapse
Affiliation(s)
- Inessa Schwab
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstrasse 3, 91058 Erlangen, Germany
| | | |
Collapse
|
249
|
JE-ADVAX vaccine protection against Japanese encephalitis virus mediated by memory B cells in the absence of CD8(+) T cells and pre-exposure neutralizing antibody. J Virol 2013; 87:4395-402. [PMID: 23388724 DOI: 10.1128/jvi.03144-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
JE-ADVAX is a new, delta inulin-adjuvanted, Japanese encephalitis (JE) candidate vaccine with a strong safety profile and potent immunogenicity that confers efficient immune protection not only against JE virus but also against related neurotropic flaviviruses such as West Nile virus. In this study, we investigated the immunological mechanism of protection by JE-ADVAX vaccine using knockout mice deficient in B cells or CD8(+) T cells and poor persistence of neutralizing antibody or by adoptive transfer of immune splenocyte subpopulations. We show that memory B cells induced by JE-ADVAX provide long-lived protection against JE even in the absence of detectable pre-exposure serum neutralizing antibodies and without the requirement of CD8(+) T cells. Upon virus encounter, these vaccine-induced memory B cells were rapidly triggered to produce neutralizing antibodies that then protected immunized mice from morbidity and mortality. The findings suggest that the extent of the B-cell memory compartment might be a better immunological correlate for clinical efficacy of JE vaccines than the currently recommended measure of serum neutralizing antibody. This may explain the paradox where JE protection is observed in some subjects even in the absence of detectable serum neutralizing antibody. Our investigation also established the suitability of a novel flavivirus challenge model (β(2)-microglobulin-knockout mice) for studies of the role of B-cell memory responses in vaccine protection.
Collapse
|
250
|
Xu Z, Davis HM, Zhou H. Rational development and utilization of antibody-based therapeutic proteins in pediatrics. Pharmacol Ther 2013; 137:225-47. [DOI: 10.1016/j.pharmthera.2012.10.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 10/08/2012] [Indexed: 12/15/2022]
|