101
|
Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Arrojo R, Liu D, Rossi EA, Chang CH, Goldenberg DM. Sacituzumab Govitecan (IMMU-132), an Anti-Trop-2/SN-38 Antibody-Drug Conjugate: Characterization and Efficacy in Pancreatic, Gastric, and Other Cancers. Bioconjug Chem 2015; 26:919-31. [PMID: 25915780 DOI: 10.1021/acs.bioconjchem.5b00223] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) made from a humanized anti-Trop-2 monoclonal antibody (hRS7) conjugated with the active metabolite of irinotecan, SN-38. In addition to its further characterization, as the clinical utility of IMMU-132 expands to an ever-widening range of Trop-2-expressing solid tumor types, its efficacy in new disease models needs to be explored in a nonclinical setting. Unlike most ADCs that use ultratoxic drugs and stable linkers, IMMU-132 uses a moderately toxic drug with a moderately stable carbonate bond between SN-38 and the linker. Flow cytometry and immunohistochemistry disclosed that Trop-2 is expressed in a wide range of tumor types, including gastric, pancreatic, triple-negative breast (TNBC), colonic, prostate, and lung. While cell-binding experiments reveal no significant differences between IMMU-132 and parental hRS7 antibody, surface plasmon resonance analysis using a Trop-2 CM5 chip shows a significant binding advantage for IMMU-132 over hRS7. The conjugate retained binding to the neonatal receptor, but it lost greater than 60% of the antibody-dependent cell-mediated cytotoxicity activity compared to that of hRS7. Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Pharmacokinetics of the intact ADC in mice reveals a mean residence time (MRT) of 15.4 h, while the carrier hRS7 antibody cleared at a similar rate as that of the unconjugated antibody (MRT ∼ 300 h). IMMU-132 treatment of mice bearing human gastric cancer xenografts (17.5 mg/kg; twice weekly × 4 weeks) resulted in significant antitumor effects compared to that of mice treated with a nonspecific control. Clinically relevant dosing schemes of IMMU-132 administered either every other week, weekly, or twice weekly in mice bearing human pancreatic or gastric cancer xenografts demonstrate similar, significant antitumor effects in both models. Current Phase I/II clinical trials ( ClinicalTrials.gov , NCT01631552) confirm anticancer activity of IMMU-132 in cancers expressing Trop-2, including gastric and pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | - Robert M Sharkey
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Preeti Trisal
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Roberto Arrojo
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Donglin Liu
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Edmund A Rossi
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States
| | - Chien-Hsing Chang
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States
| | - David M Goldenberg
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States.,§Center of Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, New Jersey 07950, United States
| |
Collapse
|
102
|
Charge-mediated influence of the antibody variable domain on FcRn-dependent pharmacokinetics. Proc Natl Acad Sci U S A 2015; 112:5997-6002. [PMID: 25918417 PMCID: PMC4434771 DOI: 10.1073/pnas.1408766112] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Here, we investigated the influence of the variable fragment (Fv) of IgG antibodies on the binding to the neonatal Fc receptor (FcRn) as well as on FcRn-dependent pharmacokinetics (PK). FcRn plays a key role in IgG homeostasis, and specific manipulation in the crystallizable fragment (Fc) is known to affect FcRn-dependent PK. Although the influence of the antigen-binding fragment (Fab) on FcRn interactions has been reported, the underlying mechanism is hitherto only poorly understood. Therefore, we analyzed the two IgG1 antibodies, briakinumab and ustekinumab, that have similar Fc parts but different terminal half-lives in human and systematically engineered variants of them with cross-over exchanges and varied charge distribution. Using FcRn affinity chromatography, molecular dynamics simulation, and in vivo PK studies in human FcRn transgenic mice, we provide evidence that the charge distribution on the Fv domain is involved in excessive FcRn binding. This excessive binding prevents efficient FcRn-IgG dissociation at physiological pH, thereby reducing FcRn-dependent terminal half-lives. Furthermore, we observed a linear correlation between FcRn column retention times of the antibody variants and the terminal half-lives in vivo. Taken together, our study contributes to a better understanding of the FcRn-IgG interaction, and it could also provide profound potential in FcRn-dependent antibody engineering of the variable Fab region.
Collapse
|
103
|
Henne KR, Ason B, Howard M, Wang W, Sun J, Higbee J, Tang J, Matsuda KC, Xu R, Zhou L, Chan JCY, King C, Piper DE, Ketchem RR, Michaels ML, Jackson SM, Retter MW. Anti-PCSK9 antibody pharmacokinetics and low-density lipoprotein-cholesterol pharmacodynamics in nonhuman primates are antigen affinity-dependent and exhibit limited sensitivity to neonatal Fc receptor-binding enhancement. J Pharmacol Exp Ther 2015; 353:119-31. [PMID: 25653417 DOI: 10.1124/jpet.114.221242] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as an attractive therapeutic target for cardiovascular disease. Monoclonal antibodies (mAbs) that bind PCSK9 and prevent PCSK9:low-density lipoprotein receptor complex formation reduce serum low-density lipoprotein-cholesterol (LDL-C) in vivo. PCSK9-mediated lysosomal degradation of bound mAb, however, dramatically reduces mAb exposure and limits duration of effect. Administration of high-affinity mAb1:PCSK9 complex (1:2) to mice resulted in significantly lower mAb1 exposure compared with mAb1 dosed alone in normal mice or in PCSK9 knockout mice lacking antigen. To identify mAb-binding characteristics that minimize lysosomal disposition, the pharmacokinetic behavior of four mAbs representing a diverse range of PCSK9-binding affinities at neutral (serum) and acidic (endosomal) pH was evaluated in cynomolgus monkeys. Results revealed an inverse correlation between affinity and both mAb exposure and duration of LDL-C lowering. High-affinity mAb1 exhibited the lowest exposure and shortest duration of action (6 days), whereas mAb2 displayed prolonged exposure and LDL-C reduction (51 days) as a consequence of lower affinity and pH-sensitive PCSK9 binding. mAbs with shorter endosomal PCSK9:mAb complex dissociation half-lives (<20 seconds) produced optimal exposure-response profiles. Interestingly, incorporation of previously reported Fc-region amino acid substitutions or novel loop-insertion peptides that enhance in vitro neonatal Fc receptor binding, led to only modest pharmacokinetic improvements for mAbs with pH-dependent PCSK9 binding, with only limited augmentation of pharmacodynamic activity relative to native mAbs. A pivotal role for PCSK9 in mAb clearance was demonstrated, more broadly suggesting that therapeutic mAb-binding characteristics require optimization based on target pharmacology.
Collapse
Affiliation(s)
- Kirk R Henne
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Brandon Ason
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Monique Howard
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Wei Wang
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Jeonghoon Sun
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Jared Higbee
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Jie Tang
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Katherine C Matsuda
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Ren Xu
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Lei Zhou
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Joyce C Y Chan
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Chadwick King
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Derek E Piper
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Randal R Ketchem
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Mark Leo Michaels
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Simon M Jackson
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| | - Marc W Retter
- Departments of Pharmacokinetics and Drug Metabolism (K.R.H., K.C.M., M.W.R.), Metabolic Disorders (B.A., J.C.Y.C., S.M.J.), Therapeutic Discovery (M.H., W.W., J.S., J.H., J.T., C.K., D.E.P., R.R.K., M.L.M.), Molecular Sciences (R.X.), and Biostatistics (L.Z.), Amgen, South San Francisco, California
| |
Collapse
|
104
|
Wu Q, Lee HY, Wong PY, Jiang G, Gazzano-Santoro H. Development and applications of AlphaScreen-based FcRn binding assay to characterize monoclonal antibodies. J Immunol Methods 2015; 420:31-7. [PMID: 25837414 DOI: 10.1016/j.jim.2015.03.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/18/2015] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
IgG antibodies are important pharmaceutical molecules that successfully treat a variety of human diseases. The neonatal Fc receptor (FcRn) interacts with IgG Fc in the CH2-CH3 domain and plays a key role in IgG antibody homeostasis and affects its pharmacokinetic properties. An in vitro FcRn binding assay could be a highly valuable complementary tool to assess IgG antibody pharmacokinetics in IgG engineering and screening during the early optimization stage. In addition, it could be useful in biological characterization studies for antibody minor variants, process optimization, and comparability study at later stages of antibody development. Here we developed a homogeneous AlphaScreen-based FcRn assay to assess the binding of FcRn to IgG antibody in vitro. The assay is found to be accurate, precise, specific, and simple: donor beads loaded with FcRn and acceptor beads loaded with IgG1 mAb1 are mixed together with sample IgG at various dilutions and incubated for 1h before acquiring data with a fluorescence reader. This assay can run up to four samples per plate in 2h, which is time and cost effective compared with other FcRn binding methods such as cell-based fluorescent-activated cell scan and surface plasma resonance. Our data demonstrated that this assay is suitable for assessing the FcRn binding in vitro and provides a platform approach that can be readily applied to various antibodies.
Collapse
Affiliation(s)
- Qiang Wu
- Biological Technologies, Analytical Development & Quality Control, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ho Young Lee
- Biological Technologies, Analytical Development & Quality Control, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Pin Yee Wong
- Biological Technologies, Analytical Development & Quality Control, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Guoying Jiang
- Biological Technologies, Analytical Development & Quality Control, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Hélène Gazzano-Santoro
- Biological Technologies, Analytical Development & Quality Control, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
105
|
Targeting FcRn for the modulation of antibody dynamics. Mol Immunol 2015; 67:131-41. [PMID: 25766596 DOI: 10.1016/j.molimm.2015.02.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 01/08/2023]
Abstract
The MHC class I-related receptor, FcRn, is a multitasking protein that transports its IgG ligand within and across cells of diverse origins. The role of this receptor as a global regulator of IgG homeostasis and transport, combined with knowledge of the molecular details of FcRn-IgG interactions, has led to opportunities to modulate the in vivo dynamics of antibodies and their antigens through protein engineering. Consequently, the generation of half-life extended antibodies has shown a rapid expansion over the past decade. Further, FcRn itself can be targeted by inhibitors to induce decreased levels of circulating IgGs, which could have applications in multiple clinical settings. The engineering of antibody-antigen interactions to reduce antibody-mediated buffering of soluble ligand has also developed into an active area of investigation, leading to novel antibody platforms designed to result in more effective antigen clearance. Similarly, the target-mediated elimination of antibodies by internalizing, membrane bound antigens (receptors) can be decreased using novel engineering approaches. These strategies, combined with subcellular trafficking analyses of antibody/antigen/FcRn behavior in cells to predict in vivo behavior, have considerable promise for the production of next generation therapeutics and diagnostics.
Collapse
|
106
|
Morris GC, Wiggins RC, Woodhall SC, Bland JM, Taylor CR, Jespers V, Vcelar BA, Lacey CJ. MABGEL 1: first phase 1 trial of the anti-HIV-1 monoclonal antibodies 2F5, 4E10 and 2G12 as a vaginal microbicide. PLoS One 2014; 9:e116153. [PMID: 25546420 PMCID: PMC4278856 DOI: 10.1371/journal.pone.0116153] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 12/03/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) which potently neutralize a broad range of HIV isolates are potential microbicide candidates. To date, topical application of mAbs in humans and their stability in vaginal secretions has not been studied. OBJECTIVES To assess the pharmacokinetics and safety of the mAbs 2F5, 4E10 and 2G12 when applied vaginally in women. DESIGN A randomized, double-blind, placebo-controlled phase 1 trial. METHODS Twenty-eight healthy, sexually abstinent women administered 2.5 g of gel daily for 12 days containing either 10 or 20 mg/g of each mAb (MABGEL) or placebo. Main clinical evaluations and sampling occurred at baseline, 1, 8, and 24 hours post-1st dose and 12 and 36 hours post-12th dose. RESULTS After adjustment for dilution factors, median levels of 2F5, 4E10 and 2G12 in vaginal secretions at 1 hour post high-dose MABGEL were 7.74, 5.28 and 7.48 mg/ml respectively. Levels of 2F5 and 4E10 declined exponentially thereafter with similar estimated half-lives (4.6 and 4.3 hours). In contrast, 2G12 levels declined more rapidly in the first 8 hours, with an estimated half-life of 1.4 hours during this period. There was no evidence of systemic absorption. There were no significant differences in local or systemic adverse event rates or vaginal flora changes (by qPCR) between active and placebo gel arms. Whilst at least 1 adverse event was recorded in 96% of participants, 95% were mild and none were serious. CONCLUSIONS Vaginal application of 50 mg of each mAb daily was safe over a 12 day period. Median mAb concentrations detected at 8 hours post dose were potentially sufficient to block HIV transmission.2G12 exhibited more rapid elimination from the human vagina than 4E10 and 2F5, likely due to poor stability of 2G12 in acidic human vaginal secretions. Further research is needed to develop mAb-based vaginal microbicides and delivery systems. TRIAL REGISTRATION ISRCTN 64808733 UK CRN Portfolio 6470.
Collapse
Affiliation(s)
- Georgina C. Morris
- Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
| | - Rebecca C. Wiggins
- Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
| | - Sarah C. Woodhall
- Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
| | - J. Martin Bland
- Department of Health Sciences, University of York, York, United Kingdom
| | - Carol R. Taylor
- Hull York Medical School Experimental Medicine Unit, York Teaching Hospitals NHS Foundation Trust, York, United Kingdom
| | | | | | - Charles J. Lacey
- Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
107
|
Borrok MJ, Wu Y, Beyaz N, Yu XQ, Oganesyan V, Dall'Acqua WF, Tsui P. pH-dependent binding engineering reveals an FcRn affinity threshold that governs IgG recycling. J Biol Chem 2014; 290:4282-90. [PMID: 25538249 DOI: 10.1074/jbc.m114.603712] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Fc domain of IgG has been the target of multiple mutational studies aimed at altering the pH-dependent IgG/FcRn interaction to modulate IgG pharmacokinetics. These studies have yielded antibody variants with disparate pharmacokinetic characteristics, ranging from extended in vivo half-life to those exhibiting extremely rapid clearance. To better understand pH-dependent binding parameters that govern these outcomes and limit FcRn-mediated half-life extension, we generated a panel of novel Fc variants with high affinity binding at acidic pH that vary in pH 7.4 affinities and assessed pharmacokinetic outcomes. Pharmacokinetic studies in human FcRn transgenic mice and cynomolgus monkeys showed that multiple variants with increased FcRn affinities at acidic pH exhibited extended serum half-lives relative to the parental IgG. Importantly, the results reveal an underappreciated affinity threshold of neutral pH binding that determines IgG recycling efficiency. Variants with pH 7.4 FcRn affinities below this threshold recycle efficiently and can exhibit increased serum persistence. Increasing neutral pH FcRn affinity beyond this threshold reduced serum persistence by offsetting the benefits of increased pH 6.0 binding. Ultra-high affinity binding to FcRn at both acidic and neutral pH leads to rapid serum clearance.
Collapse
Affiliation(s)
- M Jack Borrok
- From the Departments of Antibody Discovery and Protein Engineering and
| | - Yanli Wu
- From the Departments of Antibody Discovery and Protein Engineering and
| | - Nurten Beyaz
- From the Departments of Antibody Discovery and Protein Engineering and
| | - Xiang-Qing Yu
- Clinical Pharmacology and Drug Metabolism and Pharmacokinetics, MedImmune, Inc., Gaithersburg, Maryland 20878
| | - Vaheh Oganesyan
- From the Departments of Antibody Discovery and Protein Engineering and
| | | | - Ping Tsui
- From the Departments of Antibody Discovery and Protein Engineering and
| |
Collapse
|
108
|
Matsushima S, Huang Y, Suzuki H, Nishino J, Lloyd P. Ethnic sensitivity assessment – pharmacokinetic comparability between Japanese and non-Japanese healthy subjects on selected mAbs. Expert Opin Drug Metab Toxicol 2014; 11:179-91. [DOI: 10.1517/17425255.2015.990438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
109
|
Jensen PF, Larraillet V, Schlothauer T, Kettenberger H, Hilger M, Rand KD. Investigating the interaction between the neonatal Fc receptor and monoclonal antibody variants by hydrogen/deuterium exchange mass spectrometry. Mol Cell Proteomics 2014; 14:148-61. [PMID: 25378534 DOI: 10.1074/mcp.m114.042044] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The recycling of immunoglobulins by the neonatal Fc receptor (FcRn) is of crucial importance in the maintenance of antibody levels in plasma and is responsible for the long half-lives of endogenous and recombinant monoclonal antibodies. From a therapeutic point of view there is great interest in understanding and modulating the IgG-FcRn interaction to optimize antibody pharmacokinetics and ultimately improve efficacy and safety. Here we studied the interaction between a full-length human IgG(1) and human FcRn via hydrogen/deuterium exchange mass spectrometry and targeted electron transfer dissociation to map sites perturbed by binding on both partners of the IgG-FcRn complex. Several regions in the antibody Fc region and the FcRn were protected from exchange upon complex formation, in good agreement with previous crystallographic studies of FcRn in complex with the Fc fragment. Interestingly, we found that several regions in the IgG Fab region also showed reduced deuterium uptake. Our findings indicate the presence of hitherto unknown FcRn interaction sites in the Fab region or a possible conformational link between the IgG Fc and Fab regions upon FcRn binding. Further, we investigated the role of IgG glycosylation in the conformational response of the IgG-FcRn interaction. Removal of antibody glycans increased the flexibility of the FcRn binding site in the Fc region. Consequently, FcRn binding did not induce a similar conformational stabilization of deglycosylated IgG as observed for the wild-type glycosylated IgG. Our results provide new molecular insight into the IgG-FcRn interaction and illustrate the capability of hydrogen/deuterium exchange mass spectrometry to advance structural proteomics by providing detailed information on the conformation and dynamics of large protein complexes in solution.
Collapse
Affiliation(s)
- Pernille Foged Jensen
- From the ‡Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Vincent Larraillet
- §Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Nonnenwald 2, 82377 Penzberg, Germany
| | - Tilman Schlothauer
- §Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Nonnenwald 2, 82377 Penzberg, Germany
| | - Hubert Kettenberger
- §Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Nonnenwald 2, 82377 Penzberg, Germany
| | - Maximiliane Hilger
- §Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Nonnenwald 2, 82377 Penzberg, Germany
| | - Kasper D Rand
- From the ‡Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark;
| |
Collapse
|
110
|
Datta-Mannan A, Wroblewski VJ. Application of FcRn binding assays to guide mAb development. Drug Metab Dispos 2014; 42:1867-72. [PMID: 25024401 DOI: 10.1124/dmd.114.059089] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Monoclonal antibodies (mAbs) represent an important class of therapeutic modalities. To optimize their pharmaceutical properties, studies have focused on improving mAb pharmacokinetic/pharmacodynamic profiles by modulating their interactions with the neonatal Fc receptor (FcRn). The influence of both the chemical and physical properties of IgGs has been examined in the context of FcRn interactions. In this regard, a variety of FcRn binding assays and tools have been developed and used to characterize the interaction with IgGs. However, a predictive relationship between the FcRn binding interaction of IgGs in vitro and their pharmacokinetics in vivo broadly across mAbs remains elusive. Many studies have increasingly suggested that the interplay between the characteristics of the mAb and the nature of its target can influence disposition and elimination. Thus, it is becoming increasingly evident that along with FcRn interactions, consideration of the non-FcRn-based biologic processes active in mAb disposition should be integrated into mAb development and optimization. Herein, we describe how the pharmacokinetics of mAbs can be modulated through FcRn interactions and provide perspectives on interpreting the receptor binding parameters in relation to other mechanisms involved in antibody disposition to aid in guiding mAb development.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Department of Drug Disposition Development/Commercialization, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| | - Victor J Wroblewski
- Department of Drug Disposition Development/Commercialization, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| |
Collapse
|
111
|
Scott BJ, Klein AV, Wang J. Biosimilar monoclonal antibodies: A Canadian regulatory perspective on the assessment of clinically relevant differences and indication extrapolation. J Clin Pharmacol 2014; 55 Suppl 3:S123-32. [PMID: 24965228 DOI: 10.1002/jcph.339] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/01/2014] [Indexed: 12/21/2022]
Abstract
Monoclonal antibodies have become mainstays of treatment for many diseases. After more than a decade on the Canadian market, a number of authorized monoclonal antibody products are facing patent expiry. Given their success, most notably in the areas of oncology and autoimmune disease, pharmaceutical and biotechnology companies are eager to produce their own biosimilar versions and have begun manufacturing and testing for a variety of monoclonal antibody products. In October of 2013, the first biosimilar monoclonal antibody products were approved by the European Medicines Agency (Remsima™ and Inflectra™). These products were authorized by Health Canada shortly after; however, while the EMA allowed for extrapolation to all of the indications held by the reference product, Health Canada limited extrapolation to a subset of the indications held by the reference product, Remicade®. The purpose of this review is to discuss the Canadian regulatory framework for the authorization of biosimilar mAbs with specific discussion around the clinical requirements for establishing (bio)-similarity and to present the principles that are used in the clinical assessment of New Drug Submissions for intended biosimilar monoclonal antibodies. Health Canada's current views regarding indication extrapolation, product interchangeability, and post-market surveillance are discussed as well.
Collapse
Affiliation(s)
- Bradley J Scott
- Centre for the Evaluation of Radiopharmaceuticals and Biotherapeutics, Biologics and Genetic Therapies Directorate, Health Products and Food Branch, Health Canada, Ottawa, K1A 0K9, Canada
| | | | | |
Collapse
|
112
|
Drake PM, Albers AE, Baker J, Banas S, Barfield RM, Bhat AS, de Hart GW, Garofalo AW, Holder P, Jones LC, Kudirka R, McFarland J, Zmolek W, Rabuka D. Aldehyde tag coupled with HIPS chemistry enables the production of ADCs conjugated site-specifically to different antibody regions with distinct in vivo efficacy and PK outcomes. Bioconjug Chem 2014; 25:1331-41. [PMID: 24924618 PMCID: PMC4215875 DOI: 10.1021/bc500189z] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
It is becoming increasingly clear
that site-specific conjugation
offers significant advantages over conventional conjugation chemistries
used to make antibody–drug conjugates (ADCs). Site-specific
payload placement allows for control over both the drug-to-antibody
ratio (DAR) and the conjugation site, both of which play an important
role in governing the pharmacokinetics (PK), disposition, and efficacy
of the ADC. In addition to the DAR and site of conjugation, linker
composition also plays an important role in the properties of an ADC.
We have previously reported a novel site-specific conjugation platform
comprising linker payloads designed to selectively react with site-specifically
engineered aldehyde tags on an antibody backbone. This chemistry results
in a stable C–C bond between the antibody and the cytotoxin
payload, providing a uniquely stable connection with respect to the
other linker chemistries used to generate ADCs. The flexibility and
versatility of the aldehyde tag conjugation platform has enabled us
to undertake a systematic evaluation of the impact of conjugation
site and linker composition on ADC properties. Here, we describe the
production and characterization of a panel of ADCs bearing the aldehyde
tag at different locations on an IgG1 backbone conjugated using Hydrazino-iso-Pictet-Spengler (HIPS) chemistry. We demonstrate that
in a panel of ADCs with aldehyde tags at different locations, the
site of conjugation has a dramatic impact on in vivo efficacy and
pharmacokinetic behavior in rodents; this advantage translates to
an improved safety profile in rats as compared to a conventional lysine
conjugate.
Collapse
Affiliation(s)
- Penelope M Drake
- Redwood Bioscience , 5703 Hollis Street, Emeryville, California 94608, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Neuber T, Frese K, Jaehrling J, Jäger S, Daubert D, Felderer K, Linnemann M, Höhne A, Kaden S, Kölln J, Tiller T, Brocks B, Ostendorp R, Pabst S. Characterization and screening of IgG binding to the neonatal Fc receptor. MAbs 2014; 6:928-42. [PMID: 24802048 DOI: 10.4161/mabs.28744] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The neonatal Fc receptor (FcRn) protects immunoglobulin G (IgG) from degradation and increases the serum half-life of IgG, thereby contributing to a higher concentration of IgG in the serum. Because altered FcRn binding may result in a reduced or prolonged half-life of IgG molecules, it is advisable to characterize Fc receptor binding of therapeutic antibody lead candidates prior to the start of pre-clinical and clinical studies. In this study, we characterized the interactions between FcRn of different species (human, cynomolgus monkey, mouse and rat) and nine IgG molecules from different species and isotypes with common variable heavy (VH) and variable light chain (VL) domains. Binding was analyzed at acidic and neutral pH using surface plasmon resonance (SPR) and biolayer interferometry (BLI). Furthermore, we transferred the well-accepted, but low throughput SPR-based method for FcRn binding characterization to the BLI-based Octet platform to enable a higher sample throughput allowing the characterization of FcRn binding already during early drug discovery phase. We showed that the BLI-based approach is fit-for-purpose and capable of discriminating between IgG molecules with significant differences in FcRn binding affinities. Using this high-throughput approach we investigated FcRn binding of 36 IgG molecules that represented all VH/VL region combinations available in the fully human, recombinant antibody library Ylanthia®. Our results clearly showed normal FcRn binding profiles for all samples. Hence, the variations among the framework parts, complementarity-determining region (CDR) 1 and CDR2 of the fragment antigen binding (Fab) domain did not significantly change FcRn binding.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anne Höhne
- MorphoSys AG; Martinsried/Planegg, Germany
| | | | | | | | | | | | | |
Collapse
|
114
|
da Silva A, Kronthaler U, Koppenburg V, Fink M, Meyer I, Papandrikopoulou A, Hofmann M, Stangler T, Visser J. Target-directed development and preclinical characterization of the proposed biosimilar rituximab GP2013. Leuk Lymphoma 2014; 55:1609-17. [PMID: 24024472 PMCID: PMC4133973 DOI: 10.3109/10428194.2013.843090] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Biosimilar development involves a target-directed iterative process to ensure a similar product to the originator. Here we report the preclinical development of the proposed biosimilar rituximab (GP2013). Post-translational modifications and bioactivities of GP2013 versus originator rituximab were engineered and monitored to ensure similar pharmacological profiles. Antibody-dependent cellular cytotoxicity (ADCC) was used to illustrate how different glycosylation patterns and structure–function relationships were controlled during process development. Pharmacological comparability between GP2013 and originator rituximab were confirmed in preclinical studies using clinical scale drug product. Similar in vitro ADCC potency was demonstrated when compared in a dose–response manner against two lymphoma cell lines using freshly purified human natural killer (NK) cells. In vivo efficacy was demonstrated in two well characterized mouse xenograft models, testing at sensitive sub-therapeutic dose levels. Pharmacokinetics and pharmacodynamics (CD20 cell depletion) were likewise comparable in cynomolgus monkeys. This preclinical comparability exercise confirms that GP2013 and originator rituximab are pharmacologically similar.
Collapse
|
115
|
Monnet C, Jorieux S, Souyris N, Zaki O, Jacquet A, Fournier N, Crozet F, de Romeuf C, Bouayadi K, Urbain R, Behrens CK, Mondon P, Fontayne A. Combined glyco- and protein-Fc engineering simultaneously enhance cytotoxicity and half-life of a therapeutic antibody. MAbs 2014; 6:422-36. [PMID: 24492301 DOI: 10.4161/mabs.27854] [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: 12/24/2022] Open
Abstract
While glyco-engineered monoclonal antibodies (mAbs) with improved antibody-dependent cell-mediated cytotoxicity (ADCC) are reaching the market, extensive efforts have also been made to improve their pharmacokinetic properties to generate biologically superior molecules. Most therapeutic mAbs are human or humanized IgG molecules whose half-life is dependent on the neonatal Fc receptor FcRn. FcRn reduces IgG catabolism by binding to the Fc domain of endocytosed IgG in acidic lysosomal compartments, allowing them to be recycled into the blood. Fc-engineered mAbs with increased FcRn affinity resulted in longer in vivo half-life in animal models, but also in healthy humans. These Fc-engineered mAbs were obtained by alanine scanning, directed mutagenesis or in silico approach of the FcRn binding site. In our approach, we applied a random mutagenesis technology (MutaGen™) to generate mutations evenly distributed over the whole Fc sequence of human IgG1. IgG variants with improved FcRn-binding were then isolated from these Fc-libraries using a pH-dependent phage display selection process. Two successive rounds of mutagenesis and selection were performed to identify several mutations that dramatically improve FcRn binding. Notably, many of these mutations were unpredictable by rational design as they were located distantly from the FcRn binding site, validating our random molecular approach. When produced on the EMABling(®) platform allowing effector function increase, our IgG variants retained both higher ADCC and higher FcRn binding. Moreover, these IgG variants exhibited longer half-life in human FcRn transgenic mice. These results clearly demonstrate that glyco-engineering to improve cytotoxicity and protein-engineering to increase half-life can be combined to further optimize therapeutic mAbs.
Collapse
|
116
|
Hötzel I, Theil FP, Bernstein LJ, Prabhu S, Deng R, Quintana L, Lutman J, Sibia R, Chan P, Bumbaca D, Fielder P, Carter PJ, Kelley RF. A strategy for risk mitigation of antibodies with fast clearance. MAbs 2014; 4:753-60. [PMID: 23778268 DOI: 10.4161/mabs.22189] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
A majority of human therapeutic antibody candidates show pharmacokinetic properties suitable for clinical use, but an unexpectedly fast antibody clearance is sometimes observed that may limit the clinical utility. Pharmacokinetic data in cynomolgus monkeys collected for a panel of 52 antibodies showed broad distribution of target-independent clearance values (2.4-61.3 mL/day/kg), with 15 (29%) having clearance > 10 mL/day/kg. Alteration in the interaction with the recycling FcRn receptor did not account for the faster than expected clearance observed for the antibodies; off-target binding was presumed to account for the fast clearance. We developed an assay based on ELISA detection of non-specific binding to baculovirus particles that can identify antibodies having increased risk for fast clearance. This assay can be used during lead generation or optimization to identify antibodies with increased risk of having fast clearance in both humans and cynomolgus monkeys, and thus increase the likelihood of obtaining a suitable drug candidate.
Collapse
Affiliation(s)
- Isidro Hötzel
- Antibody Engineering Department, Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Wang Y, Tian Z, Thirumalai D, Zhang X. Neonatal Fc receptor (FcRn): a novel target for therapeutic antibodies and antibody engineering. J Drug Target 2014; 22:269-78. [PMID: 24404896 DOI: 10.3109/1061186x.2013.875030] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The biomedical applications of antibodies as prophylactics, therapeutics and diagnostics are developing rapidly. Neonatal Fc receptor (FcRn) is a major IgG Fc receptor capable of facilitating the translocation of IgG. FcRn can protect IgG from intracellular catabolism, thereby increasing its half-life. In recent decade, the interaction between FcRn and the Fc region has been reported with the focuses on either prolonging the plasma half-life of therapeutic IgG or shortening the half-life of pathogenic IgG. The FcRn-IgG interaction can be altered by modifying the Fc region to change their affinity (increase or decrease), and/or by reducing the Fc fragments of IgG to enhance its penetration into tissues or cells. By over expression of FcRn, the exogenous catabolism can be reduced, meanwhile the circulating IgG level could be enhanced. It has been confirmed in different FcRn over-expressed transgenic mice models, substantial humoral responses against antigens with weak immunogenicity can be mounted. In addition, designing inhibitors for FcRn-IgG interaction is another application prospect for treating IgG-mediated autoimmune diseases. Recent research advancements strengthen the understanding that FcRn is a key and promising drugable target in IgG intervention in the field of antibody engineering.
Collapse
Affiliation(s)
- Yuan Wang
- College of Veterinary Medicine, Northwest A&F University , Yangling, Shannxi Province , P.R. China
| | | | | | | |
Collapse
|
118
|
Crystal Structure of an HSA/FcRn Complex Reveals Recycling by Competitive Mimicry of HSA Ligands at a pH-Dependent Hydrophobic Interface. Structure 2013; 21:1966-78. [DOI: 10.1016/j.str.2013.08.022] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/17/2013] [Accepted: 08/23/2013] [Indexed: 11/23/2022]
|
119
|
A novel investigational Fc-modified humanized monoclonal antibody, motavizumab-YTE, has an extended half-life in healthy adults. Antimicrob Agents Chemother 2013; 57:6147-53. [PMID: 24080653 DOI: 10.1128/aac.01285-13] [Citation(s) in RCA: 263] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The study objective was to evaluate the pharmacokinetics (PK), antidrug antibody (ADA), and safety of motavizumab-YTE (motavizumab with amino acid substitutions M252Y/S254T/T256E [YTE]), an Fc-modified anti-respiratory syncytial virus (RSV) monoclonal antibody. Healthy adults (n = 31) were randomized to receive a single intravenous (i.v.) dose of motavizumab-YTE or motavizumab (0.3, 3, 15, or 30 mg/kg) and followed for 240 days. Clearance of motavizumab-YTE was significantly lower (71% to 86%) and the half-life (t1/2) was 2- to 4-fold longer than with motavizumab. However, similar peak concentrations and volume-of-distribution values, indicative of similar distribution properties, were seen at all dose levels. The sustained serum concentrations of motavizumab-YTE were fully functional, as shown by RSV neutralizing activity that persisted for 240 days with motavizumab-YTE versus 90 days postdose for motavizumab. Safety and incidence of ADA were comparable between groups. In this first study of an Fc-modified monoclonal antibody in humans, motavizumab-YTE was well tolerated and exhibited an extended half-life of up to 100 days. (This study has been registered at ClinicalTrials.gov under registration no. NCT00578682.).
Collapse
|
120
|
Huang X, Zheng F, Zhan CG. Binding structures and energies of the human neonatal Fc receptor with human Fc and its mutants by molecular modeling and dynamics simulations. MOLECULAR BIOSYSTEMS 2013; 9:3047-58. [PMID: 24057047 DOI: 10.1039/c3mb70231f] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Homology modeling and molecular dynamics simulations have been carried out to model the detailed structures of the human neonatal Fc receptor (FcRn) binding with the wild-type Fc of human immunoglobulin G1 (IgG1) and its various mutants. Based on the modeled human FcRn-Fc binding structures, it has been proposed that the protein-protein binding interface is composed of three subsites. The first subsite is a hydrophobic core where residue I39 of human Fc can be accommodated very well, and the other two subsites are all composed of critical salt bridges between human FcRn and human Fc. All of the modeled structures and the calculated binding energies are qualitatively consistent with the available experimental data, suggesting that the modeled human FcRn-Fc binding structures are reasonable. The modeled human FcRn-Fc binding structure may be valuable for future rational design of novel mutants of human Fc and Fc-fused therapeutic proteins with a potentially higher binding affinity for human FcRn and, thus, a longer in vivo half-life in humans.
Collapse
Affiliation(s)
- Xiaoqin Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, USA.
| | | | | |
Collapse
|
121
|
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
|
122
|
ADME of monoclonal antibody biotherapeutics: knowledge gaps and emerging tools. Bioanalysis 2013; 5:2003-14. [DOI: 10.4155/bio.13.144] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Absorption, distribution, metabolism and excretion (ADME) data are pivotal for small-molecule drug development, with well-developed in vitro and in vivo correlation tools and guidances from regulatory agencies. In the past two decades, monoclonal antibody (mAb) biotherapeutics have been successfully approved, including derived novel conjugates of active molecules (toxins or bioactive peptides) for specific target delivery or half-life extension. However, ADME information of mAb therapeutics lags behind that of small molecules due to the complex nature of the molecules and lack of appropriate tools to study drug exposure, biotransformation, and target engagement in the vascular and tissue spaces. In this perspective, the current knowledge gaps on ADME of mAb-related therapeutics are reviewed with potential solutions from emerging analytical technologies.
Collapse
|
123
|
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
|
124
|
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
|
125
|
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
|
126
|
Schlothauer T, Rueger P, Stracke JO, Hertenberger H, Fingas F, Kling L, Emrich T, Drabner G, Seeber S, Auer J, Koch S, Papadimitriou A. Analytical FcRn affinity chromatography for functional characterization of monoclonal antibodies. MAbs 2013; 5:576-86. [PMID: 23765230 DOI: 10.4161/mabs.24981] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The neonatal Fc receptor (FcRn) is important for the metabolic fate of IgG antibodies in vivo. Analysis of the interaction between FcRn and IgG in vitro might provide insight into the structural and functional integrity of therapeutic IgG that may affect pharmacokinetics (PK) in vivo. We developed a standardized pH gradient FcRn affinity liquid chromatography method with conditions closely resembling the physiological mechanism of interaction between IgG and FcRn. This method allows the separation of molecular IgG isoforms, degradation products and engineered molecules based on their affinity to FcRn. Human FcRn was immobilized on the column and a linear pH gradient from pH 5.5 to 8.8 was applied. FcRn chromatography was used in comparison to surface plasmon resonance to characterize different monoclonal IgG preparations, e.g., oxidized or aggregated species. Wild-type and engineered IgGs were compared in vitro by FcRn chromatography and in vivo by PK studies in huFcRn transgenic mice. Analytical FcRn chromatography allows differentiation of IgG samples and variants by peak pattern and retention time profile. The method can distinguish: 1) IgGs with different Fabs, 2) oxidized from native IgG, 3) aggregates from monomer and 4) antibodies with mutations in the Fc part from wild-type IgGs. Changes in the FcRn chromatographic behavior of mutant IgGs relative to the wild-type IgG correlate to changes in the PK profile in the FcRn transgenic mice. These results demonstrate that FcRn affinity chromatography is a useful new method for the assessment of IgG integrity.
Collapse
Affiliation(s)
- Tilman Schlothauer
- Department of Protein Analytics, Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Ishino T, Wang M, Mosyak L, Tam A, Duan W, Svenson K, Joyce A, O'Hara DM, Lin L, Somers WS, Kriz R. Engineering a monomeric Fc domain modality by N-glycosylation for the half-life extension of biotherapeutics. J Biol Chem 2013; 288:16529-16537. [PMID: 23615911 DOI: 10.1074/jbc.m113.457689] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human IgG is a bivalent molecule that has two identical Fab domains connected by a dimeric Fc domain. For therapeutic purposes, however, the bivalency of IgG and Fc fusion proteins could cause undesired properties. We therefore engineered the conversion of the natural dimeric Fc domain to a highly soluble monomer by introducing two Asn-linked glycans onto the hydrophobic C(H)3-C(H)3 dimer interface. The monomeric Fc (monoFc) maintained the binding affinity for neonatal Fc receptor (FcRn) in a pH-dependent manner. We solved the crystal structure of monoFc, which explains how the carbohydrates can stabilize the protein surface and provides the rationale for molecular recognition between monoFc and FcRn. The monoFc prolonged the in vivo half-life of an antibody Fab domain, and a tandem repeat of the monoFc further prolonged the half-life. This monoFc modality can be used to improve the pharmacokinetics of monomeric therapeutic proteins with an option to modulate the degree of half-life extension.
Collapse
Affiliation(s)
- Tetsuya Ishino
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140.
| | - Mengmeng Wang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Andover, Massachusetts 01810
| | - Lidia Mosyak
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| | - Amy Tam
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| | - Weili Duan
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| | - Kristine Svenson
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| | - Alison Joyce
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Andover, Massachusetts 01810
| | - Denise M O'Hara
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Andover, Massachusetts 01810
| | - Laura Lin
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| | - William S Somers
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| | - Ronald Kriz
- From Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, Massachusetts 02140
| |
Collapse
|
128
|
Tam SH, McCarthy SG, Brosnan K, Goldberg KM, Scallon BJ. Correlations between pharmacokinetics of IgG antibodies in primates vs. FcRn-transgenic mice reveal a rodent model with predictive capabilities. MAbs 2013; 5:397-405. [PMID: 23549129 DOI: 10.4161/mabs.23836] [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] [Indexed: 11/19/2022] Open
Abstract
Transgenic mice expressing human neonatal Fc receptor (FcRn) instead of mouse FcRn are available for IgG antibody pharmacokinetic (PK) studies. Given the interest in a rodent model that offers reliable predictions of antibody PK in monkeys and humans, we set out to test whether the PK of IgG antibodies in such mice correlated with the PK of the same antibodies in primates. We began by using a single research antibody to study the influence of: (1) different transgenic mouse lines that differ in FcRn transgene expression; (2) homozygous vs. hemizygous FcRn transgenic mice; (3) the presence vs. absence of coinjected high-dose human intravenous immunoglobulin (IVIG), and (4) the presence vs. absence of coinjected high-dose human serum albumin (HSA). Results of those studies suggested that use of hemizygous Tg32 mice (Tg32 hemi) not treated with IVIG or HSA offered potential as a predictive model for PK in humans. Mouse PK studies were then done under those conditions with a panel of test antibodies whose PK in mice and primates is not significantly affected by target binding, and for which monkey or human PK data were readily available. Results from the studies revealed significant correlations between terminal half-life or clearance values observed in the mice and the corresponding values reported in humans. A significant relationship in clearance values between mice and monkeys was also observed. These correlations suggest that the Tg32 hemi mouse model, which is both convenient and cost-effective, can offer value in predicting antibody half-life and clearance in primates.
Collapse
Affiliation(s)
- Susan H Tam
- Biologics Research; Janssen Research & Development, LLC; Spring House, PA USA
| | - Stephen G McCarthy
- Biologics Research; Janssen Research & Development, LLC; Spring House, PA USA
| | - Kerry Brosnan
- Biologics Toxicology; Janssen Research & Development, LLC; Spring House, PA USA
| | - Kenneth M Goldberg
- Nonclinical Statistics & Computing; Janssen Research & Development, LLC; Spring House, PA USA
| | - Bernard J Scallon
- Biologics Research; Janssen Research & Development, LLC; Spring House, PA USA
| |
Collapse
|
129
|
Aggregates, crystals, gels, and amyloids: intracellular and extracellular phenotypes at the crossroads of immunoglobulin physicochemical property and cell physiology. Int J Cell Biol 2013; 2013:604867. [PMID: 23533417 PMCID: PMC3603282 DOI: 10.1155/2013/604867] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 01/27/2013] [Indexed: 12/20/2022] Open
Abstract
Recombinant immunoglobulins comprise an important class of human therapeutics. Although specific immunoglobulins can be purposefully raised against desired antigen targets by various methods, identifying an immunoglobulin clone that simultaneously possesses potent therapeutic activities and desirable manufacturing-related attributes often turns out to be challenging. The variable domains of individual immunoglobulins primarily define the unique antigen specificities and binding affinities inherent to each clone. The primary sequence of the variable domains also specifies the unique physicochemical properties that modulate various aspects of individual immunoglobulin life cycle, starting from the biosynthetic steps in the endoplasmic reticulum, secretory pathway trafficking, secretion, and the fate in the extracellular space and in the endosome-lysosome system. Because of the diverse repertoire of immunoglobulin physicochemical properties, some immunoglobulin clones' intrinsic properties may manifest as intriguing cellular phenotypes, unusual solution behaviors, and serious pathologic outcomes that are of scientific and clinical importance. To gain renewed insights into identifying manufacturable therapeutic antibodies, this paper catalogs important intracellular and extracellular phenotypes induced by various subsets of immunoglobulin clones occupying different niches of diverse physicochemical repertoire space. Both intrinsic and extrinsic factors that make certain immunoglobulin clones desirable or undesirable for large-scale manufacturing and therapeutic use are summarized.
Collapse
|
130
|
Mathur A, Arora T, Liu L, Crouse-Zeineddini J, Mukku V. Qualification of a homogeneous cell-based neonatal Fc receptor (FcRn) binding assay and its application to studies on Fc functionality of IgG-based therapeutics. J Immunol Methods 2013; 390:81-91. [PMID: 23384837 DOI: 10.1016/j.jim.2013.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 10/20/2012] [Accepted: 01/16/2013] [Indexed: 11/16/2022]
Abstract
The Fc region of IgG-based molecules plays an important role in determining their in vivo pharmacokinetic profile by its pH-dependent binding to the neonatal Fc receptor (FcRn) which is expressed on the endothelial cells lining blood vessels. By virtue of this pH-specific interaction with IgG-Fc, FcRn mediates IgG homeostasis in human adults by maintaining serum IgG levels, and also transfers maternal IgGs from mother to fetus via the placenta. The Fc-FcRn interaction is also critical for keeping IgG-based therapeutic molecules in circulation thereby enhancing their serum half life. A homogeneous cell-based flow cytometric FcRn binding assay was established to characterize the Fc-FcRn interaction of therapeutic IgG-based molecules. It is a competition-based assay, wherein the IgG-Fc containing test molecule competes with a fixed concentration of fluorescently-labeled IgG-Fc moiety in solution for binding to the cell-expressed FcRn. The cell-bound fluorescence is read on a flow cytometer. Response of the test sample is analyzed relative to the standard sample and the results are reported as % relative binding. The assay is robust and meets the qualification criteria for specificity, method linearity, accuracy and precision over the relative binding range of 60%-160%. This assay was shown to effectively characterize altered Fc-FcRn interactions for photo-stressed, heat-stressed, oxidized, and Fc mutant samples. It was observed that the relative binding of the IgG-Fc to the cell-surface-expressed FcRn in the assay varies across different molecules, even within the same IgG subclass. This indicates that the Fc-FcRn binding can be influenced by the antigen-binding region of the molecules in addition to the IgG subclass. Overall, this assay is reflective of the in vivo mechanism of immunoglobulin binding to membrane-bound FcRn, and can be used as an analytical tool for assessing lot-to-lot consistency and stability testing across different batches of the same molecule. Additionally, the assay can be used as an effective tool for elucidating the amino acids in the IgG-Fc domain that are critical for FcRn binding and also for comparing the binding of different IgG-Fc containing molecules to FcRn.
Collapse
Affiliation(s)
- Abhishek Mathur
- Bioassay and Biological Characterization, Amgen, Inc., One Amgen Center Dr, Thousand Oaks, CA 91320, United States.
| | | | | | | | | |
Collapse
|
131
|
Rossi EA, Chang CH, Cardillo TM, Goldenberg DM. Optimization of multivalent bispecific antibodies and immunocytokines with improved in vivo properties. Bioconjug Chem 2012; 24:63-71. [PMID: 23116517 DOI: 10.1021/bc300488f] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Multifunctional antibody-based biologics, such as bispecific antibodies and immunocytokines, can be difficult to produce with sufficient yield and stability, and often exhibit inferior pharmacokinetics. Dock-and-Lock (DNL) is a modular method that combines recombinant engineering with site-specific conjugation, allowing the construction of various complex, yet defined, biostructures with multivalency and multispecificity. The technology platform exploits the natural interaction between two interactive human protein binding domains that are modified to provide covalent fusion. We explored the potential application of a new class of IgG-based DNL modules with an anchor domain fused at the C-terminal end of the kappa light chain (C(k)), instead of the C-terminal end of the Fc. Two C(k)-derived prototypes, an anti-CD22/CD20 bispecific hexavalent antibody, comprising epratuzumab (anti-CD22) and four Fabs of veltuzumab (anti-CD20), and a CD20-targeting immunocytokine, comprising veltuzumab and four molecules of interferon-α2b, were compared to their Fc-derived counterparts. The Ck-based conjugates exhibited superior Fc-effector functions in vitro, as well as improved pharmacokinetics, stability, and anti-lymphoma activity in vivo. These results favor the selection of DNL conjugates with the C(k)-design for future clinical development.
Collapse
|
132
|
The Impact of Glycosylation on the Pharmacokinetics of a TNFR2:Fc Fusion Protein Expressed in Glycoengineered Pichia Pastoris. Pharm Res 2012; 30:803-12. [DOI: 10.1007/s11095-012-0921-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 10/18/2012] [Indexed: 10/27/2022]
|
133
|
Thorn M, Piche-Nicholas N, Stedman D, Davenport SW, Zhang N, Collinge M, Bowman CJ. Embryo-fetal transfer of bevacizumab (Avastin) in the rat over the course of gestation and the impact of neonatal Fc receptor (FcRn) binding. ACTA ACUST UNITED AC 2012; 95:363-75. [PMID: 22969064 DOI: 10.1002/bdrb.21026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 08/21/2012] [Indexed: 01/06/2023]
Abstract
BACKGROUND There is concern about embryo-fetal exposure to antibody-based biopharmaceuticals based on the increase of such therapies being prescribed to women of childbearing potential. Therefore, there is a desire to better characterize embryo-fetal exposure of these molecules. The pregnant rat is a standard model for evaluating the potential consequences of exposure but placental transfer of antibody-based biopharmaceuticals is not well understood in this model. METHODS The relative embryo-fetal distribution of an antibody-based biopharmaceutical was evaluated in the rat. Bevacizumab (Avastin) was chosen as a tool antibody since it does not have significant target binding in the rat that might influence embryo-fetal biodistribution. Avastin was labeled with a fluorescent dye, characterized, and injected into pregnant rats at different gestation ages. Labeled Avastin in fetal tissues was visualized ex vivo using an IVIS 200 (Caliper, A PerkinElmer Company, Alameda, CA). RESULTS Avastin localized to the fetus as early as 24-hr post intravenous injection of the dam, and was taken up by the fetus in a dose-dependent manner. Avastin was detectable in the developing embryo as early as gestation day 13 and continued to be transferred until the end of gestation. Fetal transfer of Avastins mutated in the portion of the antibody that binds the neonatal Fc receptor (FcRn) was tested in late gestation and was found to correlate with affinities of the mutant Avastin antibody to FcRn. CONCLUSIONS The novel application of this imaging technology was used to characterize the onset and duration of Avastin maternal-fetal transfer in rats and the importance of FcRn binding.
Collapse
Affiliation(s)
- Mitchell Thorn
- Drug Safety Research & Development, Pfizer, Inc., Groton, CT 06340, USA
| | | | | | | | | | | | | |
Collapse
|
134
|
Xu X, Vugmeyster Y. Challenges and opportunities in absorption, distribution, metabolism, and excretion studies of therapeutic biologics. AAPS JOURNAL 2012; 14:781-91. [PMID: 22864668 DOI: 10.1208/s12248-012-9388-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/26/2012] [Indexed: 01/18/2023]
Abstract
With the advancement of biotechnology in the last two decades, optimized and novel modalities and platforms of biologic moieties have emerged rapidly in drug discovery pipelines. In addition, new technologies for delivering therapeutic biologics (e.g., needle-free devices, nanoparticle complexes), as well as novel approaches for disease treatments (e.g., stem cell therapy, individualized medicine), continue to be developed. While pharmacokinetic studies are routinely carried out for therapeutic biologics, experiments that elucidate underlying mechanisms for clearance and biodistribution or identify key factors that govern absorption, distribution, metabolism, and excretion (ADME) of biologics often are not thoroughly conducted. Realizing the importance of biologics as therapeutic agents, pharmaceutical industry has recently begun to move the research focus from small molecules only to a blended portfolio consisting of both small molecules and biologics. This trend brings many opportunities for scientists working in the drug disposition research field. In anticipation of these opportunities and associated challenges, this review highlights impact of ADME studies on clinical and commercial success of biologics, with a particular focus on emerging applications and technologies and linkage with mechanistic pharmacokinetic/pharmacodynamic modeling and biomarker research.
Collapse
Affiliation(s)
- Xin Xu
- National Center for Translational Therapeutics, National Institutes of Health, 9800 Medical Center Dr, Rockville, Maryland 20850, USA.
| | | |
Collapse
|
135
|
Vugmeyster Y, Xu X, Theil FP, Khawli LA, Leach MW. Pharmacokinetics and toxicology of therapeutic proteins: Advances and challenges. World J Biol Chem 2012; 3:73-92. [PMID: 22558487 PMCID: PMC3342576 DOI: 10.4331/wjbc.v3.i4.73] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 01/18/2012] [Accepted: 01/25/2012] [Indexed: 02/05/2023] Open
Abstract
Significant progress has been made in understanding pharmacokinetics (PK), pharmacodynamics (PD), as well as toxicity profiles of therapeutic proteins in animals and humans, which have been in commercial development for more than three decades. However, in the PK arena, many fundamental questions remain to be resolved. Investigative and bioanalytical tools need to be established to improve the translation of PK data from animals to humans, and from in vitro assays to in vivo readouts, which would ultimately lead to a higher success rate in drug development. In toxicology, it is known, in general, what studies are needed to safely develop therapeutic proteins, and what studies do not provide relevant information. One of the major complicating factors in nonclinical and clinical programs for therapeutic proteins is the impact of immunogenicity. In this review, we will highlight the emerging science and technology, as well as the challenges around the pharmacokinetic- and safety-related issues in drug development of mAbs and other therapeutic proteins.
Collapse
Affiliation(s)
- Yulia Vugmeyster
- Yulia Vugmeyster, Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Andover, MA 01810, United States
| | | | | | | | | |
Collapse
|
136
|
Prueksaritanont T, Tang C. ADME of biologics-what have we learned from small molecules? AAPS JOURNAL 2012; 14:410-9. [PMID: 22484625 DOI: 10.1208/s12248-012-9353-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/23/2012] [Indexed: 12/14/2022]
Abstract
Thorough characterization and in-depth understanding of absorption, distribution, metabolism, and elimination (ADME) properties of a drug candidate have been well recognized as an important element in small molecule (SM) drug discovery and development. This has been the area of focus for drug metabolism and pharmacokinetics (DMPK) scientists, whose role has been evolving over the past few decades from primarily being involved in the development space after a preclinical candidate was selected to extending their involvement into the discovery stage prior to candidate selection. This paradigm shift has ensured the entry into development of the best candidates with optimal ADME properties, and thus has greatly impacted SM drug development through significant reduction of the failure rate for pharmacokinetics related reasons. In contrast, the sciences of ADME and DMPK have not been fully integrated into the discovery and development processes for large molecule (LM) drugs. In this mini-review, we reflect on the journey of DMPK support of SM drug discovery and development and highlight the key enablers that have allowed DMPK scientists to make such impacts, with the aim to provide a perspective on relevant lessons learned from SM drugs that are applicable to DMPK support strategies for LMs.
Collapse
Affiliation(s)
- Thomayant Prueksaritanont
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Sharp and Dohme Corp., West Point, Pennsylvania 19486, USA.
| | | |
Collapse
|
137
|
Li S, Zhang D, Sun J, Li Z, Deng L, Zou B, Zhan J, Jiang W. Pharmacokinetics and tolerability of human mouse chimeric anti-CD22 monoclonal antibody in Chinese patients with CD22-positive non-Hodgkin lymphoma. MAbs 2012; 4:256-66. [PMID: 22453099 PMCID: PMC3361661 DOI: 10.4161/mabs.4.2.19136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 12/19/2011] [Accepted: 12/20/2011] [Indexed: 12/14/2022] Open
Abstract
The safety and pharmacokinetics assessment of antibodies targeting CD22 (e.g., epratuzumab) have been established in western Caucasian populations, but there are no reports of the effects in Chinese populations. This dose-escalation study examines the safety, pharmacokinetics and biologic effects of multiple doses of anti-CD22 human-murine chimeric monoclonal antibody SM03 in 21 Chinese patients with CD22-positive non-Hodgkin lymphoma. Most of drug-related adverse events (AEs) were mild and reversible. Two patients experienced serious AEs (hemorrhage); one patient had grade 4 neutropenia; one patient had asymptomatic grade III prolongation of activated partial thromboplastin time (APTT). Major AEs included fever (71%), prolongation of APTT (42.8%), leukocytopenia (44.4%), alanine transaminase elevation (28.6%), elevated serum creatinine (23.8%) and injection site skin redness (14.3%). Circulating B cells transiently decreased without significant effects on T cells or immunoglobulin levels. Pharmacokinetic data revealed that mean maximum observed SM03 concentration and mean AUC from time zero to infinity increased in a dose-dependent manner up to 360 mg/m (2) SM03. Mean clearance was similar at doses ≤ 360 mg/m (2) and decreased significantly at dose 480 mg/m (2), supporting saturation of B-cell binding at 360 mg/m (2). Across all dose levels and histologies, one patient achieved partial response at 480 mg/m (2) dose; 14 patients had stable disease as best response and four patients progressed. Overall, SM03 was tolerated at doses ranging from 60-480 mg/m (2) and had potential efficacy in Chinese patients with follicular lymphoma.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Neoplasm/administration & dosage
- Antibodies, Neoplasm/adverse effects
- Antibodies, Neoplasm/immunology
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacokinetics
- China
- Dose-Response Relationship, Drug
- Female
- Humans
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Lymphoma, Follicular/drug therapy
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/pathology
- Male
- Mice
- Middle Aged
- Sialic Acid Binding Ig-like Lectin 2
Collapse
Affiliation(s)
- Su Li
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Clinical Trial Center; Cancer Center; Sun Yat-sen University; Guangzhou, Guangdong China
| | - Dongsheng Zhang
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Medical Oncology; Cancer Center; Sun Yat-Sen University; Guangzhou, Guangdong China
| | - Jian Sun
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Clinical Trial Center; Cancer Center; Sun Yat-sen University; Guangzhou, Guangdong China
| | - Zhinming Li
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Medical Oncology; Cancer Center; Sun Yat-Sen University; Guangzhou, Guangdong China
| | - Liting Deng
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Clinical Trial Center; Cancer Center; Sun Yat-sen University; Guangzhou, Guangdong China
| | - Benyan Zou
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Medical Oncology; Cancer Center; Sun Yat-Sen University; Guangzhou, Guangdong China
| | - Jing Zhan
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Clinical Trial Center; Cancer Center; Sun Yat-sen University; Guangzhou, Guangdong China
| | - Wenqi Jiang
- State Key Laboratory of Oncology in Southern China; Sun Yat-Sen University; Guangzhou, Guangdong China
- Department of Medical Oncology; Cancer Center; Sun Yat-Sen University; Guangzhou, Guangdong China
| |
Collapse
|
138
|
Alvarez HM, So OY, Hsieh S, Shinsky-Bjorde N, Ma H, Song Y, Pang Y, Marian M, Escandón E. Effects of PEGylation and immune complex formation on the pharmacokinetics and biodistribution of recombinant interleukin 10 in mice. Drug Metab Dispos 2012; 40:360-73. [PMID: 22083830 DOI: 10.1124/dmd.111.042531] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Interleukin 10 (IL-10) is a potent cytokine homodimer with multiple immunoregulatory functions. Here, we have characterized the effects of PEGylation and formation of human IL-10 (hIL-10)/humanized anti-human IL-10 (hαhIL-10) immune complexes in the pharmacokinetics, biodistribution, and biotransformation of IL-10 in mice. To assess the fate of native, PEGylated, and antibody-bound IL-10; we implemented an analytical set of fluorescence emission-linked assays. Plasma size exclusion chromatography analysis indicated that fluoro-labeled native and PEGylated murine IL-10 (PEG-mIL-10) are stable in the circulation. PEGylation of IL-10 resulted in a 21-fold increased exposure, 2.7-fold increase in half-life, and 20-fold reduction in clearance. Kidney is the major organ of disposition for both native and PEGylated mIL-10 with renal uptake directly related to systemic clearance. The fluorescence signal in the kidneys reached tissue/blood ratios up to 150 and 20 for native and PEG-mIL-10, respectively. hIL-10/hαhIL-10 immune complexes are detectable in the circulation without evidence of unbound or degraded hIL-10. The exposure of hIL-10 present in immune complexes versus that of hIL-10 alone increased from 0.53 to 11.28 μg · day/ml, with a half-life of 1.16 days and a 23-fold reduction in clearance. Unlike hIL-10 alone, antibody-bound hIL-10 was targeted mainly to the liver with minimal renal distribution. In addition, we found an 11-fold reduction (from 9.9 to 113 nM) in binding to the neonatal Fc receptor (FcRn) when the hαhIL10 antibody is conjugated to hIL-10. The potential changes in FcRn binding in vivo and increased liver uptake may explain the unique pharmacokinetic properties of hIL-10/hαhIL-10 immune complexes.
Collapse
Affiliation(s)
- Hamsell M Alvarez
- Biologics Discovery DMPK and Bioanalytics Department, Merck Research Laboratories, Merck Sharp and Dohme Corp., Palo Alto, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Shah DK, Betts AM. Towards a platform PBPK model to characterize the plasma and tissue disposition of monoclonal antibodies in preclinical species and human. J Pharmacokinet Pharmacodyn 2011; 39:67-86. [PMID: 22143261 DOI: 10.1007/s10928-011-9232-2] [Citation(s) in RCA: 310] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 11/17/2011] [Indexed: 12/29/2022]
Abstract
The objectives of the following investigation were (1) development of a physiologically based pharmacokinetic (PBPK) model capable of characterizing the plasma and tissue pharmacokinetics (PK) of nonspecific or antigen specific monoclonal antibodies (mAbs) in wild type, FcRn knockout, tumor bearing and non tumor bearing mice and (2) evaluation of the scale up potential of the model by characterizing the mouse, rat, monkey and human plasma PK of mAbs, simultaneously. A PBPK model containing 15 tissues, a carcass and a tumor compartment was developed by modifying/augmenting previously published PBPK models. Each tissue compartment was subdivided into plasma, blood cell, endothelial, interstitial and cellular sub-compartments. Each tissue was connected through blood and lymph flow to the systemic circulation. Lymph flow was set to a value 500 times lower than plasma flow and vascular reflection coefficients for each tissue were adjusted according to their vascular pore size. In each tissue endothelial space, mAb entered via pinocytosis and the interaction of FcRn with mAb was described by on and off rates. FcRn bound mAb was recycled and unbound mAb was eliminated by a first order process (K(deg)). The PBPK model was simultaneously fit to the following datasets to estimate four system parameters: (1) plasma and tissue PK of nonspecific mAb in wild type mouse with or without simultaneous intravenous immunoglobulin (IVIG) administration, (2) plasma and tissue PK of nonspecific mAb in FcRn knockout mouse, (3) plasma and tissue PK of nonspecific mAb in tumor bearing mouse, (4) plasma and tissue PK of tumor antigen specific mAb in tumor bearing mouse, and (5) plasma PK of mAb in rat, monkey and human. The model was able to characterize all the datasets reasonably well with a common set of parameters. The estimated value of the four system parameters i.e. FcRn concentration (FcRn), rate of pinocytosis per unit endosomal space (CL(up)), K(deg) and the proportionality constant (C_LNLF) between the rate at which antibody transfers from the lymph node compartment to the blood compartment and the plasma flow of the given species, were found to be 4.98E-05 M (CV% = 11.1), 3.66E-02 l/h/l (%CV = 3.48), 42.9 1/h (%CV = 15.7) and 9.1 (CV% > 50). Thus, a platform PBPK model has been developed that can not only simultaneously characterize mAb disposition data obtained from various previously published mouse PBPK models but is also capable of characterizing mAb disposition in various preclinical species and human.
Collapse
Affiliation(s)
- Dhaval K Shah
- Translational Research Group, Department of Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development Groton Labs, MS 8220-4573, Eastern Point Road, Groton, CT 06340, USA.
| | | |
Collapse
|
140
|
Magistrelli G, Malinge P, Anceriz N, Desmurs M, Venet S, Calloud S, Daubeuf B, Kosco-Vilbois M, Fischer N. Robust recombinant FcRn production in mammalian cells enabling oriented immobilization for IgG binding studies. J Immunol Methods 2011; 375:20-9. [PMID: 21939661 DOI: 10.1016/j.jim.2011.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 09/01/2011] [Accepted: 09/01/2011] [Indexed: 12/28/2022]
Abstract
The MHC class-I related receptor or neonatal Fc receptor (FcRn) protects IgG and albumin from degradation by rescuing them in endothelial cells in a pH dependent fashion and consequently increases their respective half-lives. Monoclonal antibody-based therapies are of increasing interest and characterizing the interaction with FcRn is important for the development of an antibody candidate. In order to facilitate the production of soluble FcRn suitable for interaction studies, we generated semi-stable pools co-expressing FcRn α-chain, β2-microglobulin, biotin ligase and EGFP using a dual promoter, multi-cistronic vector. Human and mouse FcRn were purified in the mg/L range of culture medium and a single purification step was sufficient to reach a high level of purity. The receptors were characterized by ELISA, flow cytometry and surface plasmon resonance and shown to be functional. The single site biotinylation facilitated the directional immobilization of FcRn on the sensor chip and significantly increased the response level of the surface compared to amine coupling used in previous studies. Using this system, the affinity constants of seven IgGs, from various species and isotypes, were determined for human and mouse FcRn, including two hamster isotypes. These results confirm the higher selectivity of the human receptor and the promiscuous binding of mFcRn to IgGs from different species.
Collapse
|
141
|
Abstract
The majority of potent new biologics today are IgG-based molecules that have demonstrated tissue-targeting specificity with favorable clinical response. Several factors determine the efficacy of these products, including target specificity, serum half-life and effector functions via complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity or drug conjugates. In this review, we will focus on the interaction between therapeutic antibody and neonatal Fc receptor (FcRn), which is one of the critical factors in determining the circulating antibody half-life. Specifically, we will review the fundamental biology of FcRn, FcRn functions in various organs, Fc mutations designed to modulate binding to FcRn, IgG-based therapeutics that directly exploit FcRn functions and tools and strategies used to study FcRn-IgG interactions. Comprehensive understanding of FcRn-IgG interactions not only allows for development of effective therapeutics, but also avoidance of potential adverse effects.
Collapse
Affiliation(s)
- Timothy T Kuo
- Division of Gastroenterology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|