1
|
Miller MJ, Kroenke MA, Barger T, Manning MS, Sohn W, Graham K, Mytych DT, Gupta S. Inhibition of RANKL is critical for accurate assessment of anti-drug antibody incidence to denosumab in clinical studies. J Immunol Methods 2025; 540:113864. [PMID: 40220951 DOI: 10.1016/j.jim.2025.113864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/20/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Denosumab is an approved monoclonal antibody therapeutic for the treatment of bone loss in the postmenopausal osteoporosis and oncology settings and acts by binding and neutralizing the activity of receptor activator of nuclear factor-kappa-B ligand (RANKL). Anti-drug antibodies (ADAs) to denosumab are measured via a standard electrochemiluminescence- (ECL) based bridging assay. In this format, the presence of soluble RANKL (sRANKL) in clinical samples can lead to false positive results. In a denosumab bioequivalence study, approximately 50 % of the serum samples showed reactivity to denosumab in the absence of a specific reagent to sequester the sRANKL. However, upon addition of osteoprotegerin (OPG) as a reagent to neutralize the sRANKL, the overall ADA incidence was lowered to <1 %. To address this RANKL reactivity over the long-term use of this assay, the performance of 3 RANKL inhibitors was evaluated using healthy donor sera samples spiked with different concentrations of positive control ADA, sRANKL, or both, in an ECL based immunoassay utilizing the Meso Scale Discovery (MSD) platform. Based on these data, the denosumab antibody assay was modified to include a neutralizing anti-RANKL monoclonal antibody to eliminate the assay reactivity or false positivity due to sRANKL. Use of an anti-RANKL antibody did not impact the ADA-specific signal but inhibited the false positive assay signal due to sRANKL, resulting in an accurate detection of ADA incidence. Therefore, inhibition of interference posed by sRANKL in study samples is critical for the accurate assessment of ADA incidence towards denosumab and any biosimilars for this product that are undergoing clinical development.
Collapse
Affiliation(s)
- Mieke Jill Miller
- Translational Safety and Bioanalytical Sciences, Amgen, Thousand Oaks, CA 91320, United States of America.
| | - Mark A Kroenke
- Clinical Immunology, Amgen, Thousand Oaks, CA 91320, United States of America
| | - Troy Barger
- Translational Safety and Bioanalytical Sciences, Amgen, Thousand Oaks, CA 91320, United States of America
| | - Marta Starcevic Manning
- Translational Safety and Bioanalytical Sciences, Amgen, Thousand Oaks, CA 91320, United States of America
| | - Winnie Sohn
- Clinical Pharmacology Modeling and Simulation, Amgen, Thousand Oaks, CA 91320, United States of America
| | - Kevin Graham
- Large Molecule Discovery & Research Data Science, Amgen, Thousand Oaks, CA 91320, United States of America
| | - Daniel T Mytych
- Clinical Immunology, Amgen, Thousand Oaks, CA 91320, United States of America
| | - Shalini Gupta
- Translational Safety and Bioanalytical Sciences, Amgen, Thousand Oaks, CA 91320, United States of America
| |
Collapse
|
2
|
Bhavaraju K, Dhiman MK, Desai H, Brien KO, Gadgil SS, Mohapatra S, Kumar V. Mitigating target interference challenges in bridging immunogenicity assay to detect anti-tocilizumab antibodies. Bioanalysis 2024; 16:587-602. [PMID: 39010827 PMCID: PMC11352699 DOI: 10.1080/17576180.2024.2349417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/26/2024] [Indexed: 07/17/2024] Open
Abstract
Aim: An assay to detect anti-tocilizumab antibodies in the presence of high levels of circulating target and drug is needed for immunogenicity assessment in comparative clinical studies.Methods: An assay was developed and validated using a combination of blocking agents and dilutions to overcome target interference challenges.Results: No false-positive signal was detected in serum samples spiked with 350-500 ng/ml of IL-6 receptor. As low as 50 ng/ml of positive control antibodies could be detected in the presence of either 500 ng/ml of IL-6 or 250 μg/ml of the drug product. Assay also demonstrated high sensitivity, selectivity and precision.Conclusion: A robust, easy to perform immunogenicity assay was developed and validated for detecting anti-tocilizumab antibodies.
Collapse
Affiliation(s)
- Kamala Bhavaraju
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Mamta Kumari Dhiman
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Hema Desai
- Clinical Pharmacology and Bioanalysis, Syneos Health, Princeton, NJ08540, USA
| | - Kyla O’ Brien
- Clinical Pharmacology and Bioanalysis, Syneos Health, Princeton, NJ08540, USA
| | - Sagarika Sunil Gadgil
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Soumyaranjan Mohapatra
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Vikas Kumar
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| |
Collapse
|
3
|
Deng X, Hou Y, Yuan W, Yang H, Guo R, Liu T, Liu Y, Xu J, Liu H, Gong L, Qin Q. Eliminating drug target interference with specific antibody or its F(ab') 2 fragment in the bridging immunogenicity assay. Bioanalysis 2024; 16:135-148. [PMID: 38385901 PMCID: PMC11845105 DOI: 10.4155/bio-2023-0191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
Background: DB-1003 is a humanized anti-IgE monoclonal antibody with higher affinity than omalizumab. In the affinity capture elution (ACE)-based bridging electrochemiluminescent immunoassay (ECLIA) for antibodies to DB-1003, monkey serum IgE caused false-positive results. Materials & methods: The target-specific antibody or its F(ab')2 fragment was used to mitigate drug target interference in an ACE-based bridging ECLIA for the detection of anti-DB-1003 antibodies. Results: The sensitivity of the developed assay was at least 100 ng/ml. When the anti-drug antibody concentration was 250 ng/ml, the assay tolerated at least 20.0 μg/ml of the monkey IgE. Conclusion: Incorporating the target-specific antibody or its F(ab')2 fragment can overcome the interference from monkey serum IgE in ACE-based bridging ECLIA for anti-DB-1003 antibody detection.
Collapse
Affiliation(s)
- Xiaojie Deng
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingying Hou
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenyi Yuan
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hongzhou Yang
- Longbio Pharma (Suzhou) Co., Ltd, Suzhou, 215558, China
| | - Ruowen Guo
- Longbio Pharma (Suzhou) Co., Ltd, Suzhou, 215558, China
| | - Tingting Liu
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yongzhen Liu
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Junjiu Xu
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Heng Liu
- Longbio Pharma (Suzhou) Co., Ltd, Suzhou, 215558, China
| | - Likun Gong
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiuping Qin
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
4
|
Wang X, Chang W, Khosraviani M, Phung W, Peng L, Cohen S, Andrews BT, Sun Y, Davies CW, Koerber JT, Yang J, Song A. Application of N-Terminal Site-Specific Biotin and Digoxigenin Conjugates to Clinical Anti-drug Antibody Assay Development. Bioconjug Chem 2024; 35:174-186. [PMID: 38050929 DOI: 10.1021/acs.bioconjchem.3c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Biotin- and digoxigenin (DIG)-conjugated therapeutic drugs are critical reagents used for the development of anti-drug antibody (ADA) assays for the assessment of immunogenicity. The current practice of generating biotin and DIG conjugates is to label a therapeutic antibody with biotin or DIG via primary amine groups on lysine or N-terminal residues. This approach modifies lysine residues nonselectively, which can impact the ability of an ADA assay to detect those ADAs that recognize epitopes located at or near the modified lysine residue(s). The impact of the lysine modification is considered greater for therapeutic antibodies that have a limited number of lysine residues, such as the variable heavy domain of heavy chain (VHH) antibodies. In this paper, for the first time, we report the application of site-specifically conjugated biotin- and DIG-VHH reagents to clinical ADA assay development using a model molecule, VHHA. The site-specific conjugation of biotin or DIG to VHHA was achieved by using an optimized reductive alkylation approach, which enabled the majority of VHHA molecules labeled with biotin or DIG at the desirable N-terminus, thereby minimizing modification of the protein after labeling and reducing the possibility of missing detection of ADAs. Head-to-head comparison of biophysical characterization data revealed that the site-specific biotin and DIG conjugates demonstrated overall superior quality to biotin- and DIG-VHHA prepared using the conventional amine coupling method, and the performance of the ADA assay developed using site-specific biotin and DIG conjugates met all acceptance criteria. The approach described here can be applied to the production of other therapeutic-protein- or antibody-based critical reagents that are used to support ligand binding assays.
Collapse
Affiliation(s)
- Xiangdan Wang
- BioAnalytical Sciences, Genentech, South San Francisco, California 94080, United States
| | - Wenping Chang
- Department of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| | - Mehraban Khosraviani
- BioAnalytical Sciences, Genentech, South San Francisco, California 94080, United States
| | - Wilson Phung
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, South San Francisco, California 94080, United States
| | - Lingling Peng
- Department of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| | - Sivan Cohen
- BioAnalytical Sciences, Genentech, South San Francisco, California 94080, United States
| | - Benjamin T Andrews
- BioAnalytical Sciences, Genentech, South San Francisco, California 94080, United States
| | - Yonglian Sun
- Antibody Engineering, Genentech, South San Francisco, California 94080, United States
| | - Christopher W Davies
- Antibody Engineering, Genentech, South San Francisco, California 94080, United States
| | - James T Koerber
- Antibody Engineering, Genentech, South San Francisco, California 94080, United States
| | - Jihong Yang
- BioAnalytical Sciences, Genentech, South San Francisco, California 94080, United States
| | - Aimin Song
- Department of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| |
Collapse
|
5
|
Li S, Murakami D, Nagatoishi S, Liu Y, Tsumoto K, Katayama Y, Mori T. One-pot preparation of mannan-coated antigen nanoparticles using human serum albumin as a matrix for tolerance induction. J Colloid Interface Sci 2023; 649:955-965. [PMID: 37392685 DOI: 10.1016/j.jcis.2023.06.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/10/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Nanoparticles (NPs) for allergen immunotherapy have garnered attention for their high efficiency and safety compared with naked antigen proteins. In this work, we present mannan-coated protein NPs, incorporating antigen proteins for antigen-specific tolerance induction. The heat-induced formation of protein NPs is a one-pot preparation method and can be applied to various proteins. Here, the NPs were formed spontaneously via heat denaturation of three component proteins: an antigen protein, human serum albumin (HSA) as a matrix protein, and mannoprotein (MAN) as a targeting ligand for dendritic cells (DCs). HSA is non-immunogenic, therefore suitable as a matrix protein, while MAN coats the surface of the NP. We applied this method to various antigen proteins and found that the self-disperse after heat denaturation was a requirement for incorporation into the NPs. We also established that the NPs could target DCs, and the incorporation of rapamycin into the NPs enhanced the induction of a tolerogenic phenotype of DC. The MAN coating provided steric hindrance and heat denaturation destroyed recognition structures, successfully preventing anti-antigen antibody binding, indicating the NPs may avoid anaphylaxis induction. The MAN-coated NPs proposed here, prepared by a simple method, have the potential for effective and safe allergies treatment for various antigens.
Collapse
Affiliation(s)
- Shunyi Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Daisuke Murakami
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yiwei Liu
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | - Kouhei Tsumoto
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, Fukuoka 819-0395, Japan; Centre for Advanced Medicine Innovation, Kyushu University, Fukuoka 812-8582, Japan; Department of Biomedical Engineering, Chung Yuan Christian University, Chung Li, 32023, Taiwan, ROC.
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 819-0395, Japan.
| |
Collapse
|
6
|
89Zr-labelled Obinutuzumab: a potential immuno-PET radiopharmaceutical. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08614-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
7
|
Myler H, Pedras-Vasconcelos J, Phillips K, Hottenstein CS, Chamberlain P, Devanaryan V, Gleason C, Goodman J, Manning MS, Purushothama S, Richards S, Shen H, Zoghbi J, Amaravadi L, Barger T, Bowen S, Bowsher RR, Clements-Egan A, Geng D, Goletz TJ, Gunn GR, Hallett W, Hodsdon ME, Janelsins BM, Jawa V, Kamondi S, Kirshner S, Kramer D, Liang M, Lindley K, Liu S, Liu Z, McNally J, Mikulskis A, Nelson R, Ahbari MR, Qu Q, Ruppel J, Snoeck V, Song A, Yan H, Ware M. Anti-drug Antibody Validation Testing and Reporting Harmonization. AAPS J 2021; 24:4. [PMID: 34853961 PMCID: PMC8816448 DOI: 10.1208/s12248-021-00649-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/20/2021] [Indexed: 11/30/2022] Open
Abstract
Evolving immunogenicity assay performance expectations and a lack of harmonized anti-drug antibody validation testing and reporting tools have resulted in significant time spent by health authorities and sponsors on resolving filing queries. Following debate at the American Association of Pharmaceutical Sciences National Biotechnology Conference, a group was formed to address these gaps. Over the last 3 years, 44 members from 29 organizations (including 5 members from Europe and 10 members from FDA) discussed gaps in understanding immunogenicity assay requirements and have developed harmonization tools for use by industry scientists to facilitate filings to health authorities. Herein, this team provides testing and reporting strategies and tools for the following assessments: (1) pre-study validation cut point; (2) in-study cut points, including procedures for applying cut points to mixed populations; (3) system suitability control criteria for in-study plate acceptance; (4) assay sensitivity, including the selection of an appropriate low positive control; (5) specificity, including drug and target tolerance; (6) sample stability that reflects sample storage and handling conditions; (7) assay selectivity to matrix components, including hemolytic, lipemic, and disease state matrices; (8) domain specificity for multi-domain therapeutics; (9) and minimum required dilution and extraction-based sample processing for titer reporting.
Collapse
Affiliation(s)
- Heather Myler
- Immunochemistry Department, PPD Laboratories, 2244 Dabney Road, Richmond, Virginia, 23230-3323, USA.
| | - João Pedras-Vasconcelos
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Kelli Phillips
- Immunochemistry Department, PPD Laboratories, 2244 Dabney Road, Richmond, Virginia, 23230-3323, USA
| | - Charles Scott Hottenstein
- Immunogenicity, GlaxoSmithKline Pharmaceuticals, 1250 South Collegeville Road, Collegeville, Pennsylvania, 19426, USA
| | - Paul Chamberlain
- NDA Advisory Services, Ltd., Grove House, Guildford Road, Leatherhead, KT22 9DF, Surrey, UK
| | | | - Carol Gleason
- Global Biometric and Data Sciences, Bristol-Myers Squibb, Princeton, New Jersey, 08540, USA
| | - Joanne Goodman
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Shobha Purushothama
- Diagnostics Accelerator, Alzheimer's Drug Discovery Foundation, 57W 57th Street, New York, New York, USA
| | - Susan Richards
- Translational Medicine and Early Development, Sanofi, Framingham, Massachusetts, 01701, USA
| | - Honglue Shen
- Specialty Bioanalytics, Teva Pharmaceuticals, West Chester, Pennsylvania, 19380, USA
| | - Jad Zoghbi
- Translational Medicine and Early Development, Sanofi, Framingham, Massachusetts, 01701, USA
| | | | - Troy Barger
- Bioanalytical Sciences, Amgen Research, Thousand Oaks, California, 91320, USA
| | - Steven Bowen
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Ronald R Bowsher
- B2S Life Sciences, 97 East Monroe Street, Franklin, Indiana, 46131, USA
| | | | - Dong Geng
- Legend Biotech, 10 Knightsbridge Road, Piscataway, New Jersey, 08554, USA
| | - Theresa J Goletz
- Drug Metabolism & Pharmacokinetics, EMD Serono, Billerica, Massachusetts, 01821, USA
| | - George R Gunn
- Immunogenicity, GlaxoSmithKline Pharmaceuticals, 1250 South Collegeville Road, Collegeville, Pennsylvania, 19426, USA
| | - William Hallett
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Michael E Hodsdon
- Laboratory for Experimental Medicine, Eli Lilly and Company, Indianapolis, Indiana, 46285, USA
| | - Brian M Janelsins
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Vibha Jawa
- Predictive and Clinical Immunogenicity Pharmacometrics, Pharmacodynamics and Drug Metabolism, Merck and Co., 2000 Galloping Hill Road, Kenilworth, New Jersey, 07033, USA
| | - Szilard Kamondi
- Kamondi Bioanalytical Consultancy, Rheinfelden, Switzerland / Roche Pharma Research & Early Development, Pharmaceutical Sciences, Bioanalytical R&D, Roche Innovation Center, Basel, Switzerland
| | - Susan Kirshner
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Daniel Kramer
- Translational Medicine and Early Development, Sanofi, Frankfurt am Main, Germany
| | - Meina Liang
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, South San Francisco, California, USA
| | | | - Susana Liu
- Pfizer Inc., 17300 Trans Canada Hwy, Kirkland, Quebec, Canada
| | - ZhenZhen Liu
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Jim McNally
- BioAgilytix Labs, Durham, North Carolina, 27713, USA
| | - Alvydas Mikulskis
- Clinical Biomarkers, Vertex Pharmaceuticals, Inc., Boston, Massachusetts, 02210, USA
| | - Robert Nelson
- Immunochemistry Department, Covance Laboratories Ltd., Harrogate, HG3 1PY, UK
| | - Mohsen Rajabi Ahbari
- Office of Study Integrity and Surveillance, Office of Translational Sciences, Center for Drug Evaluation and Research (CDER), Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Qiang Qu
- Global Product Development, Pfizer Inc., Andover, Massachusetts, 01810, USA
| | - Jane Ruppel
- BioAnalytical Sciences, Genentech, South San Francisco, California, USA
| | - Veerle Snoeck
- Translational Biomarkers and Bioanalysis, UCB Biopharma SRL, B-1420, Braine-l'Alleud, Belgium
| | - An Song
- Development Sciences, Immune-Onc Therapeutics, Palo Alto, California, 94303, USA
| | - Haoheng Yan
- Product Quality and Immunogenicity, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Mark Ware
- Janssen BioTherapeutics, Janssen R&D LLC, Spring House, Pennsylvania, 19477, USA
| |
Collapse
|
8
|
Effect of Mesenchymal Stem Cell-Derived Exosomes on Retinal Injury: A Review of Current Findings. Stem Cells Int 2020; 2020:8883616. [PMID: 33082789 PMCID: PMC7556062 DOI: 10.1155/2020/8883616] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 02/08/2023] Open
Abstract
In recent years, various studies have followed in the literature on the therapeutic effects of mesenchymal stem cells (MSC) on damage in retinal cells. The evidence that MSCs exert their regenerative and damage reduction effect in a paracrine way, through the release of soluble factors and exosomes, is now consolidated. Exosomes are microvesicles formed by a double layer of phospholipid membrane and carry proteins and RNA, through which they play a therapeutic role on target cells. Scientific research has recently focused on the use of exosomes derived from MSC in various models of retinal damage in vitro and in vivo as they, compared to MSCs, have similar functions and at the same time have different advantages such as greater stability and handling, a lower chance of immunological rejection and no risk of malignant transformation. The purpose of this review is to summarize current knowledge on the therapeutic use of exosomes derived from MSCs in retinal damage and to stimulate new clinical perspectives regarding their use.
Collapse
|
9
|
Mitigating target interference in bridging immunogenicity assay with target-blocking reagents and mild basic pH. Bioanalysis 2019; 11:1569-1580. [DOI: 10.4155/bio-2018-0187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background: Soluble drug target in clinical study samples generated false positive results in anti-drug antibody (ADA) bridging assays due to target-mediated bridging. Results: The combination of two target-blocking reagents and mild basic assay pH resulted in high tolerance to recombinant target protein and reduced levels of positivity in clinical study samples with pharmacokinetic profiles that did not indicate significant ADA response. Testing with low-affinity ADA positive serum from immunized rabbits and known ADA positive samples from nonclinical studies in rats confirmed the assay's ability to detect ADA positive samples and the minimal impact of basic pH and target-blocking reagents on ADA detection. Conclusion: These strategies provide alternatives for mitigating target interference when standard target-blocking antibodies alone are ineffective.
Collapse
|
10
|
Perspectives on exploring hybrid LBA/LC-MS approach for clinical immunogenicity testing. Bioanalysis 2019; 11:1605-1617. [PMID: 31208198 DOI: 10.4155/bio-2018-0107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biological drug products may elicit an antidrug antibody (ADA) response. The current widely used bridging ligand binding assay (LBA) is the gold standard for ADA assessments in drug development, which is a qualitative assay followed by a quasi-quantitative titer analysis but can be prone to interferences from biological matrices, drug targets and circulating drugs. We present our perspectives and findings in exploring a hybrid LBA/LC-MS as an orthogonal bioanalytical tool for clinical immunogenicity assessments. The hybrid LBA/LC-MS is a semiquantitative assay with acceptable specificity, drug tolerance and the capability of multiplexed detection of ADA isotypes. The assay results suggest this technology to be a promising and complementary bioanalytical tool that can provide informative immunogenicity data in drug development.
Collapse
|
11
|
Suh CH, Yoo DH, Berrocal Kasay A, Chalouhi El-Khouri E, Cons Molina FF, Shesternya P, Miranda P, Medina-Rodriguez FG, Wiland P, Jeka S, Chavez-Corrales J, Linde T, Hrycaj P, Abello-Banfi M, Hospodarskyy I, Jaworski J, Piotrowski M, Brzosko M, Krogulec M, Shevchuk S, Calvo A, Andersone D, Park W, Shim SC, Lee SJ, Lee SY. Long-Term Efficacy and Safety of Biosimilar CT-P10 Versus Innovator Rituximab in Rheumatoid Arthritis: 48-Week Results from a Randomized Phase III Trial. BioDrugs 2019; 33:79-91. [PMID: 30719632 PMCID: PMC6373391 DOI: 10.1007/s40259-018-00331-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The aim of this study was to investigate long-term clinical outcomes of extended treatment with CT-P10, a rituximab biosimilar, compared with rituximab reference products sourced from the USA and the EU (US-RTX and EU-RTX) in rheumatoid arthritis (RA) for up to 48 weeks. METHODS In this multinational, randomized, double-blind trial, adults with active RA received up to two courses of CT-P10, US-RTX, or EU-RTX alongside methotrexate. Efficacy endpoints included Disease Activity Score 28-joint count (DAS28) and American College of Rheumatology (ACR) response rates. Pharmacokinetics, pharmacodynamics, immunogenicity, and safety were also assessed. RESULTS Of 372 patients randomized to the study drug, 330 (88.7%) completed the second treatment course. Mean change from baseline to week 48 in DAS28-C-reactive protein was comparable in the CT-P10 and combined rituximab (US-RTX and EU-RTX) groups (- 2.7 and - 2.6, respectively). ACR20, ACR50, and ACR70 response rates at week 48 indicated no differences between groups (80.6%, 55.4%, and 31.7% vs. 79.8%, 53.9%, and 33.7% in the CT-P10 and combined rituximab groups, respectively). Similar improvements in the Health Assessment Questionnaire Disability Index and all medical outcomes in the Short Form 36-Item Health Survey, including physical and mental health, were seen in all groups. At week 48, antidrug antibodies were detected in 4.9%, 9.4%, and 8.6% of patients in the CT-P10, US-RTX, and EU-RTX groups, respectively. CT-P10 and rituximab displayed similar pharmacokinetic, pharmacodynamic, and safety profiles. CONCLUSION CT-P10 was similar to EU-RTX and US-RTX in terms of efficacy, pharmacokinetics, pharmacodynamics, immunogenicity, and safety up to week 48. CLINICALTRIALS. GOV IDENTIFIER NCT02149121.
Collapse
Affiliation(s)
- Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dae Hyun Yoo
- Division of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, Republic of Korea.
| | | | | | | | | | | | | | | | - Slawomir Jeka
- Department of Rheumatology and Connective Tissue Diseases, University Hospital No. 2, Collegium Medicum UMK, Bydgoszcz, Poland
| | | | - Thomas Linde
- MVZ für Rheumatologie und Autoimmundiagnostik, Halle (Salle), Germany
| | - Pawel Hrycaj
- Department of Rheumatology, Koscian Municipal Hospital, Koscian, Poland
| | | | | | | | - Mariusz Piotrowski
- Department of Rheumatology, Medical University of Lublin, Lublin, Poland
| | - Marek Brzosko
- Department of Rheumatology, Internal Diseases and Geriatrics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Sergii Shevchuk
- National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| | - Armando Calvo
- Centro de Investigación Clínica Inunoreumatología, Clínica San Felipe, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Won Park
- School of Medicine, Medicine/Rheumatology, IN-HA University, Incheon, Republic of Korea
| | - Seung Cheol Shim
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | | | | |
Collapse
|
12
|
Park W, Božić-Majstorović L, Milakovic D, Berrocal Kasay A, El-Khouri EC, Irazoque-Palazuelos F, Molina FFC, Shesternya P, Miranda P, Medina-Rodriguez FG, Wiland P, Jeka S, Chavez-Corrales J, Garmish O, Linde T, Rekalov D, Hrycaj P, Krause A, Fomina N, Piura O, Abello-Banfi M, Suh CH, Shim SC, Lee SJ, Lee SY, Kim SH, Yoo DH. Comparison of biosimilar CT-P10 and innovator rituximab in patients with rheumatoid arthritis: a randomized controlled Phase 3 trial. MAbs 2018; 10:934-943. [PMID: 30010481 PMCID: PMC6152436 DOI: 10.1080/19420862.2018.1487912] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This multinational, randomized, double-blind trial, (ClinicalTrials.gov identifier NCT02149121) was designed to demonstrate equivalence in pharmacokinetics and efficacy between CT-P10 and innovator rituximab (RTX) in patients with rheumatoid arthritis (RA). Adults with active RA were treated with CT-P10, United States-sourced RTX (US-RTX; Rituxan®), or European Union-sourced RTX (EU-RTX; MabThera®) at weeks 0 and 2. The co-primary pharmacokinetic endpoints were area under the serum concentration–time curve (AUC) from time zero to last measurable concentration (AUC0–last), AUC from time zero to infinity (AUC0–∞), and maximum concentration (Cmax) after two infusions. The primary efficacy endpoint was change from baseline to week 24 in Disease Activity Score using 28 joints-C-reactive protein (DAS28-CRP). Pharmacodynamics, immunogenicity, and safety were also assessed. 372 patients were randomly assigned to CT-P10 (n = 161) or RTX (n = 211 [US-RTX, n = 151; EU-RTX, n = 60]). For the co-primary pharmacokinetic endpoints, 90% confidence intervals (CI) for ratios of geometric means (CT-P10/US-RTX, CT-P10/EU-RTX or EU-RTX/US-RTX) all fell within the equivalence margin of 80–125%. Adjusted least squares (LS) mean (standard error) change from baseline in DAS28-CRP at week 24 was −2.13 (0.175) for CT-P10 and −2.09 (0.176) for RTX. The 95% CI (−0.29, 0.21) of the estimated treatment difference between CT-P10 and RTX (−0.04) was entirely within the efficacy equivalence margin of ±0.5. Pharmacodynamics, immunogenicity, and safety profiles were similar for CT-P10 and RTX. The pharmacokinetics of CT-P10, US-RTX, and EU-RTX were equivalent. CT-P10 and RTX were also equivalent in terms of efficacy and displayed similar pharmacodynamic, immunogenicity, and safety profiles up to week 24.
Collapse
Affiliation(s)
- Won Park
- a Medicine/Rheumatology , IN-HA University, School of Medicine , Incheon , Republic of Korea
| | | | - Dragana Milakovic
- b University Clinical Centre of the Republic of Srpska , Banja Luka , Bosnia and Herzegovina
| | | | | | | | | | | | - Pedro Miranda
- h Centro de Estudios Reumatológicos , Santiago , Chile
| | | | - Piotr Wiland
- j Medical University of Wroclaw , Wroclaw , Poland
| | - Slawomir Jeka
- k Department of Rheumatology and Connective Tissue Diseases , University Hospital No. 2, Collegium Medicum UMK , Bydgoszcz , Poland
| | | | - Olena Garmish
- m Institute of Cardiology named by M.D. Strazhesko NAMS of Ukraine , Kyiv , Ukraine
| | - Thomas Linde
- n MVZ für Rheumatologie und Autoimmundiagnostik, Halle (Salle) , Germany
| | - Dmytro Rekalov
- o Department of Internal Diseases , Zaporizhzhia State Medical University , Zaporizhzhia , Ukraine
| | - Pawel Hrycaj
- p Department of Rheumatology and Clinical Immunology , Poznań University of Medical Sciences , Poznań , Poland
| | - Andreas Krause
- q Medical Centre for Rheumatology Berlin-Buch, Immanuel Krankenhaus Berlin , Berlin , Germany
| | - Natalia Fomina
- r Kemerovo Regional Clinical Hospital , Kemerovo , Russian Federation
| | - Olena Piura
- s Department of Rheumatology , Kyiv Regional Clinical Hospital , Kyiv , Ukraine
| | | | - Chang-Hee Suh
- u Department of Rheumatology , Ajou University School of Medicine , Suwon , Republic of Korea
| | - Seung Cheol Shim
- v Department of Internal Medicine , Chungnam National University Hospital , Daejeon , Republic of Korea
| | | | | | | | - Dae Hyun Yoo
- x Division of Rheumatology , Hanyang University Hospital for Rheumatic Diseases , Seoul , Republic of Korea
| |
Collapse
|
13
|
Chang YH, Wu KC, Harn HJ, Lin SZ, Ding DC. Exosomes and Stem Cells in Degenerative Disease Diagnosis and Therapy. Cell Transplant 2018; 27:349-363. [PMID: 29692195 PMCID: PMC6038041 DOI: 10.1177/0963689717723636] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stroke can cause death and disability, resulting in a huge burden on society. Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterized by motor dysfunction. Osteoarthritis (OA) is a progressive degenerative joint disease characterized by cartilage destruction and osteophyte formation in the joints. Stem cell therapy may provide a biological treatment alternative to traditional pharmacological therapy. Mesenchymal stem cells (MSCs) are preferred because of their differentiation ability and possible derivation from many adult tissues. In addition, the paracrine effects of MSCs play crucial anti-inflammatory and immunosuppressive roles in immune cells. Extracellular vesicles (EVs) are vital mediators of cell-to-cell communication. Exosomes contain various molecules such as microRNA (miRNA), which mediates biological functions through gene regulation. Therefore, exosomes carrying miRNA or other molecules can enhance the therapeutic effects of MSC transplantation. MSC-derived exosomes have been investigated in various animal models representing stroke, PD, and OA. Exosomes are a subtype of EVs. This review article focuses on the mechanism and therapeutic potential of MSC-derived exosomes in stroke, PD, and OA in basic and clinical aspects.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- 1 Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.,2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Kung-Chi Wu
- 3 Department of Orthopedics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Horng-Jyh Harn
- 4 Department of Pathology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shinn-Zong Lin
- 5 Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Dah-Ching Ding
- 2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,6 Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
14
|
Xue L, Clements-Egan A, Amaravadi L, Birchler M, Gorovits B, Liang M, Myler H, Purushothama S, Manning MS, Sung C. Recommendations for the Assessment and Management of Pre-existing Drug-Reactive Antibodies During Biotherapeutic Development. AAPS JOURNAL 2017; 19:1576-1586. [DOI: 10.1208/s12248-017-0153-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/31/2017] [Indexed: 12/16/2022]
|
15
|
|
16
|
Zhong ZD, Clements-Egan A, Gorovits B, Maia M, Sumner G, Theobald V, Wu Y, Rajadhyaksha M. Drug Target Interference in Immunogenicity Assays: Recommendations and Mitigation Strategies. AAPS JOURNAL 2017; 19:1564-1575. [DOI: 10.1208/s12248-017-0148-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/11/2017] [Indexed: 12/13/2022]
|
17
|
Bridging immunogenicity assays for IgG4 therapeutics: mitigating interference from Fc-Fc interactions. Bioanalysis 2017; 9:707-717. [PMID: 28488898 DOI: 10.4155/bio-2017-0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM A bridging immunogenicity assay for a human IgG4 mAb therapeutic was transferred to an automation system to increase throughput. However, background signal increased five- to six-fold during the 6- to 8-h run. RESULTS Noncovalent Fc contacts formed between labeled IgG4 drugs in reagent solutions stored during the automation run. This generated substantial background signal, reducing assay sensitivity by approximately sixfold. Fc interactions also significantly impacted the confirmation assay. Fc contacts formed between labeled and unlabeled drug, significantly increasing signal inhibition (∼7-70%) in the 6-h run. CONCLUSION Storing labeled antibody solutions separately and combining them immediately before adding to samples reduced interference from Fc interactions. Preincubation time for reagent solutions should be strictly controlled for anti-drug antibody assays with IgG4 drugs to avoid false-positive results.
Collapse
|
18
|
Yoo DH, Suh CH, Shim SC, Jeka S, Cons-Molina FF, Hrycaj P, Wiland P, Lee EY, Medina-Rodriguez FG, Shesternya P, Radominski S, Stanislav M, Kovalenko V, Sheen DH, Myasoutova L, Lim MJ, Choe JY, Lee SJ, Lee SY, Kwon TS, Park W. A multicentre randomised controlled trial to compare the pharmacokinetics, efficacy and safety of CT-P10 and innovator rituximab in patients with rheumatoid arthritis. Ann Rheum Dis 2017; 76:566-570. [PMID: 27624791 PMCID: PMC5446025 DOI: 10.1136/annrheumdis-2016-209540] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 08/05/2016] [Accepted: 08/09/2016] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To demonstrate pharmacokinetic equivalence of CT-P10 and innovator rituximab (RTX) in patients with rheumatoid arthritis (RA) with inadequate responses or intolerances to antitumour necrosis factor agents. METHODS In this randomised phase I trial, patients with active RA were randomly assigned (2:1) to receive 1000 mg CT-P10 or RTX at weeks 0 and 2 (alongside continued methotrexate therapy). Primary endpoints were area under the serum concentration-time curve from time zero to last quantifiable concentration (AUC0-last) and maximum serum concentration after second infusion (Cmax). Additional pharmacokinetic parameters, efficacy, pharmacodynamics, immunogenicity and safety were also assessed. Data are reported up to week 24. RESULTS 103 patients were assigned to CT-P10 and 51 to RTX. The 90% CIs for the ratio of geometric means (CT-P10/RTX) for both primary endpoints were within the bioequivalence range of 80%-125% (AUC0-last: 97.7% (90% CI 89.2% to 107.0%); Cmax: 97.6% (90% CI 92.0% to 103.5%)). Pharmacodynamics and efficacy were comparable between groups. Antidrug antibodies were detected in 17.6% of patients in each group at week 24. CT-P10 and RTX displayed similar safety profiles. CONCLUSIONS CT-P10 and RTX demonstrated equivalent pharmacokinetics and comparable efficacy, pharmacodynamics, immunogenicity and safety. TRIAL REGISTRATION NUMBER NCT01534884.
Collapse
Affiliation(s)
- Dae Hyun Yoo
- Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | | | | | - Slawomir Jeka
- University Hospital No. 2, Collegium Medicum in Bydgoszcz, UMK in Toruń, Poland
| | | | - Pawel Hrycaj
- Poznań University of Medical Sciences, Poznań, Poland
| | | | - Eun Young Lee
- Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | - Marina Stanislav
- Research Rheumatology Institute n. a. V.A. Nassonova, Moscow, Russia
| | | | | | | | - Mie Jin Lim
- IN-HA University, School of Medicine, Incheon, Korea
| | - Jung-Yoon Choe
- Catholic University of Daegu, School of Medicine, Daegu, Korea
| | | | | | | | - Won Park
- IN-HA University, School of Medicine, Incheon, Korea
| |
Collapse
|
19
|
Liao K, Meyer E, Lee TN, Loercher A, Sikkema D. Inhibition of interleukin-5 induced false positive anti-drug antibody responses against mepolizumab through the use of a competitive blocking antibody. J Immunol Methods 2016; 441:15-23. [PMID: 27889561 DOI: 10.1016/j.jim.2016.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 11/16/2022]
Abstract
Mepolizumab, a humanized IgG1 monoclonal antibody that blocks native homodimeric interleukin-5 (IL-5) from binding to the IL-5 receptor, has recently been approved for treatment of severe eosinophilic asthma. Our initial immunogenicity assay method for phase I and II studies utilized a bridging electrochemiluminescence format with biotin and ruthenium-labelled mepolizumab linked by anti-drug antibodies (ADA). We discovered that IL-5 significantly increased in dosed subjects from a phase II study and that the increased IL-5 was in the form of a drug-bound complex. We demonstrated that the elevated drug-bound IL-5 produced false-positive response in the in vitro ADA assay, in which drug-bound IL-5 dissociated and then bridged mepolizumab conjugates to yield positive signal. To eliminate the IL-5 interference, we compared two strategies: a solid-phase immunodepletion of IL-5 and an in-solution IL-5 immunocompetition. We identified the best competitive antibody for each purpose. We found both methods demonstrated similar effectiveness in reducing the false positive signal in IL-5 spiked samples; however, the in-solution immunocompetition for IL-5 had fewer false positives in study samples. Additionally, the in-solution immunocompetition method was experimentally simpler to execute. We modified the ADA assay by adding a pre-treatment step with a mepolizumab competitive anti- IL-5 antibody. Using this new method, we retested clinical samples from two phase II studies (MEA112997 and MEA114092). The confirmed ADA positive incidence was reduced from 29% and 61% to 1% and 8% with the modified in-solution immune inhibition method. Target interference is a fairly common problem facing immunogenicity testing, and target-induced false positive cannot be distinguished from true ADA response by the commonly used drug competitive confirmation assay. The approach and method used here for resolving target interference in ADA detection will be useful for differentiating between a true ADA response and target induced false positive as well as similar challenges in other programs.
Collapse
Affiliation(s)
- Karen Liao
- Clinical Immunology, GlaxoSmithKline, King of Prussia, PA 19406, United States.
| | - Erik Meyer
- Clinical Immunology, GlaxoSmithKline, King of Prussia, PA 19406, United States
| | - Thomas N Lee
- Clinical Immunology, GlaxoSmithKline, King of Prussia, PA 19406, United States
| | - Amy Loercher
- Clinical Immunology, GlaxoSmithKline, King of Prussia, PA 19406, United States
| | - Daniel Sikkema
- Clinical Immunology, GlaxoSmithKline, King of Prussia, PA 19406, United States
| |
Collapse
|
20
|
Yu B, Shao H, Su C, Jiang Y, Chen X, Bai L, Zhang Y, Li Q, Zhang X, Li X. Exosomes derived from MSCs ameliorate retinal laser injury partially by inhibition of MCP-1. Sci Rep 2016; 6:34562. [PMID: 27686625 PMCID: PMC5043341 DOI: 10.1038/srep34562] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 09/15/2016] [Indexed: 12/20/2022] Open
Abstract
Although accumulated evidence supports the notion that mesenchymal stem cells (MSCs) act in a paracrine manner, the mechanisms are still not fully understood. Recently, MSC-derived exosomes (MSC-Exos), a type of microvesicle released from MSCs, were thought to carry functional proteins and RNAs to recipient cells and play therapeutic roles. In the present study, we intravitreally injected MSCs derived from either mouse adipose tissue or human umbilical cord, and their exosomes to observe and compare their functions in a mouse model of laser-induced retinal injury. We found that both MSCs and their exosomes reduced damage, inhibited apoptosis, and suppressed inflammatory responses to obtain better visual function to nearly the same extent in vivo. Obvious down-regulation of monocyte chemotactic protein (MCP)-1 in the retina was found after MSC-Exos injection. In vitro, MSC-Exos also down-regulated MCP-1 mRNA expression in primarily cultured retinal cells after thermal injury. It was further demonstrated that intravitreal injection of an MCP-1-neutralizing antibody promoted the recovery of retinal laser injury, whereas the therapeutic effect of exosomes was abolished when MSC-Exos and MCP-1 were administrated simultaneously. Collectively, these results suggest that MSC-Exos ameliorate laser-induced retinal injury partially through down-regulation of MCP-1.
Collapse
Affiliation(s)
- Bo Yu
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, USA
| | - Chang Su
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Yuanfeng Jiang
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Xiteng Chen
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Lingling Bai
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Yan Zhang
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Qiutang Li
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, USA
| | - Xiaomin Zhang
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| | - Xiaorong Li
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin 300384, China
| |
Collapse
|
21
|
Habibi-Anbouhi M, Azadmanesh K, Behdani M, Hajizadeh-Saffar E, Vahabpour R, Shokrgozar MA. Development and Characterization of a New Antipeptide Monoclonal Antibody Directed to Human CD20 Antigen. Cancer Biother Radiopharm 2016; 30:310-6. [PMID: 26352927 DOI: 10.1089/cbr.2015.1863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The rapid expansion of immunotherapeutic approaches for treatment of various diseases, including cancers, has been greatly facilitated by the invention of new generation of antibodies. Clinical studies have indicated that anti-CD20 mAb-based therapies represent an effective treatment for various diseases with overexpression of CD20 on their cell surface, such as non-Hodgkin's lymphoma, hemolytic anemia, as well as autoimmune diseases like rheumatoid arthritis. Technically, due to a short extra membrane domain, the recombinant CD20 protein is a difficult antigen to raise immune responses. In search for new monoclonal antibodies, the authors used an antigenic polypeptide, which yielded numbers of new binders that may lead to production of anti-CD20 antibodies, with improved diagnostic or clinical attributes. Mice were immunized with extra membrane loop of human CD20 (exCD20) polypeptide. The exCD20 antigen showed a desired immune response and was able to develop a monoclonal antibody, 3B4C10, which reacted well with peptide antigen as well as native antigen on the surface of Raji B-cell line. The antibody 3B4C10 with a balanced K(on) and K(off) may be applicable in the construction of affinity columns or beads for isolation and purification of CD20-positive cells and cancer stem cells.
Collapse
Affiliation(s)
| | | | - Mahdi Behdani
- 3 Venom & Biotherapeutics Molecules Laboratory, Biotechnology Research Center, Pasteur Institute of Iran , Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- 4 Department of Regenerative Medicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology , ACECR, Tehran, Iran
| | - Rouhollah Vahabpour
- 5 Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | | |
Collapse
|
22
|
Gorovits B, Clements-Egan A, Birchler M, Liang M, Myler H, Peng K, Purushothama S, Rajadhyaksha M, Salazar-Fontana L, Sung C, Xue L. Pre-existing Antibody: Biotherapeutic Modality-Based Review. AAPS J 2016; 18:311-20. [PMID: 26821802 PMCID: PMC4779092 DOI: 10.1208/s12248-016-9878-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/20/2016] [Indexed: 01/12/2023] Open
Abstract
Pre-existing antibodies to biotherapeutic drugs have been detected in drug-naïve subjects for a variety of biotherapeutic modalities. Pre-existing antibodies are immunoglobulins that are either specific or cross-reacting with a protein or glycan epitopes on a biotherapeutic compound. Although the exact cause for pre-existing antibodies is often unknown, environmental exposures to non-human proteins, glycans, and structurally similar products are frequently proposed as factors. Clinical consequences of the pre-existing antibodies vary from an adverse effect on patient safety to no impact at all and remain highly dependent on the biotherapeutic drug modality and therapeutic indication. As such, pre-existing antibodies are viewed as an immunogenicity risk factor requiring a careful evaluation. Herein, the relationships between biotherapeutic modalities to the nature, prevalence, and clinical consequences of pre-existing antibodies are reviewed. Initial evidence for pre-existing antibody is often identified during anti-drug antibody (ADA) assay development. Other interfering factors known to cause false ADA positive signal, including circulating multimeric drug target, rheumatoid factors, and heterophilic antibodies, are discussed.
Collapse
Affiliation(s)
- Boris Gorovits
- Pfizer Worldwide Research & Development, PDM, 1 Burtt Rd, Andover, MA, USA.
| | - Adrienne Clements-Egan
- Janssen Research & Development, LLC (Johnson & Johnson), Welsh and McKean Roads, Spring House, PA, USA
| | - Mary Birchler
- Clinical Immunology, GlaxoSmithKline, King of Prussia, PA, USA
| | - Meina Liang
- MedImmune, Clinical Pharmacology and DMPK, Mountain View, CA, USA
| | - Heather Myler
- Bristol-Myers Squibb, Analytical & Bioanalytical Development, Princeton, NJ, USA
| | - Kun Peng
- Genentech, BioAnalytical Sciences, San Francisco, CA, USA
| | | | - Manoj Rajadhyaksha
- Regeneron Pharmaceuticals, Inc. Bioanalytical Sciences, Tarrytown, NY, USA
| | - Laura Salazar-Fontana
- DSAR, Project Standards and Innovation, Immunology and Biomarkers, Sanofi R&D, Framingham, MA, USA
| | - Crystal Sung
- DSAR, Clinical Laboratory Sciences, Sanofi R&D, Framingham, MA, USA
| | - Li Xue
- Pfizer Worldwide Research & Development, PDM, 1 Burtt Rd, Andover, MA, USA
| |
Collapse
|
23
|
Indirect assessment of neutralizing anti-drug antibodies utilizing pharmacokinetic assay data. J Immunol Methods 2015; 429:28-38. [PMID: 26711311 DOI: 10.1016/j.jim.2015.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 11/23/2022]
Abstract
Neutralizing anti-drug antibodies (NAbs) can adversely impact efficacy and safety of biologic therapeutics. However, current assay formats to detect NAbs are limited in their use during the dosing phase due to interference by circulating drug, resulting in low drug tolerance. To improve the drug tolerance for NAb detection, an alternative approach for indirect NAb (iNAb) assessment was developed and qualified that uses a combination of pharmacokinetic (PK) assays to measure the serum concentrations of free and total drug. It is demonstrated that the ratio of free to total drug concentrations, referred to as F/T ratio, is a novel PK parameter that can indicate neutralizing activity in test samples. The iNAb assessment correctly identified NAb-positive samples with high drug concentrations that led to false negative results in a conventional NAb assay. Moreover, iNAb reliably distinguished between NAbs and non-neutralizing anti-drug antibodies over a wide range of concentrations. A proposal on how to deploy iNAb assessment within a broader immunogenicity testing strategy is presented.
Collapse
|
24
|
Rup B, Pallardy M, Sikkema D, Albert T, Allez M, Broet P, Carini C, Creeke P, Davidson J, De Vries N, Finco D, Fogdell-Hahn A, Havrdova E, Hincelin-Mery A, C Holland M, H Jensen PE, Jury EC, Kirby H, Kramer D, Lacroix-Desmazes S, Legrand J, Maggi E, Maillère B, Mariette X, Mauri C, Mikol V, Mulleman D, Oldenburg J, Paintaud G, R Pedersen C, Ruperto N, Seitz R, Spindeldreher S, Deisenhammer F. Standardizing terms, definitions and concepts for describing and interpreting unwanted immunogenicity of biopharmaceuticals: recommendations of the Innovative Medicines Initiative ABIRISK consortium. Clin Exp Immunol 2015; 181:385-400. [PMID: 25959571 PMCID: PMC4557374 DOI: 10.1111/cei.12652] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2015] [Indexed: 12/17/2022] Open
Abstract
Biopharmaceuticals (BPs) represent a rapidly growing class of approved and investigational drug therapies that is contributing significantly to advancing treatment in multiple disease areas, including inflammatory and autoimmune diseases, genetic deficiencies and cancer. Unfortunately, unwanted immunogenic responses to BPs, in particular those affecting clinical safety or efficacy, remain among the most common negative effects associated with this important class of drugs. To manage and reduce risk of unwanted immunogenicity, diverse communities of clinicians, pharmaceutical industry and academic scientists are involved in: interpretation and management of clinical and biological outcomes of BP immunogenicity, improvement of methods for describing, predicting and mitigating immunogenicity risk and elucidation of underlying causes. Collaboration and alignment of efforts across these communities is made difficult due to lack of agreement on concepts, practices and standardized terms and definitions related to immunogenicity. The Innovative Medicines Initiative (IMI; http://www.imi-europe.org), ABIRISK consortium [Anti-Biopharmaceutical (BP) Immunization Prediction and Clinical Relevance to Reduce the Risk; http://www.abirisk.eu] was formed by leading clinicians, academic scientists and EFPIA (European Federation of Pharmaceutical Industries and Associations) members to elucidate underlying causes, improve methods for immunogenicity prediction and mitigation and establish common definitions around terms and concepts related to immunogenicity. These efforts are expected to facilitate broader collaborations and lead to new guidelines for managing immunogenicity. To support alignment, an overview of concepts behind the set of key terms and definitions adopted to date by ABIRISK is provided herein along with a link to access and download the ABIRISK terms and definitions and provide comments (http://www.abirisk.eu/index_t_and_d.asp).
Collapse
Affiliation(s)
- B Rup
- Pfizer, Immunogenicity Sciences Disciple, Pharmacokinetics, Dynamics and Metabolism
| | - M Pallardy
- INSERM, UMR996, Faculté Pharmacie, Université Paris Sud, France
| | - D Sikkema
- GlaxoSmithKline, Clinical Immunology-Biopharm, King of Prussia, PA, USA
| | - T Albert
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | - M Allez
- Hôpital Saint-Louis, Department of Gastroenterology, GETAID, Paris, France
| | - P Broet
- INSERM, UMR669, University of Paris Sud, France
| | - C Carini
- Pfizer, Early Biotech Clinical Development, Cambridge, MA, USA
| | - P Creeke
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - J Davidson
- GlaxoSmithKline, Worldwide Epidemiology, Southall, UK
| | - N De Vries
- Clinical Immunology and Rheumatology, University of Amsterdam, Amsterdam, the Netherlands
| | - D Finco
- Pfizer, Drug Safety R&D, Groton, CT, USA
| | - A Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - E Havrdova
- Department of Neurology and Center for Clinical Neuroscience, MS Center, Charles University in Prague, Prague, Czech Republic
| | - A Hincelin-Mery
- Sanofi-Aventis, Clinical Exploratory and Pharmacology, Chilly-Mazerin, FR
| | - M C Holland
- GlaxoSmithKline, Clinical Immunology-Biopharm R&D, King of Prussia, PA, USA
| | - P E H Jensen
- Department of Neurology, University of Copenhagen, Copenhagen, Denmark
| | - E C Jury
- Centre for Rheumatology, University College London, London, UK
| | - H Kirby
- UCB Pharma, Bioanalytical R&D, Slough, UK
| | - D Kramer
- Merck-Serono, Institute of Drug Metabolism and Pharmacokinetics, Grafing, Germany
| | | | - J Legrand
- Ipsen Innovation, Pharmacokinetics Drug Metabolism Department, Les Ulis, France
| | - E Maggi
- Dipartimento di Medicina Sperimentale e Clinica, Universita di Firenze, Firenze, Italy
| | - B Maillère
- CEA-Saclay Institute of Biology and Technologies, Gif sur Yvette, France
| | - X Mariette
- INSERM, U1012, Hôpitaux Universitaires Paris Sud, Rhumatologie, Paris, France
| | - C Mauri
- Centre for Rheumatology Research, University College London, London, UK
| | - V Mikol
- Sanofi Aventis, Structural Biology, Paris, France
| | - D Mulleman
- University of Tours Francois Rabelais, CNRS UMR 7292, Tours, France
| | - J Oldenburg
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | - G Paintaud
- CNRS UMR 7292 'GICC', Faculty of Medicine, Tours, France
| | | | - N Ruperto
- Istituto Giannina Gaslini, Pediatria II, Rheumatology, Genova, Italy
| | - R Seitz
- Division of Haematology/Transfusion Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - S Spindeldreher
- Drug Metabolism Pharmacokinetics-Biologics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - F Deisenhammer
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | | |
Collapse
|
25
|
Xin H, Li Y, Chopp M. Exosomes/miRNAs as mediating cell-based therapy of stroke. Front Cell Neurosci 2014; 8:377. [PMID: 25426026 PMCID: PMC4226157 DOI: 10.3389/fncel.2014.00377] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/22/2014] [Indexed: 12/19/2022] Open
Abstract
Cell-based therapy, e.g., multipotent mesenchymal stromal cell (MSC) treatment, shows promise for the treatment of various diseases. The strong paracrine capacity of these cells and not their differentiation capacity, is the principal mechanism of therapeutic action. MSCs robustly release exosomes, membrane vesicles (~30–100 nm) originally derived in endosomes as intraluminal vesicles, which contain various molecular constituents including proteins and RNAs from maternal cells. Contained among these constituents, are small non-coding RNA molecules, microRNAs (miRNAs), which play a key role in mediating biological function due to their prominent role in gene regulation. The release as well as the content of the MSC generated exosomes are modified by environmental conditions. Via exosomes, MSCs transfer their therapeutic factors, especially miRNAs, to recipient cells, and therein alter gene expression and thereby promote therapeutic response. The present review focuses on the paracrine mechanism of MSC exosomes, and the regulation and transfer of exosome content, especially the packaging and transfer of miRNAs which enhance tissue repair and functional recovery. Perspectives on the developing role of MSC mediated transfer of exosomes as a therapeutic approach will also be discussed.
Collapse
Affiliation(s)
- Hongqi Xin
- Department of Neurology, Henry Ford Hospital Detroit, MI, USA
| | - Yi Li
- Department of Neurology, Henry Ford Hospital Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital Detroit, MI, USA ; Department of Physics, Oakland University Rochester, MI, USA
| |
Collapse
|
26
|
Interference in immunoassays to support therapeutic antibody development in preclinical and clinical studies. Bioanalysis 2014; 6:1939-51. [DOI: 10.4155/bio.14.127] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
During preclinical and clinical studies, immunoassays are used to measure the concentration of the therapeutic antibody, anti-drug antibodies and soluble protein biomarkers. The reliability of these assays is crucial since the results are routinely used for safety assessment and dose selection. Furthermore, soluble protein biomarkers can provide information about target engagement, proof of mechanism, proof of principle and prediction of response. Study samples mostly consist of complex matrices that can exhibit considerable interference, resulting in inaccurate measurements. This perspective discusses the source of interference and strategies to mitigate or eliminate interference in immunoassays used during preclinical and clinical drug development of drugs with a focus on the development of therapeutic antibodies.
Collapse
|
27
|
Matrix effect in ligand-binding assay: the importance of evaluating emerging technologies. Bioanalysis 2014; 6:1033-6. [DOI: 10.4155/bio.14.39] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
28
|
Resolution of matrix interference: quantitative and quasi-quantitative ligand-binding assays case studies. Bioanalysis 2014; 6:1093-101. [DOI: 10.4155/bio.14.74] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: Matrix effects pose a constant challenge in developing robust ligand-binding assays to be validated for use in nonclinical and clinical study support. When notable matrix effects of any kind are present, it can render an otherwise sound method ineffective. We present two case studies detailing the mitigation of observed matrix effects. Method: A dimeric protein was removed from unknown samples in an anti-therapeutic antibody assay through protein extraction. Nonspecific matrix effects in a quantitative ligand-binding assays were mitigated through development of a specialized buffer. Results: The protein extraction method reproducibly reduced the artificially high responses of naïve samples, enabling the accurate detection of anti-therapeutic antibodies. Design of experiments was used to evaluate and select the optimal components and associated concentrations in order to reduce the observed matrix effect to acceptable limits. Conclusion: Our results suggest there are multiple techniques available for the bioanalytical scientist to mitigate both matrix effects in ligand-binding assays.
Collapse
|
29
|
Dai S, Schantz A, Clements-Egan A, Cannon M, Shankar G. Development of a method that eliminates false-positive results due to nerve growth factor interference in the assessment of fulranumab immunogenicity. AAPS JOURNAL 2014; 16:464-77. [PMID: 24590506 DOI: 10.1208/s12248-014-9581-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 02/13/2014] [Indexed: 11/30/2022]
Abstract
Fulranumab, a human IgG2 monoclonal antibody that neutralizes nerve growth factor (NGF), is currently in development for the treatment of pain. Our initial immunogenicity test method was found to be prone to NGF interference, leading to a high apparent incidence of anti-drug antibody (ADA) in phase 1 studies. The ADA immunoassay comprised a homogeneous bridging electrochemiluminescence (ECL) format with biotin and ruthenium-labeled fulranumab bound together ("bridged") by ADA in test samples for detection. In this assay, NGF produced a false-positive signal due to its ability to bridge fulranumab molecules. Thus, we developed a specificity assay to eliminate the NGF false-positive results. We encountered the challenge of eliminating drug interference as well as drug target interference, and discovered that the acid-dissociation-based pretreatment of samples used for mitigating drug interference dramatically increased drug target interference. Several strategies were investigated to eliminate the NGF interference; yet only one strategy specifically removed NGF and produced true fulranumab-specific ADA results by using competitive inhibition with fulranumab and utilizing an alternative NGF binding antibody to eliminate NGF interference. Using this new method, we confirmed that the high apparent anti-fulranumab antibody incidence (>60%) in clinical study samples was in fact due to fulranumab-bound NGF released during the acid-dissociation step of the ADA testing method. We conclude that our revised method accurately identifies anti-fulranumab antibodies by incorporating steps to eliminate fulranumab and NGF interference. We advise that acid-dissociation pretreatment must not be universally applied to improve ADA assays without investigating its bioanalytical risks versus benefits.
Collapse
Affiliation(s)
- Sheng Dai
- Biologics Clinical Pharmacology, Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477, USA,
| | | | | | | | | |
Collapse
|