1
|
Sinha A, Rinker S, Souliotis MR, Kumar R, Kumar S, Cowan KJ. Critical reagent considerations for immunogenicity assay development for bispecific biotherapeutic candidates. Bioanalysis 2024; 16:781-790. [PMID: 39023274 DOI: 10.1080/17576180.2024.2366091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Aim: To demonstrate the importance of critical reagent characterization for immunogenicity assay development for multi-specific drugs using two case studies.Methods: Bridging anti-drug antibody (ADA) assay with acid-dissociated samples were used for both cases.Results: In the first case study, the unexpected interference in an ADA assay from clinical samples was identified; a model was created to replicate the issue, and an anti-target antibody was identified to mitigate the target interference. In the second case study, an issue due to non-specific binding of a domain-specific confirmatory reagent was identified, and various mitigation techniques were evaluated.Conclusion: A thorough characterization of the critical reagents helped identify the issues with these ADA case studies and provided strategies for resolving them.
Collapse
Affiliation(s)
- Arkadeep Sinha
- Clinical Bioanalytical Sciences, EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
- Upstream Bio, 460 Totten Pond Rd. Suite 420, Waltham, MA 02451, USA
| | - Sherri Rinker
- B2S Life Sciences, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Mallory Rose Souliotis
- Clinical Bioanalytical Sciences, EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
- Bioagilytix, 1320 Soldiers Field Rd, Boston, MA 02135, USA
| | - Rajesh Kumar
- Clinical Bioanalytical Sciences, EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Seema Kumar
- Clinical Bioanalytical Sciences, EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
- Pioneering Medicines at Flagship Pioneering, 140 First St, Cambridge, MA 02141, USA
| | - Kyra J Cowan
- Merck Healthcare KGaA, Frankfurter Str. 250, Postcode D50/225, 64293, Darmstadt, Germany
| |
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
|
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
|
4
|
Qin Q, Gong L. Current Analytical Strategies for Antibody-Drug Conjugates in Biomatrices. Molecules 2022; 27:6299. [PMID: 36234836 PMCID: PMC9572530 DOI: 10.3390/molecules27196299] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of biotherapeutics, consisting of a cytotoxic payload covalently bound to an antibody by a linker. Ligand-binding assay (LBA) and liquid chromatography-mass spectrometry (LC-MS) are the favored techniques for the analysis of ADCs in biomatrices. The goal of our review is to provide current strategies related to a series of bioanalytical assays for pharmacokinetics (PK) and anti-drug antibody (ADA) assessments. Furthermore, the strengths and limitations of LBA and LC-MS platforms are compared. Finally, potential factors that affect the performance of the developed assays are also provided. It is hoped that the review can provide valuable insights to bioanalytical scientists on the use of an integrated analytical strategy involving LBA and LC-MS for the bioanalysis of ADCs and related immunogenicity evaluation.
Collapse
Affiliation(s)
- Qiuping Qin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| |
Collapse
|
5
|
Partridge MA, Chen J, Karayusuf EK, Sirimanne T, Stefan C, Lai CH, Gathani M, DeStefano L, Rozanski M, McAfee S, Rajadhyaksha M, Andisik MD, Torri A, Sumner G. Pre-existing Reactivity to an IgG4 Fc-Epitope: Characterization and Mitigation of Interference in a Bridging Anti-drug Antibody Assay. AAPS J 2022; 24:78. [DOI: 10.1208/s12248-022-00729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
AbstractTwenty percent of baseline patient samples exhibited a pre-existing response in a bridging anti-drug antibody (ADA) assay for a human IgG4 monoclonal antibody (mAb) therapeutic. In some cases, assay signals were more than 100-fold higher than background, potentially confounding detection of true treatment-emergent ADA responses. The pre-existing reactivity was mapped by competitive inhibition experiments using recombinant proteins or chimeric human mAbs with IgG4 heavy chain regions swapped for IgG1 sequences. These experiments demonstrated that the majority of the samples had reactivity to an epitope containing leucine 445 in the CH3 domain of human IgG4. The pre-existing reactivity in baseline patient samples was mitigated by replacing the ADA assay capture reagent with a version of the drug containing a wild type IgG1 proline substitution at residue 445 without impacting detection of drug-specific, treatment-emergent ADA. Finally, purification on Protein G or anti-human IgG (H + L) columns indicated the pre-existing response was likely due to immunoglobulins in patient samples.
Graphical abstract
Collapse
|
6
|
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
|
7
|
Overcoming multimeric target interference in a bridging immunogenicity assay with soluble target receptor, target immunodepletion and mild acidic assay pH. Bioanalysis 2020; 12:1071-1085. [PMID: 32735128 DOI: 10.4155/bio-2020-0110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Soluble multimeric target proteins can generate a target-mediated false-positive signal in bridging anti-drug antibody (ADA) assays. A high background signal due to target interference was observed in our anti-REGN-Y antibody assay, and two different strategies were evaluated to mitigate this false-positive signal. Results: Multiple anti-target antibodies were tested and found to be ineffective at reducing target interference, so soluble target receptor and co-factor proteins were used in combination to inhibit the target-mediated signal. These competitive blockers synergistically inhibited target interference and increased target tolerance levels, especially when the assay was performed under mild acidic conditions. A separate approach, target immunodepletion using magnetic beads conjugated with an anti-target antibody, was also effective at mitigating the target-mediated signal, also in combination with mild acidic assay pH. Both methods allowed detection of a true ADA signal in monkey and human post-dose serum samples. Conclusion: These methods provide alternative strategies for mitigating target interference when standard anti-target antibodies are ineffective, with the competitive blocker method being recommended, if possible, due to its higher throughput and easier execution.
Collapse
|
8
|
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
|
9
|
Immunogenicity of antibody-drug conjugates: observations across 8 molecules in 11 clinical trials. Bioanalysis 2019; 11:1555-1568. [PMID: 31208199 DOI: 10.4155/bio-2018-0259] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To evaluate the clinical immunogenicity of eight antibody-drug conjugates (ADCs), multi-domain biotherapeutics that could theoretically pose a greater immunogenicity risk than monoclonal antibodies (mAbs) because they contain non-natural structural motifs. Methodology & results: Immunogenicity strategies and assays for these ADCs included those commonly used for conventional biotherapeutics with additional characterization. A tiered approach was adopted for testing Phase I and II clinical study samples with screening, confirmatory assays and additional domain characterization. Antidrug antibody incidences with these ADCs were within those reported for mAb biotherapeutics with no apparent impact on clinical outcomes. Conclusion: These data suggest that the ADC hapten-like structure across these eight ADCs does not appear to increase patient immune responses beyond those generally observed for mAb biotherapeutics.
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
|
Dingman R, Balu-Iyer SV. Immunogenicity of Protein Pharmaceuticals. J Pharm Sci 2019; 108:1637-1654. [PMID: 30599169 PMCID: PMC6720129 DOI: 10.1016/j.xphs.2018.12.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Protein therapeutics have drastically changed the landscape of treatment for many diseases by providing a regimen that is highly specific and lacks many off-target toxicities. The clinical utility of many therapeutic proteins has been undermined by the potential development of unwanted immune responses against the protein, limiting their efficacy and negatively impacting its safety profile. This review attempts to provide an overview of immunogenicity of therapeutic proteins, including immune mechanisms and factors influencing immunogenicity, impact of immunogenicity, preclinical screening methods, and strategies to mitigate immunogenicity.
Collapse
Affiliation(s)
- Robert Dingman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214.
| |
Collapse
|
12
|
A bridging immunogenicity assay for monoclonal antibody: case study with SHR-1222. Bioanalysis 2018; 10:1115-1127. [DOI: 10.4155/bio-2017-0289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Aim: SHR-1222 is a humanized monoclonal antibody targeted to soluble sclerostin. To support the preclinical study of SHR-1222 in cynomolgus monkeys, a method for the detection of anti-drug antibodies is required. Results: A bridging immunogenicity method for the detection of anti-SHR-1222 antibodies was developed and validated. In the method, minimal required dilution, normalization factor and confirmatory cut point were 1:20, 4.35 and 10.45%, respectively. The method was successfully applied to evaluate a multiple-dose toxicity study in monkeys. Conclusion: The proposed method allows for the detection of anti-SHR-1222 antibodies in preclinical studies and aids in the interpretation of pharmacokinetic changes in certain animals. The soluble targets interference on anti-drug antibody detection can be blocked or decreased by the therapeutic drug.
Collapse
|
13
|
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]
|
14
|
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]
|
15
|
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
|
16
|
Emerging Technologies and Generic Assays for the Detection of Anti-Drug Antibodies. J Immunol Res 2016; 2016:6262383. [PMID: 27556048 PMCID: PMC4983396 DOI: 10.1155/2016/6262383] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/26/2016] [Accepted: 06/09/2016] [Indexed: 12/27/2022] Open
Abstract
Anti-drug antibodies induced by biologic therapeutics often impact drug pharmacokinetics, pharmacodynamics response, clinical efficacy, and patient safety. It is critical to assess the immunogenicity risk of potential biotherapeutics in producing neutralizing and nonneutralizing anti-drug antibodies, especially in clinical phases of drug development. Different assay methodologies have been used to detect all anti-drug antibodies, including ELISA, radioimmunoassay, surface plasmon resonance, and electrochemiluminescence-based technologies. The most commonly used method is a bridging assay, performed in an ELISA or on the Meso Scale Discovery platform. In this report, we aim to review the emerging new assay technologies that can complement or address challenges associated with the bridging assay format in screening and confirmation of ADAs. We also summarize generic anti-drug antibody assays that do not require drug-specific reagents for nonclinical studies. These generic assays significantly reduce assay development efforts and, therefore, shorten the assay readiness timeline.
Collapse
|
17
|
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
|
18
|
Affinity capture elution bridging assay: A novel immunoassay format for detection of anti-therapeutic protein antibodies. J Immunol Methods 2016; 431:45-51. [PMID: 26874304 DOI: 10.1016/j.jim.2016.02.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND Increased emphasis on the development of biologics has placed a significant focus on anti-drug antibody (ADA) detection. To address this need, several immunoassay formats have been described for use in characterizing potential immune responses. Two commonly utilized methods include the affinity capture elution (ACE) and bridging formats. While these approaches have been effective in supporting many clinical initiatives, both possess potential disadvantages. Here, we compare these standard methods to a novel format that addresses these noted drawbacks. RESULTS A novel assay format has been designed to incorporate the benefits of the ACE and bridging methods while overcoming the disadvantages incurred with each approach. The described ACE-Bridge format exhibits excellent sensitivity and precision while providing superior drug tolerance when compared to bridging formats. Further, this assay format is not susceptible to the endogenous target interference that can be an issue in the ACE format. CONCLUSIONS The ACE-Bridge format provides an often superior option as a screening method to monitor patient ADA responses. This method is unique in its ability to measure ADA in the presence of high circulating endogenous target concentrations (>100 ng/mL) while demonstrating very high drug tolerance.
Collapse
|
19
|
|
20
|
How the bioanalytical scientist plays a key role in interdisciplinary project teams in the development of biotherapeutics – a reflection of the European Bioanalysis Forum. Bioanalysis 2014; 6:1339-48. [DOI: 10.4155/bio.14.90] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The bioanalytical scientist plays a key role in the project team for the drug development of biotherapeutics from the discovery to the marketing phase. Information from the project team members is required for assay development and sample analysis during the discovery, preclinical and clinical phases of the project and input is needed from the bioanalytical scientist to help data interpretation. The European Bioanalysis Forum target team 20 discussed many of the gaps in information and communication between the bioanalytical scientist and project team members as a base for providing a perspective on the bioanalytical scientist's role and interactions within the project team.
Collapse
|
21
|
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
|
22
|
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
|
23
|
Development of a biosensor-based immunogenicity assay capable of blocking soluble drug target interference. J Immunol Methods 2013; 396:44-55. [PMID: 23933325 DOI: 10.1016/j.jim.2013.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 07/23/2013] [Accepted: 07/23/2013] [Indexed: 01/13/2023]
Abstract
As with other protein therapeutics, trebananib (AMG 386), an investigational peptide Fc-fusion protein ("peptibody") that inhibits angiogenesis by neutralizing the interaction of angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) with the Tie2 receptor, has the potential to trigger an immune response in cancer patients treated with the therapeutic. An electrochemiluminescence bridging anti-drug antibody (ADA) assay that was utilized to support early-phase clinical trials in the development of trebananib was found to lack adequate sensitivity and drug tolerance in later-phase clinical studies when higher doses of trebananib were administered. Therefore, we developed a surface plasmon resonance (SPR) immunoassay method utilizing a secondary confirmatory detector antibody (goat anti-human IgG F[ab']2) known to cross-react with human IgG and IgM to better assess the potential impact of immunogenicity on the pharmacokinetics, pharmacodynamics, and toxicity of trebananib. The SPR method was more sensitive than the electrochemiluminescence bridging assay because of signal amplification from the confirmatory binding of the detector antibody; drug tolerance was improved since antibody binding avidity does not affect detection on this platform. Despite the inability of the confirmatory detector antibody to bind angiopoietins in protein-free buffer, false-positive ADA results were generated from patient serum samples containing Ang1 and Ang2 through an apparently specific binding between the angiopoietins and the confirmatory detector antibody, likely mediated by the interaction of the angiopoietins with serum immunoglobulins. Addition to the sample diluent of a human antibody that specifically binds to Ang1 and Ang2 with high affinity resulted in a complete block of angiopoietin interference without affecting ADA detection. This biosensor-based assay provides a reliable method for assessing immunogenicity in phase 3 clinical trials.
Collapse
|
24
|
Novel data analysis methods to overcome cut point challenges and enable comprehensive assessment of antidrug binding activity in confirmatory assays. J Immunol Methods 2013; 392:38-48. [DOI: 10.1016/j.jim.2013.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 10/27/2022]
|
25
|
Immunogenicity assays for antibody–drug conjugates: case study with ado-trastuzumab emtansine. Bioanalysis 2013; 5:1007-23. [DOI: 10.4155/bio.13.64] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Antibody–drug conjugates (ADCs) such as Kadcyla™ (ado-trastuzumab emtansine [T-DM1]) present covalently bound cytotoxic drugs, which may influence their immunogenicity potential compared with antibody therapies. Therefore, ADCs require assay strategies that allow measurement of responses to all the molecular components. Results: The immunogenicity strategy for T-DM1 used a risk-based, tiered approach that included screening and titration to detect antitherapeutic antibodies; confirmation of positive responses; and characterization to assess whether the immune response is primarily to the antibody or to the linker–drug and/or new epitopes in trastuzumab resulting from conjugation. Conclusion: The tiered immunogenicity assay strategy for T-DM1 allowed detection of antitherapeutic antibodies to all components of the ADC in multiple nonclinical and clinical studies. Characterization strategies implemented in clinical studies provided additional insights into the specificity of the immune response.
Collapse
|
26
|
False-positive immunogenicity responses are caused by CD20+ B cell membrane fragments in an anti-ofatumumab antibody bridging assay. J Immunol Methods 2013; 394:22-31. [PMID: 23639298 DOI: 10.1016/j.jim.2013.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/11/2013] [Accepted: 04/23/2013] [Indexed: 11/23/2022]
Abstract
An electrochemiluminescent (ECL) bridging assay to detect anti-ofatumumab antibodies (ADA) in human serum samples was developed and validated. Using this assay format, clinical samples were first screened to identify potential ADA positive samples, which were then further tested by adding excess drug, confirming the positive signals as drug specific. However, when the method was implemented into clinical studies for ADA testing, a high positive rate was observed in the pre-dose samples collected from patients with chronic lymphocytic leukemia (CLL). Since the positive signals were not associated with ofatumumab (Ofa) treatment, and diminished after treatment, it was suspected that matrix interference might be responsible, resulting in false-positive responses. We performed a series of experimental investigations to identify, characterize, minimize or eliminate the possible false-positive responses. One possible source was identified to be CD20 (the target of Ofa) present on cell membrane fragments (CMFs). The false-positive responses caused by CD20(+) CMFs could be reduced by solid-phase immunodepletion, ultracentrifugation, or inhibited by adding another anti-CD20 antibody (rituximab). As a consequence, the ADA method was modified to minimize the matrix interference caused by CD20(+) CMFs and, then, validated for sample testing.
Collapse
|
27
|
Bioanalytical assay strategies for the development of antibody–drug conjugate biotherapeutics. Bioanalysis 2013; 5:201-26. [DOI: 10.4155/bio.12.299] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Antibody–drug conjugates (ADCs) are monoclonal antibodies with covalently bound cytotoxic drugs. They are designed to target tumor antigens selectively and offer the hope of cancer treatment without the debilitating side-effects of conventional therapies. The concept of ADCs is not new; however, development of these therapeutics is challenging and only recently are promising clinical data emerging. These challenges include ADC bioanalysis, such as quantifying in serum/plasma for PK studies and strategies for assessing immunogenicity. ADCs have complex molecular structures incorporating large- and small-molecule characteristics and require diverse analytical methods, including ligand-binding assays and MS-based methods. ADCs are typically mixtures with a range of drug-to-antibody ratios. Biotransformations in vivo can lead to additional changes in drug-to-antibody ratios resulting in dynamically changing mixtures. Thus, a standard calibration curve consisting of the reference standard may not be appropriate for quantification of analytes in vivo and represents a unique challenge. This paper will share our perspective on why ADC bioanalysis is so complex and describe the strategies and rationale that we have used for ADCs, with highlights of original data from a variety of nonclinical and clinical case studies. Our strategy has involved novel protein structural characterization tools to help understand ADC biotransformations in vivo and use of the analyte knowledge gained to guide the development of quantitative bioanalytical assays.
Collapse
|