1
|
Muccio S, Hirtz C, Kramer D, Paris J, Descloux S, Fedeli O, Deiteren A, Tribula A, Lehmann S, Vialaret J. In-depth characterization and semi-quantification of anti-drug antibodies in clinical samples using specific hybrid IC-LC-MS/MS methods. Anal Biochem 2025; 701:115797. [PMID: 39921137 DOI: 10.1016/j.ab.2025.115797] [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: 10/08/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
Anti-drug antibodies (ADAs) generated by biotherapeutics can impair the drug clearance, prevent the binding to its target or lead to hypersensitivity reactions, thereby affecting efficacy and safety. It is therefore essential to assess the immunogenicity of potential biotherapeutics, particularly in clinical development. Ligand binding assays (LBA) are the gold standard for ADA detection because of their high sensitivity and throughput. However, LBA assays don't provide details on the isotypes produced and their relative abundance. As certain isotypes are known to be associated with ADA mediated adverse events, this information could be helpful to anticipate or better characterize the immunogenicity risk of biotherapeutics. A hybrid IC-LC-MS/MS strategy was developed for the detection of specific isotypes/subclasses of ADAs in a phase I clinical study. A first approach using the biotinylated drug to capture ADAs in human serum allowed the simultaneous semi-quantification of all IgG subclasses and the detection of ADAs of the IgM isotype in clinical samples. These results enabled a detailed characterization of the immune response against the biotherapeutic. A second assay was developed using a sequential immunocapture to measure drug specific IgEs known to be potentially associated with hypersensitivity reactions. The overall results were consistent with the clinical adverse events observed in some healthy volunteers.
Collapse
Affiliation(s)
- Stéphane Muccio
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France.
| | - Christophe Hirtz
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Daniel Kramer
- Sanofi, TMED-BCB, Industriepark Höchst 65926 Frankfurt, Germany
| | - Johanna Paris
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Sandrine Descloux
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Olivier Fedeli
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Annemie Deiteren
- Ablynx R&D, TMED-TMCP, Technologiepark-Zwijnaarde 21, 9052 Gent/Zwijnaarde, Belgium
| | | | - Sylvain Lehmann
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France.
| | - Jérôme Vialaret
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| |
Collapse
|
2
|
Pap T, Király N, Fekete A, Tóth JP, Kónya A. Target tolerance improvement of the immunogenicity assay developed for analysing samples from clinical trials of RGB-14, a proposed biosimilar to Prolia/Xgeva. J Immunol Methods 2025; 541:113878. [PMID: 40368321 DOI: 10.1016/j.jim.2025.113878] [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: 01/23/2025] [Revised: 04/24/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
An immunogenicity assay was developed to support the clinical program of RGB-14, a proposed biosimilar to Prolia/Xgeva. The ADA assay was successfully validated following current guidelines and white papers. The validation included an assessment of target (RANKL) interference, and the assay passed the test up to a 30 pg/mL RANKL level. This concentration was anticipated to be the maximum level present in clinical samples. Despite the successful validation, most of the analysed samples from the RGB-14 Phase I clinical trial yielded false-positive results for anti-denosumab antibodies. The investigation concluded that unexpectedly high levels of RANKL were present in the samples and caused interference in the assay. Consequently, the assay was re-developed by adding target-specific reagent, eliminating acid dissociation step and doubling the assay dilution to enhance target tolerance to 10,000 pg/mL RANKL, a requirement for clinical sample testing. The re-developed assay underwent successful validation, and the samples from the clinical trials of RGB-14 were subsequently analysed with the improved assay. The article outlines a strategy for enhancing the target (RANKL) tolerance of the ADA assay. It also underscores the importance of monitoring clinical sample analysis results, even if a validated assay is used for sample testing, as clinical samples may differ from the spiked samples prepared for validation. If unexpected results are obtained, an investigation should be initiated, and the assay should be improved if needed to ensure the reliability of the results.
Collapse
Affiliation(s)
- Tímea Pap
- Developmental Drug Metabolism & Pharmacokinetics, Gedeon Richter Plc, 19-21 Gyömrői st, Budapest H-1103, Hungary.
| | - Nikolett Király
- Developmental Drug Metabolism & Pharmacokinetics, Gedeon Richter Plc, 19-21 Gyömrői st, Budapest H-1103, Hungary
| | - Anna Fekete
- Developmental Drug Metabolism & Pharmacokinetics, Gedeon Richter Plc, 19-21 Gyömrői st, Budapest H-1103, Hungary
| | - János Pál Tóth
- Developmental Drug Metabolism & Pharmacokinetics, Gedeon Richter Plc, 19-21 Gyömrői st, Budapest H-1103, Hungary
| | - Attila Kónya
- Developmental Drug Metabolism & Pharmacokinetics, Gedeon Richter Plc, 19-21 Gyömrői st, Budapest H-1103, Hungary
| |
Collapse
|
3
|
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
|
4
|
Nishio R, Nakanaga K, Yoshinaga R, Kurihara A, Okano F. Development and validation of an antidrug antibody assay for TRK-950 using Melon™ gel and SPEAD pretreatment. Bioanalysis 2025; 17:515-524. [PMID: 40243353 PMCID: PMC12087920 DOI: 10.1080/17576180.2025.2493607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
AIM & METHODS TRK-950, a humanized IgG1 monoclonal antibody against CAPRIN-1, which is reported to be expressed on the cell surface of various forms of cancer, is currently being developed for multiple types of solid cancer. We developed an antidrug antibody (ADA) assay that includes two pretreatment processes for use in clinical trials, namely, Melon™ gel treatment and solid-phase extraction with acid dissociation (SPEAD), to reduce baseline variability between individual serum samples and improve drug tolerance. RESULTS Comparing the assay with or without the pretreatment process using commercially available human serum samples, the mean response calculated from 18 individual human samples decreased from 327 to 270, and the coefficient of variation decreased from 41% to 16%. The new assay with the two-step pretreatment met the criteria set by the regulatory agency throughout the validation study. We also confirmed the recovery rates of IgG, IgM, and ADA - drug immune complexes after treatment with Melon™ gel. Clinical trials (NCT05423262) employing this improved analytical method have been conducted, and the results confirm the low immunogenicity of TRK-950. CONCLUSION The combination of pretreatment with Melon™ gel and SPEAD is applicable for clinical ADA assays.
Collapse
Affiliation(s)
- Reiji Nishio
- New Projects Development Division, Toray Industries, Inc, Kamakura, Japan
| | - Keita Nakanaga
- New Frontiers Research Laboratories, Toray Industries, Inc, Kamakura, Japan
| | - Ryosuke Yoshinaga
- Biomedical Analysis Laboratories, Toray Research Center, Inc, Kamakura, Japan
| | - Akira Kurihara
- New Projects Development Division, Toray Industries, Inc, Kamakura, Japan
| | - Fumiyoshi Okano
- New Projects Development Division, Toray Industries, Inc, Kamakura, Japan
- New Frontiers Research Laboratories, Toray Industries, Inc, Kamakura, Japan
| |
Collapse
|
5
|
Chen F, He X, Mao Y, Coble K. Method development for the detection of anti-drug antibodies against a therapeutic peptide: assay format selection. Bioanalysis 2025; 17:537-548. [PMID: 40346877 DOI: 10.1080/17576180.2025.2501937] [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: 01/09/2025] [Accepted: 05/01/2025] [Indexed: 05/12/2025] Open
Abstract
AIM Monitoring immune responses to therapeutic peptides with endogenous counterparts is crucial for evaluating drug safety and efficacy. In this paper, we focused on the selection of an optimal assay format to develop a sensitive, robust, and drug-tolerant immunoassay for the detection of anti-drug antibody (ADA) against a therapeutic peptide. RESULTS We assessed distinct ADA assay formats for preclinical and clinical studies, such as direct binding with labeled protein A/G, direct binding with labeled multiple species-specific antibodies for detection, bridging and affinity capture elution (ACE) formats. The assay formats were evaluated based on multiple assay parameters including sensitivity, drug tolerance, individual matrix variability and inter-assay precision. Overall, direct binding assay with labeled protein A/G for detection, which utilized less labeled peptide drug and achieved desired sensitivity and drug tolerance, is appropriate for preclinical studies. Bridging assay is more suitable format to support clinical studies as bridging assay has less assay variability than ACE assay. CONCLUSION This study highlighted advantages and limitations of each ADA assay format for peptide drugs and evaluated the performance of different assay formats in the assay development process to aid in the selection of the best fit-for-purpose assay formats for preclinical and clinical phases.
Collapse
Affiliation(s)
- Fangfang Chen
- Bioanalytical Sciences, DMPK, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Xiaolong He
- Bioanalytical Sciences, DMPK, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Yan Mao
- Bioanalytical Sciences, DMPK, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Kelly Coble
- Bioanalytical Sciences, DMPK, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| |
Collapse
|
6
|
Nielsen OH, Hammerhøj A, Ainsworth MA, Gubatan J, D'Haens G. Immunogenicity of Therapeutic Antibodies Used for Inflammatory Bowel Disease: Treatment and Clinical Considerations. Drugs 2025; 85:67-85. [PMID: 39532820 DOI: 10.1007/s40265-024-02115-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
The introduction of tumor necrosis factor inhibitors has led to a paradigm shift in the management of inflammatory bowel disease (IBD). The subsequent introduction of both anti-integrins and cytokine blockers has since expanded the biologic armamentarium. However, immunogenicity, defined as the production of anti-drug antibodies (ADAs) to the prescribed biopharmaceutical, means a significant fraction of patients exposed to biologic agents will experience a secondary loss of response to one or more of the drugs. In clinical settings, immunogenicity may be caused by several factors, both patient related (e.g., underlying chronic disease, systemic immune burden, including previous biologic therapy failure, and [epi]genetic background) and treatment related (e.g., dose and administration regimens, drug physical structure, photostability, temperature, and agitation). Here, we outline these elements in detail to enhance biopharmaceutical delivery and therapy for patients with IBD. Moreover, concurrent immunomodulator medication may reduce the risks of ADA generation, especially when using the chimeric drug infliximab. Summarizing the latest developments and knowledge in the field, this review aims to provide strategies to prevent ADA production and information on managing non-responsiveness or loss of response to biologics. Better understanding of the molecular mechanisms underlying the formation of ADAs and the critical factors influencing the immunogenicity of biopharmaceuticals may lead to improved health outcomes in the IBD community that may benefit both the individual patient and society through lower healthcare expenses.
Collapse
Affiliation(s)
- Ole Haagen Nielsen
- Department of Gastroenterology D112, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Copenhagen, Denmark.
| | - Alexander Hammerhøj
- Department of Gastroenterology D112, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Copenhagen, Denmark
| | - Mark Andrew Ainsworth
- Department of Gastroenterology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - John Gubatan
- Department of Gastroenterology & Hepatology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Geert D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Leisegang R, Silber Baumann HE, Lennon-Chrimes S, Ito H, Miya K, Genin JC, Plan EL. Immunogenicity dynamics and covariate effects after satralizumab administration predicted with a hidden Markov model. CPT Pharmacometrics Syst Pharmacol 2024; 13:2171-2184. [PMID: 39380259 DOI: 10.1002/psp4.13230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 10/10/2024] Open
Abstract
Immunogenicity is the propensity of a therapeutic protein to generate an immune response to itself. While reporting of antidrug antibodies (ADAs) is increasing, model-based analysis of such data is seldom performed. Model-based characterization of factors affecting the emergence and dissipation of ADAs may inform drug development and/or improve understanding in clinical practice. This analysis aimed to predict ADA dynamics, including the potential influence of individual covariates, following subcutaneous satralizumab administration. Satralizumab is a humanized IgG2 monoclonal recycling IL-6 receptor antagonist antibody approved for treating neuromyelitis optica spectrum disorder (NMOSD). Longitudinal pharmacokinetic (PK) and ADA data from 154 NMOSD patients in two pivotal Phase 3 studies (NCT02028884, NCT02073279) and PK data from one Phase 1 study (SA-001JP) in 72 healthy volunteers were available for this analysis. An existing population PK model was adapted to derive steady-state concentration without ADA for each patient. A mixed hidden Markov model (mHMM) was developed whereby three different states were identified: one absorbing Markov state for non-ADA developer, and two dynamic and inter-connected Markov states-transient ADA negative and positive. Satralizumab exposure and body mass index impacted transition probabilities and, therefore, the likelihood of developing ADAs. In conclusion, the mHMM model was able to describe the time course of ADA development and identify patterns of ADA development in NMOSD patients following treatment with satralizumab, which may allow for the formulation of strategies to reduce the emergence or limit the impact of ADA in the clinical setting.
Collapse
Affiliation(s)
- Rory Leisegang
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hanna E Silber Baumann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | | | | | | | - Jean-Christophe Genin
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Elodie L Plan
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Xu X, Xu Y, Liang S. Analytical Interference in Chemiluminescence Assay-Measured Angiotensin I, Angiotensin II, Aldosterone, and Renin. J Clin Lab Anal 2024; 38:e25045. [PMID: 38822626 PMCID: PMC11211672 DOI: 10.1002/jcla.25045] [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: 11/16/2023] [Revised: 03/08/2024] [Accepted: 05/05/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND The interference can be a significant source of laboratory errors with the potential to cause immunoassay results to drift. Therefore, we evaluated the interference in various endogenous and exogenous substances on immunoassay for angiotensin I (Ang I), angiotensin II (Ang II), aldosterone, and renin in vitro. METHODS Ten endogenous and eight exogenous substances were evaluated at supraphysiologic or supratherapeutic plasma levels using the screening study to identify potential interfering substances. Subsequently, potential interfering substances were further tested within maximum pathological or therapeutic plasma concentration ranges using the dose-response study to determine whether the interference has a significant bias. According to preset acceptance criteria, the interference in potential interfering substances for Ang I, Ang II, and renin and aldosterone assays was determined. RESULTS Six potential interfering substances for Ang I immunoassays were identified, namely valsartan, nifedipine, spironolactone, cholesterol, hemoglobin, and triglyceride. Meanwhile, ethanol, nifedipine, spironolactone, heparin sodium, warfarin, hemoglobin, uric acid, cholesterol, and triglyceride appeared to have potential interference in the Ang II assay. Three identified as possible interferents for aldosterone immunoassays were glucose, valsartan, and spironolactone. Moreover, warfarin, valsartan, spironolactone, uric acid, cholesterol, bilirubin unconjugated, triglyceride, and hemoglobin were potential interfering substances for renin immunoassays. However, only spironolactone of these potential interfering substances exceeded preset mean bias limits (less than ±10.0%) in aldosterone immunoassays. CONCLUSION Exogenous spironolactone caused clinically significant interference in aldosterone immunoassays. Moreover, the interference in other substances was acceptable in Ang I, Ang II, and renin and aldosterone immunoassays.
Collapse
Affiliation(s)
- Xiaohua Xu
- Department of Clinical Laboratory, The 909th Hospital, School of MedicineXiamen UniversityZhangzhouFujianChina
| | - Yongzhi Xu
- Department of Clinical Laboratory, The 909th Hospital, School of MedicineXiamen UniversityZhangzhouFujianChina
| | - Shengqiang Liang
- Department of Clinical Laboratory, The 909th Hospital, School of MedicineXiamen UniversityZhangzhouFujianChina
| |
Collapse
|
11
|
Fu R, Xu J, Guo Q, Liu T, Su X, Xu M, Zhao X, Wang F, Ji L, Qian W, Hou S, Li J, Zhang D, Guo H. Highly drug/target-tolerant neutralizing antibody (NAb) assay development through target-based drug depletion and drug-based NAb extraction for an anti-EGFR therapeutic monoclonal antibody. J Pharm Biomed Anal 2024; 241:116006. [PMID: 38309099 DOI: 10.1016/j.jpba.2024.116006] [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: 11/10/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/05/2024]
Abstract
The reduction of immunogenicity is fundamental for the development of biobetter Erbitux, given that the development of an immune response reduces treatment efficacy and may lead to potential side effects. One of the requirements for the clinical research of a Erbitux biobetter candidate (CMAB009) is to develop a neutralizing antibody (NAb) assay, and sufficient drug and target tolerance for the assay is necessary. Here, we describe the development of a competitive ligand binding (CLB) assay for CMAB009 with high drug and target tolerance through target-based drug depletion and drug-based NAb extraction, the integrated experimental strategy was implemented to simultaneously mitigate drug interference and enhance target tolerance. Following troubleshooting and optimization, the NAb assay was validated for clinical sample analysis with the sensitivity of 92 ng/mL, drug tolerance of 70 μg/mL and target tolerance of 798 ng/mL. The innovative drug depletion and NAb extraction achieved though the combination of drug and target beads would enable the development of reliable NAb assays for many other therapeutics that overcome drug and its target interference for more precise and sensitive NAb assessment.
Collapse
Affiliation(s)
- Rongrong Fu
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Jin Xu
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qingcheng Guo
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Taizhou Mabtech Pharmaceuticals Co., Ltd, Taizhou, China
| | - Tao Liu
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinyi Su
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, Shanghai Zhangjiang Biotechnology Co., Ltd, Shanghai, China
| | - Mengjiao Xu
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Xiang Zhao
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Fugui Wang
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China
| | - Lusha Ji
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weizhu Qian
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sheng Hou
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jun Li
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Dapeng Zhang
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Huaizu Guo
- State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China; NMPA Key Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai, China; State key laboratory of macromolecular drugs and large-scale manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; State key laboratory of macromolecular drugs and large-scale manufacturing, Shanghai Zhangjiang Biotechnology Co., Ltd, Shanghai, China.
| |
Collapse
|
12
|
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
|
13
|
Polsky R, Gunn G, Reese KJ, Hottenstein CS, Gehman A, Schwartz A, Root D, Concannon A. Strategy and validation of a nonclinical generic plug-and-play antidrug antibody method for human monoclonal antibody biotherapeutics. Bioanalysis 2024; 16:277-287. [PMID: 38334073 DOI: 10.4155/bio-2023-0184] [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] [Indexed: 02/10/2024] Open
Abstract
The measurement of antidrug antibodies (ADA) in nonclinical studies provides limited value because the formation and incidence of nonclinical ADA does not translate to clinical experience. The formation and presence of ADA in nonclinical species can, however, correlate to reduced drug exposure and safety observations including vasculitis and immune complex disease. Generic ADA methods for humanized monoclonal antibody biotherapeutics mitigate the need to develop bespoke ADA methods during nonclinical drug development. A drug-tolerant, sensitive, generic ADA immunoassay has been developed and validated for measuring ADA in cynomolgus monkey serum samples, allowing for immediate qualification of future monoclonal antibody biotherapeutics. This approach allows us to differentiate complexed and free ADA in a rapidly deployable manner when needed.
Collapse
Affiliation(s)
- Rodd Polsky
- GSK, Immunogenicity Group, Collegeville, PA 19426, USA
| | - George Gunn
- GSK, Immunogenicity Group, Collegeville, PA 19426, USA
| | | | | | - Andrew Gehman
- GSK, Immunogenicity Group, Collegeville, PA 19426, USA
| | - Ann Schwartz
- GSK, Immunogenicity Group, Collegeville, PA 19426, USA
| | - Devin Root
- GSK, Immunogenicity Group, Collegeville, PA 19426, USA
| | - Amy Concannon
- GSK, Immunogenicity Group, Collegeville, PA 19426, USA
| |
Collapse
|
14
|
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
|
15
|
Xiang D, Li N, Liu L, Yu H, Li X, Zhao T, Liu D, Gong X. Development and validation of enzyme-linked immunosorbent assays for the measurement of infliximab and anti-drug antibody levels. Heliyon 2023; 9:e21858. [PMID: 38034789 PMCID: PMC10682623 DOI: 10.1016/j.heliyon.2023.e21858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/11/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Infliximab and its anti-drug antibody (ADA) serum concentrations exhibit a strong correlation with clinical response and loss of response. The use of therapeutic drug monitoring to measure the concentration of infliximab and ADA can facilitate clinical decision-making, helping patients attain optimal therapeutic effects. However, there are still limitations to the existing infliximab and its ADA detection methods. Therefore, this study aimed to develop and validate enzyme-linked immunosorbent assay (ELISA)-based methods for measuring infliximab and its ADA levels in human plasma according to the general recommendations for immunoassays. Free infliximab is bound by recombinant TNF-α and detected using HRP-labeled anti-human antibody. The ADA is captured by on-plate-coated infliximab and recognized by biotin-labeled infliximab. Two bridging ELISA assays were developed and after assay optimization and validation, these assays have been applied in ten patients with inflammatory bowel disease (IBD). In infliximab detection assay, a standard curve ranging from 0.10 μg/mL to 8.0 μg/mL with great precision and accuracy has been established. Drug tolerance of the ADA assay was that 100 ng/mL ADA could tolerate at least 5.0 μg/mL infliximab in the plasma using a commercially available monoclonal anti-infliximab antibody as the positive control. The ADA screening and confirmatory assays achieved a sensitivity of 36.74 ng/mL and 37.15 ng/mL, respectively. All other assay characteristics met the requirements. The mean concentration of infliximab in eight patients with IBD was 7.88 (1.87-21.1) μg/mL, and the ADA levels were all negative. Moreover, the concentrations of infliximab in the remaining two patients were below the LLOQ and the ADAs were positive. Thus, accurate and sensitive ELISA methods have been developed and validated for the detection of infliximab and its ADA concentrations and have been successfully applied to clinical therapeutic drug monitoring.
Collapse
Affiliation(s)
- Dong Xiang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ninghong Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pharmacy, Nanchang First Hospital, Nanchang, 330008, China
| | - Lu Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hengyi Yu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiping Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tinghui Zhao
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuepeng Gong
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
16
|
Yang X, Siradze K, Sperinde G, Arjomandi A, Fischer S. Evaluation of multiple immunoassay formats for detection of anti-drug antibodies to zinpentraxin alfa. J Immunol Methods 2023; 522:113573. [PMID: 37816404 DOI: 10.1016/j.jim.2023.113573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/31/2023] [Accepted: 10/06/2023] [Indexed: 10/12/2023]
Abstract
Zinpentraxin alfa (rhPTX-2; PRM-151) is currently being developed for the treatment of fibrotic diseases such as idiopathic pulmonary fibrosis and myelofibrosis. Notably, because it is administered chronically and has an endogenously expressed counterpart, clinical studies of zinpentraxin alpha must include immunogenicity assessments. Since the typical homogenous bridging ELISA assay does not adequately measure anti-drug antibodies (ADAs) against zinpentraxin alfa, additional assay formats have been developed to evaluate immunogenicity of this therapeutic. Here, we present the evaluation of four distinct assay formats that were used to measure zinpentraxin alpha ADA: step-wise bridging, direct binding, total ADA, and the semi-homogeneous formats, based on multiple parameters including assay sensitivity, precision, and drug tolerance. This paper presents the full details of method development for each of the aforementioned assay formats including evaluation of sample pre-treatment, determination of cut point, and assessment of assay performance by analyzing a subset of clinical samples. Overall, the semi-homogenous ADA assay format with no sample pre-treatment was selected for the measurement of zinpentraxin alpha immunogenicity as it provided the desired sensitivity, drug tolerance, and reproducibility. Our study emphasizes the importance of assay format evaluation during drug development and the necessity to select the most suitable assay format and sample pre-treatment method by which to evaluate therapeutic drug immunogenicity.
Collapse
Affiliation(s)
- Xiaoyun Yang
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Ketevan Siradze
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Gizette Sperinde
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Audrey Arjomandi
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Saloumeh Fischer
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
17
|
Luong M, Wang Y, Donnelly BB, Lepsy C. Addressing Domain Specificity in the Development of a Cell-Based Binding Assay for the Detection of Neutralizing Antibodies Against a CD47xPD-L1 Bispecific Antibody. AAPS J 2023; 25:91. [PMID: 37740131 DOI: 10.1208/s12248-023-00856-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/23/2023] [Indexed: 09/24/2023] Open
Abstract
PF-07257876 is a bispecific antibody being developed for the treatment of certain advanced or metastatic solid tumors. To support clinical development of PF-07257876, neutralizing antibody (NAb) assays were developed as part of a tiered immunogenicity testing approach. Because PF-07257876 targets both CD47 and PD-L1, determination of domain specificity of a NAb response may provide additional insight relating to PK, efficacy, and safety. Due to limitations of functional cell systems, two cell-based binding assays were developed using electrochemiluminescence to detect domain-specific NAb. While both NAb assays utilized a cell-based binding approach and shared certain requirements, such as sensitivity and tolerance to potentially interfering substances, the development of each assay faced unique challenges. Among the hurdles encountered, achieving drug tolerance while preserving domain specificity for CD47 proved particularly challenging. Consequently, a sample pretreatment procedure to isolate NAb from potentially interfering substances was necessary. The sample pretreatment procedure developed was based on a bead-extraction and acid dissociation (BEAD) approach. However, the use of the standard BEAD approach with whole drug to capture NAb resulted in loss of NAb detection under certain circumstances. Specifically, mock samples containing a mixture of NAb positive controls against both binding domains of the bispecific antibody produced false-negative results in the cell-based binding assay. An adaptation made to the standard BEAD approach restored domain-specific NAb detection, while also contributing to an assay sensitivity of 1 µg/mL in the presence of a clinically relevant drug tolerance level of up to 400 µg/mL.
Collapse
Affiliation(s)
- Michael Luong
- BioMedicine Design, Pfizer, Inc., 1 Burtt Road, Andover, Massachusetts, 01810, USA.
| | - Ying Wang
- BioMedicine Design, Pfizer, Inc., 1 Burtt Road, Andover, Massachusetts, 01810, USA
| | - Brianna B Donnelly
- Janssen R&D, Lower Gwynedd Township, Montgomery County, Pennsylvania, USA
| | - Christopher Lepsy
- BioMedicine Design, Pfizer, Inc., 1 Burtt Road, Andover, Massachusetts, 01810, USA
| |
Collapse
|
18
|
Donley J, Jani D, Zhu T, Xiang Y, Gorovits B, Arkin S. Evolution of Antidrug Antibody Assays During the Development of Anti-Tissue Factor Pathway Inhibitor Monoclonal Antibody Marstacimab. AAPS J 2023; 25:84. [PMID: 37610502 DOI: 10.1208/s12248-023-00847-w] [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: 12/06/2022] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Tissue factor pathway inhibitor (TFPI) is an endogenous inhibitor of the extrinsic coagulation pathway. In patients with hemophilia A or B, inhibition of TFPI is an alternative therapeutic approach that augments the extrinsic coagulation pathway. Marstacimab is an investigational fully human monoclonal antibody that binds and neutralizes TFPI and is being evaluated as a prophylactic treatment to prevent or reduce the frequency of bleeding episodes in patients with severe hemophilia A or B, with or without inhibitors (antibodies against coagulation factors). However, the efficacy, safety, and pharmacokinetics of marstacimab may be affected by the induction of antidrug antibody (ADA) responses. Here, we describe the evolution and validation of three quasi-quantitative electrochemiluminescence-based methods to detect marstacimab ADAs, starting from their use in a first-in-human phase 1 study to their use in phase 2 and 3 clinical studies of patients with severe hemophilia. For all three methods, validation criteria evaluated the performance of the assays in screening and confirmatory cut points, precision, selectivity, drug tolerance, target interference, and stability. Additional criteria for validation were dilution linearity (Methods 1 and 2) and low positive control concentration, prozone effect, plate homogeneity, and robustness (Method 3). The three methods met validation criteria and are a potentially valuable tool in detecting the induction of marstacimab ADAs during treatment in patients with hemophilia.
Collapse
Affiliation(s)
- Jean Donley
- Pfizer Biomedicine Design, Andover, Massachusetts, USA
| | | | - Tong Zhu
- Pfizer Inc, Cambridge, Massachusetts, USA
| | - Yuhong Xiang
- Pfizer Biomedicine Design, Andover, Massachusetts, USA
| | | | - Steven Arkin
- Rare Disease Research Unit, Pfizer Worldwide Research & Development, 610 Main St., 2nd Floor, Cambridge, Massachusetts, 02139, USA.
| |
Collapse
|
19
|
Yin Z, Guerrero J, Melendez R, Andrews B, Peng K. Development of a Cell-based Neutralizing Antibody Assay for Zinpentraxin Alfa: Challenges and Mitigation Strategies. AAPS J 2023; 25:75. [PMID: 37468730 DOI: 10.1208/s12248-023-00841-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/01/2023] [Indexed: 07/21/2023] Open
Abstract
Therapeutic protein drugs can potentially induce immune responses in patients and result in the production of anti-drug antibodies (ADAs), including a subset of ADAs called neutralizing antibodies (NAbs) that might cause loss of efficacy by inhibiting clinical activities of the drug. Herein, we describe the unique challenges encountered during the development of a fit-for-purpose cell-based NAb assay for a new protein modality, zinpentraxin alfa, including our strategies for assay design to overcome various matrix interferences and improve assay drug tolerance. We demonstrated that a typical biotin-drug extraction with acid dissociation (BEAD) approach alone was not sufficient to eliminate matrix interferences in this assay. Instead, the combination of the BEAD and ZebaTM spin size exclusion plate (SEP) was required to achieve the desirable assay performance. We also demonstrated that appropriate acidic buffers were critical in sample pretreatment to improve assay drug tolerance, which not only dissociated the drug/NAb immune complex but also effectively and irreversibly denatured the free drug. The final assay performed well with confirmed assay robustness and suitability for the clinical applications.
Collapse
Affiliation(s)
- Zhaojun Yin
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Joyce Guerrero
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Rachel Melendez
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Ben Andrews
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Kun Peng
- BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| |
Collapse
|
20
|
Myler H, Pedras-Vasconcelos J, Lester T, Civoli F, Xu W, Wu B, Vainshtein I, Luo L, Hassanein M, Liu S, Ramaswamy SS, Mora J, Pennucci J, McCush F, Lavelle A, Jani D, Ambakhutwala A, Baltrukonis D, Barker B, Carmean R, Chung S, Dai S, DeWall S, Dholakiya SL, Dodge R, Finco D, Yan H, Hays A, Hu Z, Inzano C, Kamen L, Lai CH, Meyer E, Nelson R, Paudel A, Phillips K, Poupart ME, Qu Q, Abhari MR, Ryding J, Sheldon C, Spriggs F, Warrino D, Wu Y, Yang L, Pasas-Farmer S. Neutralizing Antibody Validation Testing and Reporting Harmonization. AAPS J 2023; 25:69. [PMID: 37421491 DOI: 10.1208/s12248-023-00830-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/05/2023] [Indexed: 07/10/2023] Open
Abstract
Evolving immunogenicity assay performance expectations and a lack of harmonized neutralizing antibody validation testing and reporting tools have resulted in significant time spent by health authorities and sponsors on resolving filing queries. A team of experts within the American Association of Pharmaceutical Scientists' Therapeutic Product Immunogenicity Community across industry and the Food and Drug Administration addressed challenges unique to cell-based and non-cell-based neutralizing antibody assays. Harmonization of validation expectations and data reporting will facilitate filings to health authorities and are described in this manuscript. This team provides validation testing and reporting strategies and tools for the following assessments: (1) format selection; (2) cut point; (3) assay acceptance criteria; (4) control precision; (5) sensitivity including positive control selection and performance tracking; (6) negative control selection; (7) selectivity/specificity including matrix interference, hemolysis, lipemia, bilirubin, concomitant medications, and structurally similar analytes; (8) drug tolerance; (9) target tolerance; (10) sample stability; and (11) assay robustness.
Collapse
Affiliation(s)
- Heather Myler
- Bioanalytical and Biomarker Science and Technologies, Takeda, Cambridge, MA, 02139, USA.
- 25 Creekview Ln, Yardley, Pennsylvania, 19067, USA.
| | - João Pedras-Vasconcelos
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Todd Lester
- BioAgilytix Labs, Durham, North Carolina, 27713, USA
| | - Francesca Civoli
- Coherus BioSciences, 333 Twin Dolphin Drive, Redwood City, California, 94065, USA
| | - Weifeng Xu
- Regulated Global Bioanalytics, Merck & Co., Inc, Rahway, New Jersey, 07065, USA
| | - Bonnie Wu
- Bioanalytical Discovery and Development Sciences, Janssen Research and Development, Spring House, Pennsylvania, 19477, USA
| | - Inna Vainshtein
- Discovery and Translational Research, Exelixis, 1851 Harbor Bay Pkwy, Alameda, California, 94502, USA
| | - Linlin Luo
- Regulated Global Bioanalytics, Merck & Co., Inc, Rahway, New Jersey, 07065, USA
| | - Mohamed Hassanein
- Early Clinical Development, Precision Medicine, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Susana Liu
- Global Product Development, , Pfizer Inc, 17300 Trans Canada Hwy, Kirkland, Quebec, Canada
| | - Swarna Suba Ramaswamy
- Regulated Bioanalysis Department, B2S Life Sciences, 97 East Monroe Street, Franklin, Indiana, 46131, USA
| | - Johanna Mora
- Non-Clinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jason Pennucci
- Quantitative Bioanalytics, Moderna, 200 Technology Square, Cambridge, Massachusetts, 02139, USA
| | - Fred McCush
- Clinical Pharmacology, Pfizer Inc, Eastern Point Road, Groton, CT, 06379, USA
| | - Amy Lavelle
- Bioanalytical Lab, PPD Clinical Research, , 2244 Dabney Road, Richmond, Virginia, 23230-3323, USA
| | - Darshana Jani
- Preclinical and Clinical Bioanalysis, , Moderna Tx, 200 Technology Square, Cambridge, Massachusetts, 02142, USA
| | - Angela Ambakhutwala
- Immunology Sciences, Kriya Therapeutics, 4105 Hopson Road, Morrisville, North Carolina, 27560, USA
| | - Daniel Baltrukonis
- Clinical Pharmacology, Pfizer Inc, Eastern Point Road, Groton, CT, 06379, USA
| | - Breann Barker
- Drug Metabolism and Biopharmaceuticals, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware, 19803, USA
| | - Rebecca Carmean
- Bioanalytical Lab, PPD Clinical Research, , 2244 Dabney Road, Richmond, Virginia, 23230-3323, USA
| | - Shan Chung
- Department of BioAnalytical Sciences, Genentech Inc, South San Francisco, California, USA
| | - Sheng Dai
- Quantitative Clinical Pharmacology & Translational Sciences (QCP), Daiichi Sankyo, Inc, 211 Mt. Airy Road, Basking Ridge, New Jersey, 07920, USA
| | - Stephen DeWall
- Bioanalytical Sciences, Scholar Rock, 301 Binney Street, 3rd Floor, Cambridge, Massachusetts, 02142-1071, USA
| | - Sanjay L Dholakiya
- Non-Clinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Robert Dodge
- Novartis Institutes for BioMedical Research, 1 Health Plaza, East Hanover, New Jersey, 07936, USA
| | - Deborah Finco
- Deborah Finco Consulting LLC, 101 Prospect Hill Road, Groton, Connecticut, 06340, USA
| | - Haoheng Yan
- Global Regulatory Affairs, Shanghai Henlius Biotech. Inc, 430 N. McCarthy Blvd, Milpitas, California, 95035, USA
| | - Amanda Hays
- BioAgilytix Labs, Durham, North Carolina, 27713, USA
| | - Zheng Hu
- Translation Safety & Bioanalytical Science, Amgen Inc, Thousand Oaks, California, 91360, USA
| | - Cynthia Inzano
- Non-Clinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Lynn Kamen
- BioAgilytix Labs, Durham, North Carolina, 27713, USA
| | - Ching-Ha Lai
- Bioanalytical Sciences, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Erik Meyer
- Immunogenicity, GSK Pharmaceuticals, 1250 South Collegeville Road, Collegeville, Pennsylvania, 19426, USA
| | - Robert Nelson
- Bioanalytical Services, Labcorp Drug Development, Otley Road, Harrogate, HG3 1PY, UK
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering GmbH (RCPE), Inffeldgasse 13, 8010, Graz, Austria
| | - Kelli Phillips
- Clinical Pharmacology, Pfizer Inc, Eastern Point Road, Groton, CT, 06379, USA
| | - Marie-Eve Poupart
- Immunology, Charles River Laboratories, Montreal ULC, Transcanada Highway, Senneville, Quebec, 22022, Canada
| | - Qiang Qu
- Quantitative Bioanalytics, Moderna, 200 Technology Square, Cambridge, Massachusetts, 02139, USA
| | - Mohsen Rajabi Abhari
- Office of Clinical PharmacologyOffice of Translational SciencesCenter for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Janka Ryding
- Bioanalysis-Biologics, Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | - Curtis Sheldon
- Technical Operations, , IVERIC Bio, Inc, 8 Sylvan Way, Parsippany, New Jersey, 07054, USA
| | - Franklin Spriggs
- Spriggs Bioanalytical Consulting LLC, 15632 W 83rd Terrace, Lenexa, Kansas, 66219, USA
| | - Dominic Warrino
- Bioanalytical and Biomarker Services, KCAS, 10830 S Clay Blair Blvd, Olathe, Kansas, 66061, USA
| | - Yuling Wu
- Integrated Bioanalysis, , AstraZeneca, Gaithersburg, Maryland, 20878, USA
| | - Lin Yang
- Bioanalytical Sciences, REGENXBIO Inc, 9804 Medical Center Drive, Rockville, Maryland, 20850, USA
| | | |
Collapse
|
21
|
Pöhler A, Jany C, Butzer J, Bach T, Opolka-Hoffmann E, Staack RF, Jordan G. High ionic strength dissociation assay reduces dimeric target interference in immunogenicity testing. Bioanalysis 2023; 15:823-832. [PMID: 37326333 DOI: 10.4155/bio-2023-0082] [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] [Indexed: 06/17/2023] Open
Abstract
Aim: The presence of di-/multi-meric forms of soluble target in biological samples can interfere in anti-drug antibody (ADA) assays, leading to increased background values and potentially false positivity. The authors investigated the use of the high ionic strength dissociation assay (HISDA) to reduce target interference in two different ADA assays. Results: Interference caused by homodimeric FAP was successfully eliminated to enable cut point determination after applying HISDA. Biochemical experiments confirmed the dissociation of homodimeric FAP after treatment with high ionic strength conditions. Conclusion: HISDA is a promising approach to simultaneously achieve high drug tolerance and reduced interference by noncovalently bound dimeric target molecules in ADA assays without extensive optimization, which is particularly advantageous in routine use.
Collapse
Affiliation(s)
- Alexander Pöhler
- Roche Pharma Research & Early Development (pRED), Pharmaceutical Sciences, Bioanalysis & Biomarkers, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| | - Cordula Jany
- Roche Pharma Research & Early Development (pRED), Pharmaceutical Sciences, Bioanalysis & Biomarkers, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| | - Joachim Butzer
- Roche Pharma Research & Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| | - Thomas Bach
- Roche Pharma Research & Early Development (pRED), Pharmaceutical Sciences, Bioanalysis & Biomarkers, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| | - Eugenia Opolka-Hoffmann
- Roche Pharma Research & Early Development (pRED), Pharmaceutical Sciences, Bioanalysis & Biomarkers, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| | - Roland F Staack
- Roche Pharma Research & Early Development (pRED), Pharmaceutical Sciences, Bioanalysis & Biomarkers, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| | - Gregor Jordan
- Roche Pharma Research & Early Development (pRED), Pharmaceutical Sciences, Bioanalysis & Biomarkers, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg, 82377, Germany
| |
Collapse
|
22
|
Du J, Yang Y, Zhu L, Wang S, Yu C, Liu C, Long C, Chen B, Xu G, Zou L, Wang L. Method validation of a bridging immunoassay in combination with acid-dissociation and bead treatment for detection of anti-drug antibody. Heliyon 2023; 9:e13999. [PMID: 36915535 PMCID: PMC10006523 DOI: 10.1016/j.heliyon.2023.e13999] [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: 09/18/2022] [Revised: 01/20/2023] [Accepted: 02/17/2023] [Indexed: 03/03/2023] Open
Abstract
Anti-drug antibody (ADA) positivity is correlated with disease relapse risk when treated with monoclonal antibody (mAb) therapeutics. ADA evaluation can assist with interpreting pharmacokinetic, pharmacological, and toxicology results. Here, we established an ADA assay based on two steps of acid dissociation combined with a bridging immunoassay to provide a comprehensive validation strategy. The three-tiered sample analysis process included screening, confirmation, and titration assays using therapeutic HLX26 (targeting lymphocyte activation gene-3 [LAG-3]) as an example. The cut points were determined by testing 50 individual normal human serum samples, including screening cut point (SCP) (SNR: 1.08), confirmatory cut point (CCP) (% inhibition: 12.65), and titration cut point (TCP) (sample-to-noise ratio [SNR]: 1.17). The assay sensitivity, low positive control (LPC), and high positive control (HPC) titer acceptable range were also set up as 33.0 ng/mL, 41.0 ng/mL, and 320-1280, respectively. After full validation, both the intra-assay and inter-assay precision testing passed with coefficient of variations (CVs) < 20%. The assay enabled excellent drug tolerance up to 768.0 μg/mL at the HPC level and 291.0 μg/mL at the LPC level, while the tolerance of target interference was up to 74.0 ng/mL of soluble LAG3. Moreover, no false-positive results were observed in the presence of 5% hemolyzed serum samples and 150 mg/dL of triglyceride in the serum samples, no hook effect was observed, and the stability performed normally under room temperature for 24 h, 2-8 °C for 7 d, and six freeze/thaw cycles. In summary, this ADA assay is feasible and could be used for evaluating the immunogenicity of HLX26 in clinical trials.
Collapse
Affiliation(s)
- Jialiang Du
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yalan Yang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | | | - Shaoyi Wang
- Shanghai Henlius Biotech Inc, Shanghai, China
| | - Chuanfei Yu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | - Chunyu Liu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | - Caifeng Long
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | - Baowen Chen
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | - Gangling Xu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
| | - Linglong Zou
- Shanghai Henlius Biotech Inc, Shanghai, China
- Corresponding author. 5155# GUANGFULIN Road, Shanghai Henlius Biotech Inc, Shanghai, 201616, China.
| | - Lan Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, Beijing, China
- Corresponding author. Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Division of Monoclonal Antibody Products, National Institutes for Food and Drug Control, 31# HUATUO Road, Beijing, 102629, China.
| |
Collapse
|
23
|
Fathallah AM, Oldfield P, Fiedler‐Kelly J, Ramadan A. Immunogenicity Considerations for Therapeutic Modalities Used in Rare Diseases. J Clin Pharmacol 2022; 62 Suppl 2:S110-S118. [DOI: 10.1002/jcph.2166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/30/2022] [Indexed: 12/04/2022]
Affiliation(s)
| | - Philip Oldfield
- Scientific and Regulatory Consultant Greater Montreal Metropolitan Canada
| | | | | |
Collapse
|
24
|
Hum RM, Ho P, Nair N, Jani M, Morgan AW, Isaacs JD, Wilson AG, Hyrich KL, Plant D, Barton A. Non-Trough adalimumab and certolizumab drug levels associated with a therapeutic EULAR response in adherent patients with rheumatoid arthritis. Rheumatology (Oxford) 2022:6747167. [PMID: 36190343 DOI: 10.1093/rheumatology/keac564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/03/2022] [Accepted: 09/17/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Interventions aimed at increasing tumour-necrosis factor-alpha inhibitor serum drug levels (SDLs) may improve treatment response; however, previous studies suggesting SDL cut-offs have not accounted for treatment adherence. The aim of this study was to establish the relationship between Adalimumab/Certolizumab SDLs and EULAR good vs non/moderate response, and to define SDL cut-offs associated with good response in fully adherent patients. METHODS In a prospective observational study, 475 patients with RA were treated with Certolizumab (n = 192) or Adalimumab (n = 283). At baseline, 3/6/12-months patients had DAS28, self-reported treatment adherence, and SDLs measured. Fully adherent patients were analysed as a subgroup. Follow-up data at 3/6/12-months was analysed separately. Median SDLs were compared in good vs non/moderate response, and receiver-operator characteristics curves (ROC) were used to establish cut-off SDLs. RESULTS Fully adherent good responders had significantly higher median Adalimumab/Certolizumab SDLs compared with non/moderate responders (p= 0·04 and p= 0·0005, respectively). ROC analysis reported 3-month non-trough Adalimumab SDLs discriminated good vs non/moderate response with an AUC of 0·63 (95% CI 0·52-0·75), with a cut-off of 7·5mg/l being 39·1% specific, and 80·9% sensitive. Similarly, 3-month non-trough Certolizumab SDLs discriminated good vs non/moderate response with an AUC of 0·65 (95% CI 0·51-0·78), with a cut-off of 26·0mg/l being 43·9% specific, and 77·8% sensitive. CONCLUSION In fully adherent patients, higher SDLs are detected in good responders suggesting that interventions to improve SDLs such as encouraging adherence could improve treatment response. 3-month non-trough SDL cut-offs of 7·5mg/l for Adalimumab and 26·0mg/l for Certolizumab may be useful in clinical practice.
Collapse
Affiliation(s)
- Ryan M Hum
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom.,The Kellgren Centre for Rheumatology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Pauline Ho
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom.,The Kellgren Centre for Rheumatology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Nisha Nair
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Meghna Jani
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Ann W Morgan
- NIHR Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Leeds, Leeds, United Kingdom
| | - John D Isaacs
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Anthony G Wilson
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Kimme L Hyrich
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom.,The Kellgren Centre for Rheumatology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Darren Plant
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Anne Barton
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom.,The Kellgren Centre for Rheumatology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | | |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Novel isoelectric target depletion (ITaD) protocol reduces the need for specific reagents for immunogenicity testing. Bioanalysis 2022; 14:725-735. [PMID: 35642540 DOI: 10.4155/bio-2022-0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: The development of immunogenicity assays for clinical drug candidates targeting soluble proteins is challenging when the soluble target might produce either false-positive or false-negative signals in bridging anti-drug antibody screening assays. A generic soluble target removal protocol that uses a pH-dependent depletion was evaluated. Results: An anti-drug antibody bridging assay with a pH-dependent soluble target depletion step was successfully developed. Endogenous target levels of ∼600 nM could be depleted below 8 pM. The assay was highly drug tolerant and met regulatory requirements. Conclusion: A reagent-independent target depletion protocol can be used for immunogenicity testing in the presence of a soluble target. The generic protocol circumvents common depletion or masking protocols.
Collapse
|
28
|
Kroenke MA, Barger TE, Hu J, Miller MJ, Kalenian K, He L, Hsu H, Bartley Y, Chow VFS, Teixeira Dos Santos MC, Sullivan BA, Cheng LE, Parnes JR, Padaki R, Kuhns S, Mytych DT. Immune Complex Formation Is Associated With Loss of Tolerance and an Antibody Response to Both Drug and Target. Front Immunol 2022; 12:782788. [PMID: 34970265 PMCID: PMC8712722 DOI: 10.3389/fimmu.2021.782788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/23/2021] [Indexed: 11/15/2022] Open
Abstract
AMG 966 is a bi-specific, heteroimmunoglobulin molecule that binds both tumor necrosis factor alpha (TNFα) and TNF-like ligand 1A (TL1A). In a first-in-human clinical study in healthy volunteers, AMG 966 elicited anti-drug antibodies (ADA) in 53 of 54 subjects (98.1%), despite a paucity of T cell epitopes observed in T cell assays. ADA were neutralizing and bound to all domains of AMG 966. Development of ADA correlated with loss of exposure. In vitro studies demonstrated that at certain drug-to-target ratios, AMG 966 forms large immune complexes with TNFα and TL1A, partially restoring the ability of the aglycosylated Fc domain to bind FcγRIa and FcγRIIa, leading to the formation of ADA. In addition to ADA against AMG 966, antibodies to endogenous TNFα were also detected in the sera of subjects dosed with AMG 966. This suggests that the formation of immune complexes between a therapeutic and target can cause loss of tolerance and elicit an antibody response against the target.
Collapse
Affiliation(s)
- Mark A Kroenke
- Clinical Immunology, Translational Medicine, Amgen, Thousand Oaks, CA, United States
| | - Troy E Barger
- Translational Safety & Bioanalytical Sciences, Amgen, Thousand Oaks, CA, United States
| | - Jenny Hu
- Translational Safety & Bioanalytical Sciences, Amgen, Thousand Oaks, CA, United States
| | - Mieke Jill Miller
- Translational Safety & Bioanalytical Sciences, Amgen, Thousand Oaks, CA, United States
| | - Kevin Kalenian
- Process Development, Attribute Sciences, Amgen, Thousand Oaks, CA, United States
| | - Lidong He
- Process Development, Attribute Sciences, Amgen, Thousand Oaks, CA, United States
| | - Hailing Hsu
- Inflammation Research, Amgen, Thousand Oaks, CA, United States
| | | | - Vincent Fung-Sing Chow
- Clinical Pharmacology, Modeling and Simulation, Translational Medicine, Amgen, Thousand Oaks, CA, United States
| | | | - Barbara A Sullivan
- Clinical Biomarkers and Diagnostics, Translational Medicine, Amgen, Thousand Oaks, CA, United States
| | - Laurence E Cheng
- Early Development, Translational Medicine, Amgen, Thousand Oaks, CA, United States
| | - Jane R Parnes
- Early Development, Translational Medicine, Amgen, Thousand Oaks, CA, United States
| | - Rupa Padaki
- Process Development, Attribute Sciences, Amgen, Thousand Oaks, CA, United States
| | - Scott Kuhns
- Process Development, Attribute Sciences, Amgen, Thousand Oaks, CA, United States
| | - Daniel T Mytych
- Clinical Immunology, Translational Medicine, Amgen, Thousand Oaks, CA, United States
| |
Collapse
|
29
|
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
|
30
|
Factors associated with reduced infliximab exposure in the treatment of pediatric autoimmune disorders: a cross-sectional prospective convenience sampling study. Pediatr Rheumatol Online J 2021; 19:62. [PMID: 33933127 PMCID: PMC8088679 DOI: 10.1186/s12969-021-00548-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inadequate systemic exposure to infliximab (IFX) is associated with treatment failure. This work evaluated factors associated with reduced IFX exposure in children with autoimmune disorders requiring IFX therapy. METHODS In this single-center cross-sectional prospective study IFX trough concentrations and anti-drug antibodies (ADAs) were measured in serum from children diagnosed with inflammatory bowel disease (IBD) (n = 73), juvenile idiopathic arthritis (JIA) (n = 16), or uveitis (n = 8) receiving maintenance IFX infusions at an outpatient infusion clinic in a tertiary academic pediatric hospital. IFX concentrations in combination with population pharmacokinetic modeling were used to estimate IFX clearance. Patient demographic and clinical data were collected by chart review and evaluated for their relationship with IFX clearance. RESULTS IFX trough concentrations ranged from 0 to > 40 μg/mL and were 3-fold lower in children with IBD compared to children with JIA (p = 0.0002) or uveitis (p = 0.001). Children with IBD were found to receive lower IFX doses with longer dosing intervals, resulting in dose intensities (mg/kg/day) that were 2-fold lower compared to children with JIA (p = 0.0002) or uveitis (p = 0.02). Use of population pharmacokinetic analysis to normalize for variation in dosing practices demonstrated that increased IFX clearance was associated with ADA positivity (p = 0.004), male gender (p = 0.02), elevated erythrocyte sedimentation rate (ESR) (p = 0.02), elevated c-reactive protein (CRP) (p = 0.001), reduced serum albumin concentrations (p = 0.0005), and increased disease activity in JIA (p = 0.009) and IBD (p ≤ 0.08). No significant relationship between diagnosis and underlying differences in IFX clearance was observed. Multivariable analysis by covariate population pharmacokinetic modeling confirmed increased IFX clearance to be associated with anti-IFX antibody positivity, increased ESR, and reduced serum albumin concentrations. CONCLUSIONS Enhanced IFX clearance is associated with immunogenicity and inflammatory burden across autoimmune disorders. Higher systemic IFX exposures observed in children with rheumatologic disorders are driven primarily by provider drug dose and interval selection, rather than differences in IFX pharmacokinetics across diagnoses. Despite maintenance IFX dosing at or above the standard recommended range for IBD (i.e., 5 mg/kg every 8 weeks), the dosing intensity used in the treatment of IBD is notably lower than dosing intensities used to treat JIA and uveitis, and may place some children with IBD at risk for suboptimal maintenance IFX exposures necessary for treatment response.
Collapse
|
31
|
Heron CE, Ghamrawi RI, Balogh EA, Feldman SR. Immunogenicity of Biologic and Biosimilar Therapies for Psoriasis and Impact of Novel Immunoassays for Immunogenicity Detection. Am J Clin Dermatol 2021; 22:221-231. [PMID: 33169802 DOI: 10.1007/s40257-020-00569-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 12/21/2022]
Abstract
Anti-drug antibodies (ADAs) may develop against originator biologic and biosimilar therapies used for the treatment of psoriasis and may be the cause of initial therapeutic non-response or diminished therapeutic response over time. Comparing immunogenicity between therapeutic agents is challenging owing to the variation in assays used for detection, among other reasons. Using the results of a PubMed search for psoriasis clinical trials disclosing the rates of ADAs for originator biologic and biosimilar therapies approved for the treatment of psoriasis within the last 5 years, this review discusses the rates and potential clinical impact of ADA formation in patients with psoriasis managed with originator biologic and biosimilar therapies, along with novel methods of ADA testing. Anti-drug antibodies are detectable in all biologic and biosimilar therapies approved for the treatment of psoriasis in the last 5 years, and the effect of ADAs on clinical response varies by agent. Novel immunoassays used for the detection of ADAs may have increased sensitivity compared with traditional assays, although the increased rate of detection may not correlate with decreased clinical response and the decision to test for the presence of ADAs may vary from patient to patient. Though ADA formation seems ubiquitous with the use of biologic agents for the treatment of psoriasis, the increased rates of ADAs detected by novel immunoassays may not necessarily correlate with decreased treatment efficacy.
Collapse
|
32
|
Cohen JM, Ning X, Kessler Y, Rasamoelisolo M, Campos VR, Seminerio MJ, Krasenbaum LJ, Shen H, Stratton J. Immunogenicity of biologic therapies for migraine: a review of current evidence. J Headache Pain 2021; 22:3. [PMID: 33413094 PMCID: PMC7791637 DOI: 10.1186/s10194-020-01211-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/12/2020] [Indexed: 12/16/2022] Open
Abstract
Background Monoclonal antibodies (mAbs) targeting the calcitonin gene-related peptide (CGRP) pathway have been shown to be effective in migraine prevention. Eptinezumab, erenumab, fremanezumab, and galcanezumb have shown efficacy in clinical trials along with favorable safety and tolerability profiles. Although erenumab is a human mAb and the others have been humanized to varying degrees, they all have the capacity to provoke immune reactions. The present review article aims to discuss the current relationship between mAbs targeting the CGRP pathway (CGRP mAbs) and immunogenicity and their potential clinical implications. Findings The incidence of patients developing anti-drug antibodies (ADAs), their titer, and clinical significance are highly variable and depend on a variety of different drug and patient factors. Neutralizing ADAs (NAbs) bind to and inhibit or reduce the pharmacologic activity of the biologic drug molecule, whereas non-neutralizing antibodies (Non-NAbs) bind to the biologic drug molecule without affecting pharmacologic activity in an in vitro test, although pharmacokinetics and drug clearance may be affected. A direct comparison of immunogenicity data across clinical trials with different biologics is not possible due to a lack of standardized assays. Several phase 2, phase 3, and long-term studies evaluating CGRP mAbs for migraine prevention have reported immunogenicity data (5 studies each for eptinezumab, erenumab, fremanezumab, and galcanezumab). Across these studies, prevalence of ADAs varied, ranging from < 1% to ~ 18%. Neutralizing ADAs were slightly less common, with a prevalence ranging from 0 to 12%. Adverse events related to ADA formation were rare. Conclusions As more CGRP mAb studies are conducted and more long-term follow-up data become available, evidence is increasing that immunogenicity rates of biologic therapies for migraine are low, and adverse events related to ADAs are rare. Taken together, these results add to the growing body of evidence for the safety and tolerability of this class of migraine medications.
Collapse
|
33
|
Strand V, Goncalves J, Isaacs JD. Immunogenicity of biologic agents in rheumatology. Nat Rev Rheumatol 2020; 17:81-97. [PMID: 33318665 DOI: 10.1038/s41584-020-00540-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Biologic agents have become a core component of therapeutic strategies for many inflammatory rheumatic diseases. However, perhaps reflecting the specificity and generally high affinity of biologic agents, these therapeutics have been used by rheumatologists with less consideration of their pharmacokinetics than that of conventional synthetic DMARDs. Immunogenicity was recognized as a potential limitation to the use of biologic agents at an early stage in their development, although regulatory guidance was relatively limited and assays to measure immunogenicity were less sophisticated than today. The advent of biosimilars has sparked a renewed interest in immunogenicity that has resulted in the development of increasingly sensitive assays, an enhanced appreciation of the pharmacokinetic consequences of immunogenicity and the development of comprehensive and specific guidance from regulatory authorities. As a result, rheumatologists have a greatly improved understanding of the field in general, including the factors responsible for immunogenicity, its potential clinical consequences and the implications for everyday treatment. In some specialties, immunogenicity testing is becoming a part of routine clinical management, but definitive evidence of its cost-effectiveness in rheumatology is awaited.
Collapse
Affiliation(s)
- Vibeke Strand
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA.
| | - Joao Goncalves
- Research Institute for Medicines (iMed), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
34
|
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
|
35
|
Martinez JM, Hindiyeh N, Anglin G, Kalidas K, Hodsdon ME, Kielbasa W, Moser BA, Pearlman EM, Garces S. Assessment of immunogenicity from galcanezumab phase 3 trials in patients with episodic or chronic migraine. Cephalalgia 2020; 40:978-989. [PMID: 32340471 PMCID: PMC7469706 DOI: 10.1177/0333102420920642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/23/2019] [Accepted: 03/26/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND This analysis characterizes the immunogenicity profile of galcanezumab, a humanized monoclonal antibody that selectively binds calcitonin gene-related peptide and inhibits its activity, in phase 3 migraine trials. METHODS Immunogenicity data were analyzed from baseline and double-blind, placebo-controlled phases of the 3-month chronic migraine study REGAIN, the 6-month episodic migraine studies EVOLVE-1 and EVOLVE-2, and from baseline and open-label phases of the 12-month chronic and episodic migraine Study CGAJ. The incidence of baseline antidrug antibodies, treatment-emergent antidrug antibodies, neutralizing antidrug antibodies, and the effect of antidrug antibody titer on pharmacokinetics and pharmacodynamics were assessed. The relationship between antidrug antibody status and efficacy was explored using average change in monthly migraine headache days. Safety analyses assessed the potential relationship between treatment-emergent antidrug antibodies and hypersensitivity events or adverse events related to injection sites. FINDINGS Across studies, 5.9-11.2% of patients had baseline antidrug antibodies. The incidence of treatment-emergent antidrug antibodies was 2.6-12.4% in the galcanezumab group and 0.5-1.7% in the placebo group. The majority of treatment-emergent antidrug antibodies were detected approximately 3-6 months after first study drug dose. Overall, the observed antidrug antibody titer did not impact galcanezumab concentrations, calcitonin gene-related peptide concentrations, or galcanezumab efficacy. There was no evidence that hypersensitivity events or adverse events related to injection sites were mediated by treatment-emergent antidrug antibodies. INTERPRETATION These data showed that immunogenicity did not impact galcanezumab concentrations, calcitonin gene-related peptide concentrations, or the efficacy and hypersensitivity profile of galcanezumab in patients with migraine.
Collapse
|
36
|
Regulatory and strategic considerations for addressing immunogenicity and related responses in biopharmaceutical development programs. J Clin Transl Sci 2020; 4:547-555. [PMID: 33948231 PMCID: PMC8057416 DOI: 10.1017/cts.2020.493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The last three decades have seen the biotherapeutic drug market evolve from promising concept to market dominance in a range of clinical indications. This growth has been spurred by the success of established drug classes like monoclonal antibodies, but also by the introduction of biosimilars, and more recently, multiple novel cell and gene therapies. Biotherapeutic drug development presents many unique challenges, but unintended immune responses are among the most common reasons for program attrition. Anti-drug antibodies can impact the safety and efficacy of drug products, and related immune responses, like the cytokine release syndrome that occurred in the infamous TGN-1412 clinical trial, can be challenging to predict with nonclinical models. For this reason, it is important that development programs proceed with a scientifically grounded and measured approach to these responses. This process begins at the discovery stage with the application of “quality by design,” continues into the clinic with the development of quality assays and management strategies, and culminates in the effective presentation of this information in regulatory documents. This review provides an overview of some of the key strategic and regulatory considerations for biotherapeutics as they pertain to immunogenicity and related responses.
Collapse
|
37
|
Wang EQ, Bukowski JF, Yunis C, Shear CL, Ridker PM, Schwartz PF, Baltrukonis D. Assessing the Potential Risk of Cross-Reactivity Between Anti-Bococizumab Antibodies and Other Anti-PCSK9 Monoclonal Antibodies. BioDrugs 2020; 33:571-579. [PMID: 31529318 PMCID: PMC6790354 DOI: 10.1007/s40259-019-00375-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Anti-drug antibodies (ADAs) to bococizumab were detected in > 40% of subjects in the SPIRE lipid-lowering trials. The risk of cross-reactivity between anti-bococizumab antibodies and other approved anti-proprotein convertase subtilisin/kexin type-9 (PCSK9) monoclonal antibodies (mAbs) was investigated using a single-assay approach. METHODS Bococizumab immunogenicity was assessed in SPIRE-HR, a 52-week study. The highest ADA titer sample from each ADA-positive subject (n = 155) was tested in vitro for cross-reactivity to alirocumab and evolocumab using a novel ADA assay approach. Additional specificity tiers within the bococizumab ADA assay against each drug were validated using recombinant PCSK9 as a surrogate cross-reactive positive control. If the highest ADA titer sample showed cross-reactivity, additional samples from that subject were analyzed. Cross-reactivity was determined by the ability of alirocumab or evolocumab to inhibit the sample signal greater than or equal to the cross-reactivity cut-points. RESULTS ADAs were detected in 44.0% (155/352) of bococizumab-treated subjects, and 27.0% also developed neutralizing antibodies (NAbs). Median ADA and NAb titers ranged from 276 to 526 and 8 to 12 over the course of the study, respectively. From 155 ADA-positive subjects tested for cross-reactivity, one (0.6%) subject showed weak cross-reactivity to both alirocumab and evolocumab. This cross-reactivity signal was transient (from Days 337 to 373) and undetectable at the last ADA-positive timepoint (Day 407). CONCLUSION A novel, single-assay approach was validated to assess the potential cross-reactivity of anti-bococizumab antibodies to alirocumab and evolocumab. In subjects who developed ADAs to bococizumab, the likelihood of clinically relevant cross-reactivity to marketed anti-PCSK9 mAbs is remote, based on the low frequency of cross-reactivity observed, which was weak in signal inhibition and transient in nature. CLINICAL TRIAL REGISTRATION The SPIRE-HR study is registered on ClinicalTrials.gov under the identifier NCT01968954.
Collapse
Affiliation(s)
- Ellen Q Wang
- Clinical Pharmacology, Global Product Development, Pfizer Inc, New York, NY, USA.
| | | | - Carla Yunis
- Internal Medicine, Global Product Development, Pfizer Inc, New York, NY, USA
| | | | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Daniel Baltrukonis
- Clinical Pharmacology, Global Product Development, Pfizer Inc, Groton, CT, USA
| |
Collapse
|
38
|
Bharadwaj P, Riekofski C, Lin S, Seaman MS, Garber DA, Montefiori D, Sarzotti-Kelsoe M, Ackerman ME, Weiner JA. Implementation of a three-tiered approach to identify and characterize anti-drug antibodies raised against HIV-specific broadly neutralizing antibodies. J Immunol Methods 2020; 479:112764. [PMID: 32070674 PMCID: PMC7103756 DOI: 10.1016/j.jim.2020.112764] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 01/10/2020] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
The ability to detect, quantify, and interrogate the properties of immune responses raised against biological therapeutics is not only important to our understanding of these molecules, but also to their success in the clinic. A tiered assay approach to identify the presence, specificity, and titer of anti-drug antibody (ADA) responses has been adopted as a gold standard by industry leaders, the FDA, and the EMA. In order to support pre-clinical and clinical trials, these assays must be standardized, and their performance sufficiently characterized to ensure the accuracy and reproducibility of results under relevant testing conditions. Here we present implementation of electrochemiluminiscence assays that fit into the tiered paradigm of ADA testing for five HIV broadly neutralizing antibodies (3BNC117, 3BNC117-LS, 10–1074, PGT121, and PGDM1400) in compliance with Good Clinical Laboratory practices. Assay sensitivities and matrix effects were evaluated and used to inform the development of positivity cut points. Once cut points were established, assay precision, specificity, free-drug tolerance, and robustness were defined. In all cases, assay characteristics met or surpassed recommendations set forth by the FDA. To further evaluate the performance of these assays and the tiered approach, samples from non-human primates that had received a subset of the five therapeutics were evaluated. In sum, this study reports qualification of a set of ADA assays available to the scientific community as pre-clinical and clinical trials of broadly HIV-neutralizing antibodies proceed, and a framework that is easily adapted as new drug products are advanced in the clinic.
Collapse
Affiliation(s)
- Pranay Bharadwaj
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | - Shu Lin
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David A Garber
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Marcella Sarzotti-Kelsoe
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA; Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
39
|
Zhong ZD, Jiang LL, Khandelwal P, Clarke AW, Bakhtiar R, Zou L. Development and Utility of an ELISA Method for Sensitive and Specific Detection of IgE Antidrug Antibodies. AAPS J 2020; 22:36. [PMID: 31997031 DOI: 10.1208/s12248-020-0413-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/27/2019] [Indexed: 12/16/2022] Open
Abstract
Biologics can potentially induce unwanted immune responses, leading to formation of antidrug antibodies (ADA) of various affinity, isotypes, and subclasses. Among them, antigen and drug-specific immunoglobulin E (IgE) antibodies have been reported to have potential correlation with hypersensitivity and anaphylaxis in particular. Recent regulatory guidance on immunogenicity testing has recommended the measurement of antigen-specific IgE antibodies for biologics with a reported high risk of anaphylaxis using assays with sensitivities in the high pg/mL to low ng/mL range. Nevertheless, IgE ADA remains challenging to detect due to their being the least abundant isotype in blood serum samples and the potential for interference in the bioanalytical methods due to high levels of endogenous immunoglobulin G (IgG) and immunoglobulin M (IgM) ADA, not to mention the nonspecific total serum IgE antibodies. Another challenge in developing IgE ADA assays is the need to create a surrogate drug-specific IgE antibody positive control to monitor the performance of the assay for the intended use. In this case study, utilizing a human IgE antidrug antibody positive control and a human IgE receptor as capture, an enzyme-linked immunosorbent assay (ELISA) method was developed for the measurement of IgE ADA, meeting the regulatory expectations, with excellent assay sensitivity, selectivity, specificity, and tolerance towards potential interference in serum samples. This assay format could be readily adapted and implemented to assess drug-specific IgE antibodies in the event of drug-related anaphylaxis in clinical and in nonclinical development programs.
Collapse
Affiliation(s)
- Zhandong Don Zhong
- Specialty Bioanalytics, Teva Pharmaceuticals Inc., West Chester, Pennsylvania, 19380, USA.
| | - Lynn L Jiang
- Specialty Bioanalytics, Teva Pharmaceuticals Inc., West Chester, Pennsylvania, 19380, USA
| | - Puneet Khandelwal
- Specialty Bioanalytics, Teva Pharmaceuticals Inc., West Chester, Pennsylvania, 19380, USA
| | - Adam W Clarke
- R&D, Biologics, Lead Antibody Discovery, Teva Pharmaceuticals, Sydney, NSW, Australia
| | - Ray Bakhtiar
- Specialty Bioanalytics, Teva Pharmaceuticals Inc., West Chester, Pennsylvania, 19380, USA
| | - Linglong Zou
- Biologics Assays & Technology, Teva Pharmaceuticals Inc., West Chester, Pennsylvania, 19380, USA
| |
Collapse
|
40
|
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
|
41
|
Wang Y, Luong M, Guadiz C, Zhang M, Gorovits B. Addressing soluble target interference in the development of a functional assay for the detection of neutralizing antibodies against a BCMA-CD3 bispecific antibody. J Immunol Methods 2019; 474:112642. [PMID: 31400410 DOI: 10.1016/j.jim.2019.112642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/28/2019] [Accepted: 08/06/2019] [Indexed: 10/26/2022]
Abstract
Proper evaluation of immunogenicity during clinical development of biotherapeutics is a major challenge to bioanalytical scientists, in part due to matrix interference in anti-drug antibody (ADA) and neutralizing antibody (NAB) assays. If not addressed, matrix interference could confound the immunogenicity assessment of a given biotherapeutic in clinical development. To support clinical development of a B cell maturation antigen (BCMA)-CD3 bispecific antibody, a cell-based NAB assay was developed as part of a tiered approach to evaluating the immunogenicity of the drug. The assay endpoint (T cell activation) was chosen based on its strong association with the mechanism of action of the drug. The BCMA-CD3 bispecific antibody activates T cells through simultaneous binding of CD3 on T cells and BCMA on target cells. In this system, T cell activation was assessed through the measurement of luciferase activity in an engineered Jurkat cell line. In the presence of NAB, the degree of T cell activation measured by the amount of luciferase activity can be reduced. During method development, soluble BCMA (sBCMA) interference in the NAB assay was apparent. The binding of sBCMA to the anti-BCMA domain of the bispecific drug led to reduced T cell activation, which caused false positive results in NAB testing. To mitigate this interference, several strategies to eliminate sBCMA were investigated. Among the procedures tested, a bead-based approach proved most effective in depleting sBCMA, while maintaining robust assay performance and achieving fit-for-purpose sensitivity. Using this sample pretreatment procedure, the NAB assay tolerated sBCMA up to 2 μg/mL, or approximately four times the estimated median sBCMA concentration in serum samples from patients with active multiple myeloma.
Collapse
Affiliation(s)
- Ying Wang
- BioMedicine Design, Pfizer, Andover, MA, United States of America.
| | - Michael Luong
- BioMedicine Design, Pfizer, Andover, MA, United States of America
| | - Crisanto Guadiz
- BioMedicine Design, Pfizer, Andover, MA, United States of America
| | - Minlei Zhang
- BioMedicine Design, Pfizer, Andover, MA, United States of America
| | - Boris Gorovits
- BioMedicine Design, Pfizer, Andover, MA, United States of America
| |
Collapse
|
42
|
Bioanalytical assays in support of tanezumab developmental and reproductive toxicity studies: challenges and learnings. Bioanalysis 2019; 11:1207-1216. [PMID: 31204868 DOI: 10.4155/bio-2018-0314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bioanalytical challenges were encountered during developmental and reproductive toxicity studies of tanezumab in cynomolgus monkeys. Possible changes in breast milk composition over the postpartum period potentially complicated assessment of tanezumab concentration in this matrix, requiring validation of the quantification assay across different time intervals. Immunogenicity assessment in maternal serum was complicated by apparent increases in the incidence of antidrug antibody-positive results in treatment-naive samples as pregnancy progressed that were due to changes in the concentration of nerve growth factor, tanezumab's target protein. This was overcome by employing gestational day-specific cut points throughout pregnancy. Researchers should recognize potential challenges associated with dynamic matrices/physiological conditions and anticipate that assays developed under normal conditions may require adaptation for specialized situations.
Collapse
|
43
|
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
|
44
|
Gorovits B, Wang Y, Zhu L, Araya M, Kamerud J, Lepsy C. Anti-drug Antibody Assay Conditions Significantly Impact Assay Screen and Confirmatory Cut-Points. AAPS JOURNAL 2019; 21:71. [DOI: 10.1208/s12248-019-0342-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/17/2019] [Indexed: 01/12/2023]
|
45
|
Xiang Y, Parng C, Olson K, Seletskaia E, Gorovits B, Jani D, Caiazzo T, Joyce A, Donley J. Neutralizing Antibody Assay Development with High Drug and Target Tolerance to Support Clinical Development of an Anti-TFPI Therapeutic Monoclonal Antibody. AAPS JOURNAL 2019; 21:46. [PMID: 30927117 DOI: 10.1208/s12248-019-0320-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/12/2019] [Indexed: 11/30/2022]
Abstract
Immunogenicity is a major challenge for protein therapeutics which can potentially reduce drug efficacy and safety and is often being monitored by anti-drug antibody (ADA) and neutralizing antibody (NAb) assays. Circulating targets and residual drugs in matrices can have significant impacts on accuracy of results from ADA and NAb assays, and sufficient drug and target tolerance for these assays are necessary. Here, we report the development of a competitive ligand binding (CLB) NAb assay for an anti-TFPI (tissue factor pathway inhibitor) monoclonal antibody (PF-06741086) with high drug and target tolerance to support ongoing clinical studies. A double acid affinity capture elution approach was used to mitigate drug interference, and a robust target removal strategy was employed to enhance target tolerance. The validated NAb assay has sensitivity of 313 ng/mL, drug tolerance of 50 μg/mL, and target tolerance of 1200 ng/mL. A step-by-step tutorial of assay development is described in this manuscript along with the rationale for using a high drug/target tolerant NAb assay. The NAb assay cut point factor obtained was 0.78. Other assay performance characteristics, e.g., precision and selectivity, are also discussed. This validated method demonstrated a superior drug and target tolerance to warrant specific and precise characterization of the NAb responses in support of ongoing clinical studies.
Collapse
|
46
|
Perspectives on potentiating immunocapture-LC-MS for the bioanalysis of biotherapeutics and biomarkers. Bioanalysis 2018; 10:1679-1690. [PMID: 30371100 DOI: 10.4155/bio-2018-0205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The integration of ligand-binding assay and LC-MS/MS (immunocapture-LC-MS) has unleashed the combined advantages of both powerful techniques for addressing the ever increasing bioanalytical challenges for biotherapeutics and biomarker assays. The highly specific, selective and sensitive characteristics of the immunocapture-LC-MS-based assays have enabled the determination of biotherapeutics and biomarkers in biomatrices with ease of method development, less requirements on key reagents as well as structural specificity for endogenous and engineered biomolecules. In addition, the versatile immunocapture-LC-MS technology has expanded into many challenging areas to enhance mechanistic studies of drug interactions with their targets. This paper intends to summarize our perspectives on enhancing the use of immunocapture-LC-MS in drug discovery and development for emerging new modalities.
Collapse
|
47
|
Quarmby V, Phung QT, Lill JR. MAPPs for the identification of immunogenic hotspots of biotherapeutics; an overview of the technology and its application to the biopharmaceutical arena. Expert Rev Proteomics 2018; 15:733-748. [DOI: 10.1080/14789450.2018.1521279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Valerie Quarmby
- Department of BioAnalytical Sciences, Genentech Inc., San Francisco, CA, USA
| | - Qui T Phung
- Department of Microchemistry, Proteomics & Lipidomics, Genentech Inc., San Francisco, CA, USA
| | - Jennie R Lill
- Department of Microchemistry, Proteomics & Lipidomics, Genentech Inc., San Francisco, CA, USA
| |
Collapse
|
48
|
Immunogenicity testing of therapeutic antibodies in ocular fluids after intravitreal injection. Bioanalysis 2018; 10:803-814. [DOI: 10.4155/bio-2018-0047] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aim: High drug concentrations in ocular fluids after intravitreal administration preclude the use of drug-sensitive immunoassays. A drug-tolerant immunoassay is therefore desirable for immunogenicity testing in ophthalmology. Experimental: Immune complex (IC) antidrug antibody (ADA) assays were established for two species. The assays were compared with the bridging assay in ocular and plasma samples from two preclinical studies. Results: The IC assays showed high drug tolerance, which enabled a reliable ADA detection in ocular fluids after intravitreal administration. The IC assays were superior to the bridging assay in the analysis of ocular fluids with high drug concentrations. Conclusion: The IC assay allows a reliable ADA detection in matrices with high drug concentrations, such as ocular fluids.
Collapse
|