1
|
Jarab AS, Abu Heshmeh SR, Al Meslamani AZ. Biosimilars and immunogenicity: a matter of concern? Expert Opin Drug Saf 2025; 24:519-527. [PMID: 39955621 DOI: 10.1080/14740338.2025.2467817] [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: 02/06/2024] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Biosimilars have transformed treatment modalities across various medical fields such as oncology, rheumatology, and immunology. Despite their potential for reducing healthcare costs, concerns persist regarding their ability to induce an immune response, which could affect efficacy and safety. This review critically evaluates the current evidence on the immunogenicity of biosimilars and discusses the regulatory frameworks guiding their approval and monitoring. AREAS COVERED This review includes studies from databases like Scopus, PubMed, Web of Science, and ScienceDirect, published up to April 2024. It explores the 'totality of the evidence' approach used by regulatory bodies like the FDA and EMA, detailing analytical, preclinical, and clinical assessments that ensure biosimilars' similarity to their reference products in terms of structure, function, and clinical outcomes. The review also addresses the challenges and limitations in current research methodologies and the implications of immunogenicity on therapeutic efficacy and patient safety. EXPERT OPINION While substantial evidence confirms the safety and efficacy of biosimilars, the review emphasizes the need for continuous regulatory vigilance and advanced methodologies in post-marketing surveillance to capture long-term immunogenicity data effectively. It advocates for integrating cutting-edge analytical techniques and personalized medicine to better manage immunogenic risks associated with biological therapies.
Collapse
Affiliation(s)
- Anan S Jarab
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Shrouq R Abu Heshmeh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Ahmad Z Al Meslamani
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| |
Collapse
|
2
|
Williams WT, Lindley K, Liao H, Kamen L, Miller M, Hays A, Sailstad J. Development of a validated novel bead extraction method for the detection of anti-PEG antibodies in human serum. Bioanalysis 2025; 17:7-15. [PMID: 39696894 PMCID: PMC11749383 DOI: 10.1080/17576180.2024.2442198] [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: 09/17/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
AIMS Polyethylene glycol (PEG) is used in many applications including drug development. Due to exposure to environmental products, there is a high prevalence of preexisting anti-PEG antibodies in the global human population. The presence of anti-PEG antibodies is a concern for potentially reducing the efficacy of therapeutics after administration and represents a risk of safety events after exposure to PEGylated drug products. We developed and validated a creative and sensitive method for the detection of anti-PEG antibodies in human serum to support clinical programs for PEGylated drugs. METHODS In this method, biotin-PEG streptavidin beads were used to extract anti-PEG antibodies from human serum for analysis in an anti-PEG ELISA assay. The same serum sample was analyzed in an anti-drug antibody assay. RESULTS The anti-PEG antibody assay was validated with a screening cut point of 1.41 normalized signal, confirmatory cut point of 32.2% inhibition, sensitivity of 7.81 ng/mL and sufficient reproducibility, selectivity, and drug tolerance in accordance with the FDA 2019 Immunogenicity guidance. CONCLUSION This method of removal of anti-PEG antibodies enables the use of a single sample to detect anti-drug and anti-PEG antibodies to support drug development programs.
Collapse
|
3
|
Kalluri HV, Rosebraugh MR, Boehm N, Locke C, Ziemann A, Xiong H. Comparability of Elezanumab Safety, Tolerability, and Pharmacokinetics in Healthy Japanese, Chinese, and White Participants. Clin Pharmacol Drug Dev 2024; 13:180-189. [PMID: 38191982 DOI: 10.1002/cpdd.1341] [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: 07/12/2023] [Accepted: 10/23/2023] [Indexed: 01/10/2024]
Abstract
Elezanumab is a fully human monoclonal antibody, which is directed against repulsive guidance molecule A. The safety, tolerability, pharmacokinetics (PK), and immunogenicity of elezanumab were assessed in 2 Phase 1 clinical studies. The objective of this study was to assess the PK, safety, tolerability, and immunogenicity following intravenous infusion of elezanumab in healthy adult Japanese, Han Chinese, and Caucasian participants as well as Western participants from the single-ascending-dose study. Elezanumab exposures were approximately 20% higher in Japanese and Han Chinese participants compared to White participants without controlling for body weight. After statistically controlling for body weight by including it as a covariate, the PK of elezanumab in White participants were comparable to those in Japanese and Han Chinese participants. The clinical implications of these exposure differences are yet to be determined. All adverse events were assessed by the investigator as having no reasonable possibility of being related to the study drugs and were mild in severity. No positive immunogenicity effect was observed that impacted elezanumab exposure or safety.
Collapse
Affiliation(s)
- Hari V Kalluri
- Clinical Pharmacology, AbbVie Inc, North Chicago, IL, USA
| | | | - Nils Boehm
- DMPK-BA, AbbVie Inc, North Chicago, IL, USA
| | | | - Adam Ziemann
- Neuroscience Development, AbbVie Inc, North Chicago, IL, USA
| | - Hao Xiong
- Clinical Pharmacology, AbbVie Inc, North Chicago, IL, USA
| |
Collapse
|
4
|
Asher D, Dai D, Klimchak AC, Sedita LE, Gooch KL, Rodino-Klapac L. Paving the way for future gene therapies: A case study of scientific spillover from delandistrogene moxeparvovec. Mol Ther Methods Clin Dev 2023; 30:474-483. [PMID: 37674905 PMCID: PMC10477757 DOI: 10.1016/j.omtm.2023.08.002] [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: 09/08/2023]
Abstract
Gene therapies have potential to improve outcomes of severe diseases after only a single administration. Novel therapies are continually being developed using knowledge gained from prior successes, a concept known as scientific spillover. Gene therapy advancement requires extensive development at each stage: preclinical work to create and evaluate vehicles for delivery of the therapy, design of clinical development programs, and establishment of a large-scale manufacturing process. Pioneering gene therapies are generating spillover as investigators confront myriad issues specific to this treatment modality. These include frameworks for construct engineering, dose evaluation, patient selection, outcome assessment, and safety monitoring. Consequently, the benefits of these therapies extend beyond offering knowledge for treating any one disease to establishing new platforms and paradigms that will accelerate advancement of future gene therapies. This impact is even more profound in rare diseases, where developing therapies in isolation may not be possible. This review describes some instances of scientific spillover in healthcare, and specifically gene therapy, using delandistrogene moxeparvovec (SRP-9001), a gene therapy recently approved by the US Food and Drug Administration for the treatment of ambulatory pediatric patients aged 4-5 years with Duchenne muscular dystrophy with a confirmed mutation in the DMD gene, as a case study.
Collapse
Affiliation(s)
- Damon Asher
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Daisy Dai
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Alexa C. Klimchak
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Lauren E. Sedita
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | | | | |
Collapse
|
5
|
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: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [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
|
6
|
Vu T, Ortiz S, Katsuno M, Annane D, Mantegazza R, Beasley KN, Aguzzi R, Howard JF. Ravulizumab pharmacokinetics and pharmacodynamics in patients with generalized myasthenia gravis. J Neurol 2023; 270:3129-3137. [PMID: 36890354 PMCID: PMC10188401 DOI: 10.1007/s00415-023-11617-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 03/10/2023]
Abstract
INTRODUCTION The terminal complement C5 inhibitor ravulizumab has a long elimination half-life, allowing maintenance dosing every 8 weeks. In the 26-week, double-blind, randomized, placebo-controlled period (RCP) of the CHAMPION MG study, ravulizumab provided rapid and sustained efficacy and was well tolerated in adults with anti-acetylcholine receptor antibody-positive (AChR Ab+) generalized myasthenia gravis (gMG). This analysis evaluated the pharmacokinetics (PK), pharmacodynamics (PD), and potential immunogenicity of ravulizumab in adults with AChR Ab+ gMG. METHODS Data were analyzed from 86 patients who received ravulizumab in the CHAMPION MG RCP. Ravulizumab dosing was weight-based: initial loading dose of 2400/2700/3000 mg on Day 1 and maintenance doses of 3000/3300/3600 mg on Day 15 and then every 8 weeks. PK parameters were estimated from serum ravulizumab concentrations determined pre- and post-dose; PD effects of ravulizumab on serum free C5 concentrations were measured; and immunogenicity was assessed using anti-drug antibody and neutralizing-antibody assays. RESULTS Target serum ravulizumab concentrations (> 175 µg/mL) were achieved immediately after the first ravulizumab dose (within 30 min of infusion completion) and maintained throughout the 26-week treatment period irrespective of patient body weight. Following the final maintenance dose, mean Cmax was 1548 µg/mL and Ctrough 587 µg/mL; no meaningful differences were noted among body-weight categories. Inhibition of serum free C5 was immediate, complete (< 0.5 μg/mL), and sustained throughout treatment in all patients. No treatment-emergent anti-drug antibodies were observed. CONCLUSIONS PK/PD evidence supports the use of ravulizumab every 8 weeks for immediate, complete, and sustained inhibition of terminal complement C5 in adults with AChR Ab+ gMG. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT03920293 (April 18, 2019).
Collapse
Affiliation(s)
- Tuan Vu
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | - Djillali Annane
- Hôpital Raymond Poincaré, University of Versailles, Garches, France
| | | | | | - Rasha Aguzzi
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | | |
Collapse
|
7
|
Jani D, Marsden R, Gunsior M, Hay LS, Ward B, Cowan KJ, Azadeh M, Barker B, Cao L, Closson KR, Coble K, Dholakiya SL, Dusseault J, Hays A, Herl C, Hodsdon ME, Irvin SC, Kirshner S, Kolaitis G, Kulagina N, Kumar S, Lai CH, Lipari F, Liu S, Merdek KD, Moldovan IR, Mozaffari R, Pan L, Place C, Snoeck V, Manning MS, Stocker D, Tary-Lehmann M, Turner A, Vainshtein I, Verthelyi D, Williams WT, Yan H, Yan W, Yang L, Yang L, Zemo J, Zhong ZD. Anti-drug Antibody Sample Testing and Reporting Harmonization. AAPS J 2022; 24:113. [PMID: 36307592 DOI: 10.1208/s12248-022-00762-6] [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: 08/26/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
A clear scientific and operational need exists for harmonized bioanalytical immunogenicity study reporting to facilitate communication of immunogenicity findings and expedient review by industry and health authorities. To address these key bioanalytical reporting gaps and provide a report structure for documenting immunogenicity results, this cross-industry group was formed to establish harmonized recommendations and a develop a submission template to facilitate agency filings. Provided here are recommendations for reporting clinical anti-drug antibody (ADA) assay results using ligand-binding assay technologies. This publication describes the essential bioanalytical report (BAR) elements such as the method, critical reagents and equipment, study samples, results, and data analysis, and provides a template for a suggested structure for the ADA BAR. This publication focuses on the content and presentation of the bioanalytical ADA sample analysis report. The interpretation of immunogenicity data, including the evaluation of the impact of ADA on safety, exposure, and efficacy, is out of scope of this publication.
Collapse
Affiliation(s)
- Darshana Jani
- Bioanalytical and Molecular Assays, Moderna, Cambridge, Massachusetts, USA.
| | | | - Michele Gunsior
- Research and Translational Sciences, Astria Therapeutics, Boston, Massachusetts, USA
| | - Laura Schild Hay
- Bioanalytical Lab, PPD Clinical Research Services, Thermo Fisher Scientific, Richmond, Virginia, USA
| | - Bethany Ward
- Bioanalytical Lab, PPD Clinical Research Services, Thermo Fisher Scientific, Richmond, Virginia, USA
| | - Kyra J Cowan
- New Biological Entities Drug Metabolism and Pharmacokinetics, Merck KGaA, Darmstadt, Germany
| | - Mitra Azadeh
- Biomarker Operations, Translational Medicine and Early Stage Clinical Development, Alkermes, Inc., Waltham, Massachusetts, USA
| | - Breann Barker
- Drug Metabolism and Biopharmaceuticals, Incyte Corporation, Wilmington, Delaware, USA
| | - Liching Cao
- Biomarker and BioAnalytical Sciences, Sangamo Therapeutics, California, USA
| | - Kristin R Closson
- Laboratory Operations, Immunologix Laboratories, Tampa, Florida, USA
| | - Kelly Coble
- DMPK/Bioanalytical Sciences, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Sanjay L Dholakiya
- Non-Clinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Julie Dusseault
- Laboratory Sciences, Charles River Laboratories, Quebec, Canada
| | | | - Carina Herl
- Clinical Pharmacology and Translational Sciences, Exelixis, Alameda, California, USA
| | - Michael E Hodsdon
- Laboratory for Experimental Medicine, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Susan C Irvin
- Bioanalytical Sciences, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Susan Kirshner
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gerry Kolaitis
- Non-Clinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Nadia Kulagina
- Pharmaceutical Development Services, Smithers, Gaithersburg, Maryland, USA
| | - Seema Kumar
- EMD Serono Research and Development Institute (A business of Merck KGaA, Darmstadt, Germany), Billerica, Massachusetts, USA
| | - Ching Ha Lai
- Bioanalytical Sciences, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Francesco Lipari
- Nexelis, a Q2 Solutions Company, Vaccine Sciences, Laval, Quebec, Canada
| | - Susana Liu
- Global Product Development, Clinical Assay Group, Pfizer Inc., Kirkland, Quebec, Canada
| | - Keith D Merdek
- Biomarkers and Clinical Bioanalyses (TMED), Sanofi, Framingham, Massachusetts, USA
| | | | - Reza Mozaffari
- Bioanalysis, Immunogenicity and Biomarkers (BIB), IVIVT, Research, GSK, Collegeville, Pennsylvania, USA
| | - Luying Pan
- Clinical Biomarker Innovation and Development, Takeda Development Center Americas Inc., Cambridge, Massachusetts, USA
| | - Corina Place
- DMPK/Bioanalytical Sciences, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Veerle Snoeck
- Translational Biomarkers and Bioanalysis, UCB Biopharma SRL, Braine-l'Alleud, Belgium
| | | | - Dennis Stocker
- Non-Clinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Amy Turner
- Pharmaceutical Development Services, Smithers, Gaithersburg, Maryland, USA
| | - Inna Vainshtein
- Clinical Pharmacology and Translational Sciences, Exelixis, Alameda, California, USA
| | - Daniela Verthelyi
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Haoheng Yan
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drugs Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Weili Yan
- Department of Bioanalytical Sciences, Genentech, South San Francisco, California, USA
| | - Lili Yang
- Clinical Biomarker Innovation and Development, Takeda Development Center Americas Inc., Cambridge, Massachusetts, USA
| | - Lin Yang
- Bioanalytical Sciences, REGENXBIO Inc., Rockville, Maryland, USA
| | - Jennifer Zemo
- Bioanalytical Operations, BioAgilytix Labs, Durham, North Carolina, USA
| | - Zhandong Don Zhong
- Development Sciences, Denali Therapeutics, South San Francisco, California, USA
| |
Collapse
|
8
|
Irvin SC, D’Orvilliers A, Bloch N, Boccio K, Pennucci J, Brouwer-Visser J, Ullman E, Rajadhyaksha M, Hassanein M, Potocky T, Torri A, Hermann A, Partridge MA. Interference in a Neutralizing Antibody Assay for Odronextamab, a CD20xCD3 Bispecific mAb, from Prior Rituximab Therapy and Possible Mitigation Strategy. AAPS J 2022; 24:76. [DOI: 10.1208/s12248-022-00724-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
AbstractA cell-based assay was developed to detect neutralizing anti-drug antibodies (NAbs) against odronextamab, a CD20xCD3 bispecific monoclonal antibody (mAb) under investigation for treatment of CD20+ B cell malignancies. In this assay, odronextamab bridges between two cell types, CD20-expressing HEK293 cells and CD3-expressing Jurkat T cells that generate a luciferase signal upon CD3 clustering. Patient samples containing NAbs directed to either arm of the bispecific drug block the odronextamab bridge formation between the cell lines thus preventing the generation of the luciferase signal. We determined that other anti-CD20 therapeutics also block bridge formation, resulting in false-positive results. In patient samples from odronextamab clinical trials, approximately 30% of baseline samples had a strong false-positive NAb signal that correlated with the presence of prior rituximab (anti-CD20) therapy. We determined that rituximab interference can be minimized by the addition of anti-rituximab antibodies in the NAb assay. Understanding and mitigating the impact of prior biologic exposure is increasingly important for implementing a successful bioanalytical strategy to support clinical drug development, especially in the immuno-oncology field.
Graphical Abstract
Odronextamab neutralizing antibody assay, interference, and mitigation. A Design of the odronextamab neutralizing antibody (NAb) assay where anti-CD20xCD3 drug bridges between CD20-expressing HEK293 cells and Jurkat T cells expressing an NFAT response element and luciferase reporter. True NAb prevents odronextamab from bridging between target and effector cells, thus preventing the expression of luciferase. B Interference with odronextamab from other anti-CD20 therapeutic antibodies (e.g., rituximab) from prior disease treatment generates a false-positive NAb result. Assay interference can be mitigated with an anti-idiotypic antibody against the interfering therapy.
Collapse
|
9
|
Wasserstein M, Lachmann R, Hollak C, Arash-Kaps L, Barbato A, Gallagher RC, Giugliani R, Guelbert NB, Ikezoe T, Lidove O, Mabe P, Mengel E, Scarpa M, Senates E, Tchan M, Villarrubia J, Chen Y, Furey S, Thurberg BL, Zaher A, Kumar M. A randomized, placebo-controlled clinical trial evaluating olipudase alfa enzyme replacement therapy for chronic acid sphingomyelinase deficiency (ASMD) in adults: One-year results. Genet Med 2022; 24:1425-1436. [PMID: 35471153 DOI: 10.1016/j.gim.2022.03.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 12/20/2022] Open
Abstract
PURPOSE This trial aimed to assess the efficacy and safety of olipudase alfa enzyme replacement therapy for non-central nervous system manifestations of acid sphingomyelinase deficiency (ASMD) in adults. METHODS A phase 2/3, 52 week, international, double-blind, placebo-controlled trial (ASCEND; NCT02004691/EudraCT 2015-000371-26) enrolled 36 adults with ASMD randomized 1:1 to receive olipudase alfa or placebo intravenously every 2 weeks with intrapatient dose escalation to 3 mg/kg. Primary efficacy endpoints were percent change from baseline to week 52 in percent predicted diffusing capacity of the lung for carbon monoxide and spleen volume (combined with splenomegaly-related score in the United States). Other outcomes included liver volume/function/sphingomyelin content, pulmonary imaging/function, platelet levels, lipid profiles, and pharmacodynamics. RESULTS Least square mean percent change from baseline to week 52 favored olipudase alfa over placebo for percent predicted diffusing capacity of the lung for carbon monoxide (22% vs 3.0% increases, P = .0004), spleen volume (39% decrease vs 0.5% increase, P < .0001), and liver volume (28% vs 1.5% decreases, P < .0001). Splenomegaly-related score decreased in both groups (P = .64). Other clinical outcomes improved in the olipudase alfa group compared with the placebo group. There were no treatment-related serious adverse events or adverse event-related discontinuations. Most adverse events were mild. CONCLUSION Olipudase alfa was well tolerated and associated with significant and comprehensive improvements in disease pathology and clinically relevant endpoints compared with placebo in adults with ASMD.
Collapse
Affiliation(s)
- Melissa Wasserstein
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY.
| | - Robin Lachmann
- Charles Dent Metabolic Unit, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Carla Hollak
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Laila Arash-Kaps
- Villa Metabolica, Department of Pediatric and Adolescent Medicine, University Medical Center Mainz, Mainz, Germany; Clinical Science for LSD, SphinCS, Hochheim, Germany
| | - Antonio Barbato
- Department of Clinical Medicine and Surgery, "Federico II" University Hospital, Naples, Italy
| | - Renata C Gallagher
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA
| | - Roberto Giugliani
- Medical Genetics Service and DR BRASIL Research Group, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil; Department of Genetics, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute on Population Medical Genetics (INAGEMP), Porto Alegre, Brazil
| | | | - Takayuki Ikezoe
- Department of Hematology, Fukushima Medical University, Fukushima, Japan
| | - Olivier Lidove
- Service de Médecine Interne, Diaconesses Croix Saint-Simon Hospital, Paris, France
| | - Paulina Mabe
- Servicio de Pediatría, Clínica Santa María, Santiago, Chile
| | - Eugen Mengel
- Clinical Science for LSD, SphinCS, Hochheim, Germany
| | - Maurizio Scarpa
- Regional Coordinator Centre for Rare Diseases, University Hospital of Udine, Udine, Italy
| | - Eubekir Senates
- Department of Gastroenterology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Michel Tchan
- Department of Genetic Medicine, Westmead Hospital, Sydney, Australia
| | - Jesus Villarrubia
- Hematology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Yixin Chen
- Clinical Development, Sanofi, Bridgewater, NJ
| | - Sandy Furey
- Clinical Development, Sanofi, Bridgewater, NJ
| | | | - Atef Zaher
- Clinical Development, Sanofi, Bridgewater, NJ
| | | |
Collapse
|
10
|
Zhou Y, Penny HL, Kroenke MA, Bautista B, Hainline K, Chea LS, Parnes J, Mytych DT. Immunogenicity assessment of bispecific antibody-based immunotherapy in oncology. J Immunother Cancer 2022; 10:e004225. [PMID: 35444060 PMCID: PMC9024276 DOI: 10.1136/jitc-2021-004225] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/18/2022] Open
Abstract
With increasing numbers of bispecific antibodies (BsAbs) and multispecific products entering the clinic, recent data highlight immunogenicity as an emerging challenge in the development of such novel biologics. This review focuses on the immunogenicity risk assessment (IgRA) of BsAb-based immunotherapies for cancer, highlighting several risk factors that need to be considered. These include the novel scaffolds consisting of bioengineered sequences, the potentially synergistic immunomodulating mechanisms of action (MOAs) from different domains of the BsAb, as well as several other product-related and patient-related factors. In addition, the clinical relevance of anti-drug antibodies (ADAs) against selected BsAbs developed as anticancer agents is reviewed and the advances in our knowledge of tools and strategies for immunogenicity prediction, monitoring, and mitigation are discussed. It is critical to implement a drug-specific IgRA during the early development stage to guide ADA monitoring and risk management strategies. This IgRA may include a combination of several assessment tools to identify drug-specific risks as well as a proactive risk mitigation approach for candidate or format selection during the preclinical stage. The IgRA is an on-going process throughout clinical development. IgRA during the clinical stage may bridge the gap between preclinical immunogenicity prediction and clinical immunogenicity, and retrospectively guide optimization efforts for next-generation BsAbs. This iterative process throughout development may improve the reliability of the IgRA and enable the implementation of effective risk mitigation strategies, laying the foundation for improved clinical success.
Collapse
Affiliation(s)
- Yanchen Zhou
- Clinical Immunology, Amgen Inc, South San Francisco, California, USA
| | | | - Mark A Kroenke
- Clinical Immunology, Amgen Inc, Thousand Oaks, California, USA
| | - Bianca Bautista
- Clinical Immunology, Amgen Inc, Thousand Oaks, California, USA
| | - Kelly Hainline
- Clinical Immunology, Amgen Inc, Thousand Oaks, California, USA
| | - Lynette S Chea
- Clinical Immunology, Amgen Inc, South San Francisco, California, USA
| | - Jane Parnes
- Early Development, Amgen Inc, Thousand Oaks, California, USA
| | - Daniel T Mytych
- Clinical Immunology, Amgen Inc, Thousand Oaks, California, USA
| |
Collapse
|
11
|
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
|
12
|
Li Y, Qi L, Bai H, Sun C, Xu S, Wang Y, Han C, Li Y, Liu L, Cheng X, Liu J, Lei C, Tong Y, Sun M, Yan L, Chen W, Liu X, Liu Q, Xie L, Wang X. Safety, Tolerability, Pharmacokinetics, and Immunogenicity of a Monoclonal Antibody (SCTA01) Targeting SARS-CoV-2 in Healthy Adults: a Randomized, Double-Blind, Placebo-Controlled, Phase I Study. Antimicrob Agents Chemother 2021; 65:e0106321. [PMID: 34491805 PMCID: PMC8522782 DOI: 10.1128/aac.01063-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/20/2021] [Indexed: 01/08/2023] Open
Abstract
SCTA01 is a novel monoclonal antibody with promising prophylactic and therapeutic potential for COVID-19. This study aimed to evaluate the safety, tolerability, pharmacokinetics (PK) and immunogenicity of SCTA01 in healthy adults. This was a randomized, double-blind, placebo-controlled, dose escalation phase I clinical trial. Healthy adults were randomly assigned to cohort 1 (n = 5; 3:2), cohort 2 (n = 8; 6:2), cohort 3, or cohort 4 (both n = 10; 8:2) to receive SCTA01 (5, 15, 30, and 50 mg/kg, respectively) versus placebo. All participants were followed up for clinical, laboratory, PK, and immunogenicity assessments for 84 days. The primary outcomes were the dose-limiting toxicity (DLT) and maximal tolerable dose (MTD), and the secondary outcomes included PK parameters, immunogenicity, and adverse events (AE). Of the 33 participants, 18 experienced treatment-related AEs; the frequency was 52.0% (13/25) in participants receiving SCTA01 and 62.5% (5/8) in those receiving placebo. All AEs were mild. There was no serious AE or death. No DLT was reported, and the MTD of SCTA01 was not reached. SCTA01 with a dose range of 5 to 50 mg/kg had nearly linear dose-proportional increases in Cmax and AUC parameters. An antidrug antibody response was detected in four (16.0%) participants receiving SCTA01, with low titers, between the baseline and day 28, but all became negative later. In conclusion, SCTA01 up to 50 mg/kg was safe and well-tolerated in healthy participants. Its PK parameters were nearly linear dose-proportional. (This study has been registered at ClinicalTrials.gov under identifier NCT04483375.).
Collapse
Affiliation(s)
- Yinjuan Li
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lu Qi
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Haihong Bai
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chunyun Sun
- Sinocelltech Ltd., Beijing, China
- Beijing Engineering Research Center of Protein and Antibody, Beijing, China
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | - Yu Wang
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chunyu Han
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yan Li
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Long Liu
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoqiang Cheng
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ju Liu
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chunpu Lei
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuanxu Tong
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Mingli Sun
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | | | | | | | - Qing Liu
- Sinocelltech Ltd., Beijing, China
| | - Liangzhi Xie
- Sinocelltech Ltd., Beijing, China
- Beijing Engineering Research Center of Protein and Antibody, Beijing, China
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinghe Wang
- Department of Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Diaz GA, Jones SA, Scarpa M, Mengel KE, Giugliani R, Guffon N, Batsu I, Fraser PA, Li J, Zhang Q, Ortemann-Renon C. One-year results of a clinical trial of olipudase alfa enzyme replacement therapy in pediatric patients with acid sphingomyelinase deficiency. Genet Med 2021; 23:1543-1550. [PMID: 33875845 PMCID: PMC8354848 DOI: 10.1038/s41436-021-01156-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To assess olipudase alfa enzyme replacement therapy for non-central nervous system manifestations of acid sphingomyelinase deficiency (ASMD) in children. METHODS This phase 1/2, international, multicenter, open-label trial (ASCEND-Peds/NCT02292654) administered intravenous olipudase alfa every 2 weeks with intrapatient dose escalation to 3 mg/kg. Primary outcome was safety through week 64. Secondary outcomes included pharmacokinetics, spleen and liver volumes, lung diffusing capacity (DLCO), lipid profiles, and height through week 52. RESULTS Twenty patients were enrolled: four adolescents (12-17 years), nine children (6-11 years), and seven infants/early child (1-5 years). Most adverse events were mild or moderate, including infusion-associated reactions (primarily urticaria, pyrexia, and/or vomiting) in 11 patients. Three patients had serious treatment-related events: one with transient asymptomatic alanine aminotransferase increases, another with urticaria and rash (antidrug antibody positive [ADA+]), and a third with an anaphylactic reaction (ADA+) who underwent desensitization and reached the 3 mg/kg maintenance dose. Mean splenomegaly and hepatomegaly improved by >40% (p < 0.0001). Mean % predicted DLCO improved by 32.9% (p = 0.0053) in patients able to perform the test. Lipid profiles and elevated liver transaminase levels normalized. Mean height Z-scores improved by 0.56 (p < 0.0001). CONCLUSION In this study in children with chronic ASMD, olipudase alfa was generally well-tolerated with significant, comprehensive improvements in disease pathology across a range of clinically relevant endpoints.
Collapse
Affiliation(s)
- George A Diaz
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Simon A Jones
- St Mary's Hospital, Manchester University Foundation Trust, University of Manchester, Manchester, UK
| | - Maurizio Scarpa
- Regional Coordinating Center for Rare Diseases, University Hospital Udine, Udine, Italy
| | | | - Roberto Giugliani
- Dept Genetics, UFRGS, Medical Genetics Clinical Research Group, HCPA, and INAGEMP, Porto Alegre, Brazil
| | - Nathalie Guffon
- Reference Center for Inherited Metabolic Disorders, Femme Mère Enfant Hospital, Lyon, France
| | | | | | - Jing Li
- Sanofi, Bridgewater, NJ, USA
| | | | | |
Collapse
|
14
|
Wang Y, Smith JF, Araya MM, Liao KHK, Gorovits B. Development of a Highly Specific Anti-drug Antibody Assay in Support of a Nanoparticle-based Therapeutic. AAPS JOURNAL 2020; 22:81. [PMID: 32488626 DOI: 10.1208/s12248-020-00462-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/29/2020] [Indexed: 11/30/2022]
Abstract
PEGylated biotherapeutics can elicit anti-PEG (polyethylene glycol) immune responses in patients treated with this category of drugs. While anti-PEG antibody assays for this class of biotherapeutics have become a common element of the clinical immunogenicity testing strategy, the overall antibody incidence induced by the nanoparticle (NP) delivery system (such as ACCURINS®) has not been fully studied to date. To support the immunogenicity assessment of one of Pfizer's NP-based therapeutics, consisting of gedatolisib (GEDA) encapsulated in ACCURINS® (GEDA-NP), we developed an anti-GEDA-NP antibody (ADA) assay on the MSD platform for the detection of GEDA-NP induced ADA in human serum. The focus of our strategy was on developing a clinically relevant ADA assay and systematically addressing assay interference through rigorous assay optimization. Our efforts led to a fit-for-purpose assay for the detection of anti-GEDA-NP ADA in serum samples obtained from breast cancer patients. Results from method qualification indicated robust assay performance, as highlighted by inter and intra-assay precision within 25% CV for all controls, and reproducible response profiles across multiple runs during the assessment of assay cut points with breast cancer samples. The assay sensitivity was between 4.3 ng/mL and 123 ng/mL for surrogate positive controls of IgG and IgM isotypes, respectively. Additionally, assay interference from nonspecific matrix proteins and circulating drug was addressed, which ensured accurate assessment of ADA incidence that can be attributed to GEDA-NP.
Collapse
Affiliation(s)
- Ying Wang
- BioMedicine Design, Pfizer Inc, One Burtt Road, Andover, Massachusetts, 01810, USA.
| | - Judith F Smith
- BioMedicine Design, Pfizer Inc, One Burtt Road, Andover, Massachusetts, 01810, USA
| | - Marcela M Araya
- BioMedicine Design, Pfizer Inc, One Burtt Road, Andover, Massachusetts, 01810, USA
| | - Kai-Hsin Ken Liao
- Early Clinical Development, Pfizer Inc, 10777 Science Center Drive, San Diego, California, 92121, USA
| | - Boris Gorovits
- BioMedicine Design, Pfizer Inc, One Burtt Road, Andover, Massachusetts, 01810, USA
| |
Collapse
|
15
|
Analysis of regulatory guidance on antidrug antibody testing for therapeutic protein products. Bioanalysis 2020; 11:2283-2296. [PMID: 31845602 DOI: 10.4155/bio-2019-0241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Therapeutic proteins have the potential to induce unwanted immune responses. The potential impact of immunogenicity on pharmacokinetics, pharmacodynamics, safety and efficacy are well established. Here, we analyze key aspects of current US FDA and EMA guidelines on the development and validation of antidrug antibody assays. Although FDA and EMA guidance documents are in harmony on most points, EMA allows greater leeway for scientific judgement, while FDA recommends specific approaches that may not be appropriate in some situations. Many white papers suggest approaches different from the guidance documents, however, these can conflict with each other and are themselves only scientifically valid in certain situations. Here, we indicate when alternatives to guidance may be needed and what those approaches might be.
Collapse
|
16
|
Gorovits B, Roldan MA, Baltrukonis D, Cai CH, Donley J, Jani D, Kamerud J, McCush F, Thomas JS, Wang Y. Anti-drug Antibody Assay Validation: Improved Reporting of the Assay Selectivity via Simpler Positive Control Recovery Data Analysis. AAPS JOURNAL 2019; 21:76. [PMID: 31214862 DOI: 10.1208/s12248-019-0347-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 05/28/2019] [Indexed: 01/31/2023]
Abstract
Anti-drug antibody (ADA) assay selectivity is evaluated during assay validation to assess the potential for individual matrices to interfere with detection of ADA. While current EMA and FDA guideline documents suggest comparative analysis with and without matrix, they do not provide specific recommendations on the acceptance criteria such as an acceptable percent positive control (PC) recovery range or positive rate. Industry has adopted an approach where recovery of PC spiked sample is expected to fall within ± 20% (80 to 120%) vs. that for the PC material spiked in negative control matrix or assay buffer. Here, it is proposed that ADA assay selectivity evaluated using a qualitative assessment of PC recovery vs. a PK-like quantitative method may be more appropriate. The PC recovery test should focus on the reliability of the method to detect the low PC level in individual samples and avoid false-negative ADA reporting. Therefore, it is proposed that assessment of high PC level as well as the assessment of quantitative percent recovery (within ± 20%) should not be included in the test. The recovery test may be viewed as acceptable should a pre-selected number of individual samples (for example at least 8 or 9 out of 10) prepared at the low PC concentration of the assay score as ADA positive.
Collapse
Affiliation(s)
| | | | | | - Chun-Hua Cai
- Pfizer Inc., Eastern Point Road, Groton, CT, 06340, USA
| | - Jean Donley
- Pfizer Inc., 1 Burtt Rd., Andover, MA, 01810, USA
| | | | - John Kamerud
- Pfizer Inc., 1 Burtt Rd., Andover, MA, 01810, USA
| | | | | | - Ying Wang
- Pfizer Inc., 1 Burtt Rd., Andover, MA, 01810, USA
| |
Collapse
|
17
|
Brys M, Fanning L, Hung S, Ellenbogen A, Penner N, Yang M, Welch M, Koenig E, David E, Fox T, Makh S, Aldred J, Goodman I, Pepinsky B, Liu Y, Graham D, Weihofen A, Cedarbaum JM. Randomized phase I clinical trial of anti-α-synuclein antibody BIIB054. Mov Disord 2019; 34:1154-1163. [PMID: 31211448 PMCID: PMC6771554 DOI: 10.1002/mds.27738] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/03/2019] [Accepted: 05/20/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Pathological and genetic evidence implicates toxic effects of aggregated α-synuclein in the pathophysiology of neuronal dysfunction and degeneration in Parkinson's disease. Immunotherapy targeting aggregated α-synuclein is a promising strategy for delaying disease progression. OBJECTIVE This study (NCT02459886) evaluated the safety, tolerability, and pharmacokinetics of BIIB054, a human-derived monoclonal antibody that preferentially binds to aggregated α-synuclein, in healthy volunteers and participants with Parkinson's disease. METHODS A total of 48 healthy volunteers (age 40-65, 19 women) and 18 Parkinson's disease participants (age 47-75, 5 women, Hoehn and Yahr stage ≤2.5) were in the study. Volunteers were enrolled into 6 single-dose cohorts of BIIB054 (range 1-135 mg/kg) or placebo, administered intravenously; Parkinson's disease participants received a single dose of BIIB054 (15 or 45 mg/kg) or placebo. All participants were evaluated for 16 weeks with clinical, neuroimaging, electrocardiogram, and laboratory assessments. Serum and cerebrospinal fluid BIIB054 concentrations were measured. BIIB054/α-synuclein complexes were measured in plasma. RESULTS Most adverse events were mild and assessed by investigators as unrelated to the study drug. Pharmacokinetic parameters for volunteers and the Parkinson's disease participants were similar. BIIB054 serum exposure and maximum concentrations were dose proportional during the dose range studied. In volunteers and the Parkinson's disease participants, the serum half-life of BIIB054 was 28 to 35 days; the cerebrospinal fluid-to-serum ratio ranged from 0.13% to 0.56%. The presence of BIIB054/α-synuclein complexes in plasma was confirmed; all Parkinson's disease participants showed almost complete saturation of the BIIB054/α-synuclein complex formation. CONCLUSIONS BIIB054 has favorable safety, tolerability, and pharmacokinetic profiles in volunteers and Parkinson's disease participants, supporting further clinical development. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | | | | | - Aaron Ellenbogen
- Michigan Institute for Neurological Disorders, Farmington Hills, Michigan, USA.,QUEST Research Institute, Farmington, Michigan, USA
| | | | | | | | | | | | | | | | - Jason Aldred
- Selkirk Neurology & Inland Northwest Neurological, Spokane, Washington, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
2018 White Paper on Recent Issues in Bioanalysis: focus on flow cytometry, gene therapy, cut points and key clarifications on BAV (Part 3 - LBA/cell-based assays: immunogenicity, biomarkers and PK assays). Bioanalysis 2018; 10:1973-2001. [PMID: 30488726 DOI: 10.4155/bio-2018-0287] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The 2018 12th Workshop on Recent Issues in Bioanalysis took place in Philadelphia, PA, USA on April 9-13, 2018 with an attendance of over 900 representatives from pharmaceutical/biopharmaceutical companies, biotechnology companies, contract research organizations and regulatory agencies worldwide. WRIB was once again a 5-day full immersion in bioanalysis, biomarkers and immunogenicity. As usual, it was specifically designed to facilitate sharing, reviewing, discussing and agreeing on approaches to address the most current issues of interest including both small- and large-molecule bioanalysis involving LCMS, hybrid LBA/LCMS and LBA/cell-based assays approaches. This 2018 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2018 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations for large molecule bioanalysis, biomarkers and immunogenicity using LBA and cell-based assays. Part 1 (LCMS for small molecules, peptides, oligonucleotides and small molecule biomarkers) and Part 2 (hybrid LBA/LCMS for biotherapeutics and regulatory agencies' inputs) are published in volume 10 of Bioanalysis, issues 22 and 23 (2018), respectively.
Collapse
|