1
|
Liu W, Yang J, Yan W, Peng K. Reformation of a Clinical Anti-Drug Antibody Assay to Enable the Immunogenicity Assessment of a Bispecific Antibody Biotherapeutic. AAPS J 2024; 27:12. [PMID: 39663290 DOI: 10.1208/s12248-024-00996-6] [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: 09/05/2024] [Accepted: 11/09/2024] [Indexed: 12/13/2024] Open
Abstract
An enzyme-linked immunosorbent assay (ELISA) based anti-drug antibody (ADA) assay was developed to support the clinical development of a bispecific antibody biotherapeutic anti-A/B. This anti-A/B clinical ADA Version 1 (V1) assay was successfully validated initially using commercial samples from the target indication. However, applying the validation cut point factors (CPFs) led to a high untreated ADA positive rate in the Phase 1 study baseline sample analysis. While implementing the in-study CPFs was effective to mitigate the high baseline prevalence, this led to unfavorable assay sensitivity with no drug tolerance, which necessitated an assay re-optimization. The re-optimized Version 2 assay (V2) was able to mitigate the matrix interference observed in the clinical sample testing using the V1 assay, proven to be a more suitable method. The V2 assay optimization work was discussed, and the performance of the V1 and V2 assays during validation and clinical sample analysis was compared. Preliminary sample testing results generated using the two versions of the assay were compared and the ADA clinical impact was discussed. Our experience insinuates that a successfully validated method does not guarantee to be appropriate for sample testing. Adjustments of the method may be required to ensure that it performs as expected during sample testing and throughout the assay's lifecycle. This work highlights the importance of verifying the assay suitability during clinical sample testing and making appropriate adjustments as needed, especially in the first clinical study and the first study for a new indication.
Collapse
Affiliation(s)
- Wenyu Liu
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA
| | - Jie Yang
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA
| | - Weili Yan
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA
| | - Kun Peng
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA.
| |
Collapse
|
2
|
Jairath V, Yarur A, Osterman MT, James A, Balma D, Mehrotra S, Yang L, Yajnik V, Qasim Khan RM. ENTERPRET: A Randomized Controlled Trial of Vedolizumab Dose Optimization in Patients With Ulcerative Colitis Who Have Early Nonresponse. Clin Gastroenterol Hepatol 2024; 22:1077-1086.e13. [PMID: 37951560 DOI: 10.1016/j.cgh.2023.10.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND & AIMS Patients with ulcerative colitis (UC) may experience nonresponse to biologics, possibly as a result of low drug exposure. This trial assessed the efficacy of dose optimization in patients with UC who have early nonresponse to vedolizumab and high drug clearance. METHODS ENTERPRET was a phase 4, open-label, randomized, controlled trial that included patients with moderate to severe UC who had high drug clearance at week 5 (serum concentration, <50 μg/mL) and nonresponse to standard vedolizumab treatment at week 6. At week 6, eligible patients were randomized 1:1 to receive standard dosing (300 mg every 8 weeks) or dose-optimized vedolizumab (600 mg at week 6, then 300 mg every 4 weeks; or 600 mg at week 6, then 600 mg every 4 weeks [based on week 5 serum concentration]). The primary end point was endoscopic improvement at week 30. RESULTS Of 278 enrolled patients, 132 (47.5%) had a clinical response at week 6. From week 6, 108 patients received standard (n = 53) or dose-optimized vedolizumab (n = 55); among patients with nonresponse at week 6, 86.5% had high drug clearance. At week 30, 10 patients (18.9%) who received standard vedolizumab had endoscopic improvement vs 8 patients (14.5%) who received dose-optimized vedolizumab. Five patients (9.4%) who received standard vedolizumab had clinical remission at week 30 vs 5 patients (9.1%) who received dose-optimized vedolizumab; clinical response was observed in 17 (32.1%) and 17 patients (30.9%), respectively. Safety event rates were similar among treatment groups. CONCLUSIONS In patients with early nonresponse and high drug clearance, vedolizumab dose optimization is probably not required. A proportion of patients benefited from continued treatment irrespective of the dose received. CLINICALTRIALS gov: NCT03029143.
Collapse
Affiliation(s)
- Vipul Jairath
- Division of Gastroenterology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| | - Andres Yarur
- Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Mark T Osterman
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Alexandra James
- Takeda Pharmaceuticals U.S.A., Inc, Lexington, Massachusetts
| | - Diane Balma
- Takeda Pharmaceuticals U.S.A., Inc, Lexington, Massachusetts
| | | | - Lili Yang
- Takeda Pharmaceuticals U.S.A., Inc, Lexington, Massachusetts
| | - Vijay Yajnik
- Takeda Pharmaceuticals U.S.A., Inc, Lexington, Massachusetts
| | | |
Collapse
|
3
|
Weiner JA, Natarajan H, McIntosh CJ, Yang ES, Choe M, Papia CL, Axelrod KS, Kovacikova G, Pegu A, Ackerman ME. Selection of positive controls and their impact on anti-drug antibody assay performance. J Immunol Methods 2024; 528:113657. [PMID: 38479453 DOI: 10.1016/j.jim.2024.113657] [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: 10/23/2023] [Accepted: 03/03/2024] [Indexed: 03/17/2024]
Abstract
Development of assays to reliably identify and characterize anti-drug antibodies (ADAs) depends on positive control anti-idiotype (anti-id) reagents, which are used to demonstrate that the standards recommended by regulatory authorities are met. This work employs a set of therapeutic antibodies under clinical development and their corresponding anti-ids to investigate how different positive control reagent properties impact ADA assay development. Positive controls exhibited different response profiles and apparent assay analytical sensitivity values depending on assay format. Neither anti-id affinity for drug, nor sensitivity in direct immunoassays related to sensitivity in ADA assays. Anti-ids were differentially able to detect damage to drug conjugates used in bridging assays and were differentially drug tolerant. These parameters also failed to relate to assay sensitivity, further complicating selection of anti-ids for use in ADA assay development based on functional characteristics. Given this variability among anti-ids, alternative controls that could be employed across multiple antibody drugs were investigated as a more uniform means to define ADA detection sensitivity across drug products and assay protocols, which could help better relate assay results to clinical risks of ADA responses. Overall, this study highlights the importance of positive control selection to reliable detection and clinical interpretation of the presence and magnitude of ADA responses.
Collapse
Affiliation(s)
- Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, NH, USA
| | - Calum J McIntosh
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Misook Choe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cassidy L Papia
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | | | | | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA; Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, NH, USA.
| |
Collapse
|
4
|
Opolka-Hoffmann E, Edelmann MR, Otteneder MB, Hauri S, Jordan G, Schrag P, Lechmann M, Winter G, Staack RF. Biodistribution of Drug/ADA Complexes: The Impact of Immune Complex Formation on Antibody Distribution. AAPS J 2024; 26:33. [PMID: 38478197 DOI: 10.1208/s12248-024-00899-6] [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: 12/08/2023] [Accepted: 02/10/2024] [Indexed: 04/11/2024] Open
Abstract
The clinical use of therapeutic monoclonal antibodies (mAbs) for the treatment of cancer, inflammation, and other indications has been successfully established. A critical aspect of drug-antibody pharmacokinetics is immunogenicity, which triggers an immune response via an anti-drug antibody (ADA) and forms drug/ADA immune complexes (ICs). As a consequence, there may be a reduced efficacy upon neutralization by ADA or an accelerated drug clearance. It is therefore important to understand immunogenicity in biological therapies. A drug-like immunoglobulin G (IgG) was radiolabeled with tritium, and ICs were formed using polyclonal ADA, directed against the complementary-determining region of the drug-IgG, to investigate in vivo biodistribution in rodents. It was demonstrated that 65% of the radioactive IC dose was excreted within the first 24 h, compared with only 6% in the control group who received non-complexed 3H-drug. Autoradiographic imaging at the early time point indicated a deposition of immune complexes in the liver, lung, and spleen indicated by an increased radioactivity signal. A biodistribution study confirmed the results and revealed further insights regarding excretion and plasma profiles. It is assumed that the immune complexes are readily taken up by the reticuloendothelial system. The ICs are degraded proteolytically, and the released radioactively labeled amino acids are redistributed throughout the body. These are mainly renally excreted as indicated by urine measurements or incorporated into protein synthesis. These biodistribution studies using tritium-labeled immune complexes described in this article underline the importance of understanding the immunogenicity induced by therapeutic proteins and the resulting influence on biological behavior.
Collapse
Affiliation(s)
- Eugenia Opolka-Hoffmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany.
| | - Martin R Edelmann
- Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, UK
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Therapeutic Modalities, CH-4070, Basel, Switzerland
| | - Michael B Otteneder
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | - Simon Hauri
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | - Gregor Jordan
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany
| | - Peter Schrag
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | - Martin Lechmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians University, DE-80539, Munich, Germany
| | - Roland F Staack
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany
| |
Collapse
|
5
|
Desai DC, Dherai AJ, Strik A, Mould DR. Personalized Dosing of Infliximab in Patients With Inflammatory Bowel Disease Using a Bayesian Approach: A Next Step in Therapeutic Drug Monitoring. J Clin Pharmacol 2023; 63:480-489. [PMID: 36458468 DOI: 10.1002/jcph.2189] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022]
Abstract
Although biological agents have revolutionized the management of inflammatory bowel diseases (IBDs), a significant proportion of patients show primary non-response or develop secondary loss of response. Therapeutic drug monitoring (TDM) is advocated to maintain the efficacy of biologic agents. Reactive TDM can rationalize the management of primary non-response and secondary loss of response and has shown to be more cost-effective compared with empiric dose escalation. Proactive TDM is shown to increase clinical remission and the durability of the response to a biologic agent. However, the efficacy of proactive and reactive TDM has been questioned in recent studies and meta-analyses. Hence, we need a different approach to TDM, which addresses inflammatory burden, the individual patient, and disease factors. Bayesian approaches, which use population pharmacokinetic models, enable clinicians to make better use of TDM for dose adjustment. With rapid improvement in computer technology, these Bayesian model-based software packages are now available for clinical use. Bayesian dashboard systems allow clinicians to apply model-based dosing to understand an individual's pharmacokinetics and achieve a target serum drug concentration. The model is updated using previously measured drug concentrations and relevant patient factors, such as body weight, C-reactive protein, and serum albumin concentration, to maintain effective drug concentrations in the serum. Initial studies have found utility for the Bayesian approach in induction and maintenance, in adult and pediatric patients, in clinical trials, and in real-life situations for patients with IBD treated with infliximab. This needs confirmation in larger studies. This article reviews the Bayesian approach to therapeutic drug monitoring in IBD.
Collapse
Affiliation(s)
- Devendra C Desai
- Division of Gastroenterology, PD Hinduja Hospital, Veer Savarkar Marg, Mahim, Mumbai, India
| | - Alpa J Dherai
- Department of Laboratory Medicine, PD Hinduja Hospital, Veer Savarkar Marg, Mahim, Mumbai, India
| | - Anne Strik
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, the Netherlands
| | - Diane R Mould
- Projections Research Inc., Phoenixville, Pennsylvania, USA
| |
Collapse
|
6
|
Lin YC, Chen BM, Tran TTM, Chang TC, Al-Qaisi TS, Roffler SR. Accelerated clearance by antibodies against methoxy PEG depends on pegylation architecture. J Control Release 2023; 354:354-367. [PMID: 36641121 DOI: 10.1016/j.jconrel.2023.01.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/07/2023] [Accepted: 01/08/2023] [Indexed: 01/16/2023]
Abstract
Methoxy polyethylene glycol (mPEG) is attached to many proteins, peptides, nucleic acids and nanomedicines to improve their biocompatibility. Antibodies that bind PEG are present in many individuals and can be generated upon administration of pegylated therapeutics. Anti-PEG antibodies that bind to the PEG "backbone" can accelerate drug clearance and detrimentally affect drug activity and safety, but no studies have examined how anti-methoxy PEG (mPEG) antibodies, which selectively bind the terminus of mPEG, affect pegylated drugs. Here, we investigated how defined IgG and IgM monoclonal antibodies specific to the PEG backbone (anti-PEG) or terminal methoxy group (anti-mPEG) affect pegylated liposomes or proteins with a single PEG chain, a single branched PEG chain, or multiple PEG chains. Large immune complexes can be formed between all pegylated compounds and anti-PEG antibodies but only pegylated liposomes formed large immune complexes with anti-mPEG antibodies. Both anti-PEG IgG and IgM antibodies accelerated the clearance of all pegylated compounds but anti-mPEG antibodies did not accelerate clearance of proteins with a single or branched PEG molecule. Pegylated liposomes were primarily taken up by Kupffer cells in the liver, but both anti-PEG and anti-mPEG antibodies directed uptake of a heavily pegylated protein to liver sinusoidal endothelial cells. Our results demonstrate that in contrast to anti-PEG antibodies, immune complex formation and drug clearance induced by anti-mPEG antibodies depends on pegylation architecture; compounds with a single or branched PEG molecule are unaffected by anti-mPEG antibodies but are increasingly affected as the number of PEG chain in a structure increases.
Collapse
Affiliation(s)
- Yi-Chen Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Trieu Thi My Tran
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tien-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Talal Salem Al-Qaisi
- Department of Medical Laboratory Sciences, Pharmacological and Diagnostic Research Centre, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Steve R Roffler
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
7
|
2021 White Paper on Recent Issues in Bioanalysis: TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparability & Cut Point Appropriateness ( Part 3 - Recommendations on Gene Therapy, Cell Therapy, Vaccine Assays; Immunogenicity of Biotherapeutics and Novel Modalities; Integrated Summary of Immunogenicity Harmonization). Bioanalysis 2022; 14:737-793. [PMID: 35578991 DOI: 10.4155/bio-2022-0081] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term "Context of Use - COU"); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and, critical reagent and positive control generation were the special features of the 15th edition. This 2021 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 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations on TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparability & Cut Point Appropriateness. Part 1A (Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC), Part 1B (Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine) and Part 2 (ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry) are published in volume 14 of Bioanalysis, issues 9 and 10 (2022), respectively.
Collapse
|
8
|
Ishiwatari-Ogata C, Kyuuma M, Ogata H, Yamakawa M, Iwata K, Ochi M, Hori M, Miyata N, Fujii Y. Ozoralizumab, a Humanized Anti-TNFα NANOBODY ® Compound, Exhibits Efficacy Not Only at the Onset of Arthritis in a Human TNF Transgenic Mouse but Also During Secondary Failure of Administration of an Anti-TNFα IgG. Front Immunol 2022; 13:853008. [PMID: 35273620 PMCID: PMC8902368 DOI: 10.3389/fimmu.2022.853008] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 12/19/2022] Open
Abstract
Although the introduction of tumor necrosis factor (TNF) inhibitors represented a significant advance in the treatment of rheumatoid arthritis (RA), traditional anti-TNFα antibodies are somewhat immunogenic, and their use results in the formation of anti-drug antibodies (ADAs) and loss of efficacy (secondary failure). Ozoralizumab is a trivalent, bispecific NANOBODY® compound that differs structurally from IgGs. In this study we investigated the suppressant effect of ozoralizumab and adalimumab, an anti-TNFα IgG, on arthritis and induction of ADAs in human TNF transgenic mice. Ozoralizumab markedly suppressed arthritis progression and did not induce ADAs during long-term administration. We also developed an animal model of secondary failure by repeatedly administering adalimumab and found that switching from adalimumab to ozoralizumab was followed by superior anti-arthritis efficacy in the secondary-failure animal model. Moreover, ozoralizumab did not form large immune complexes that might lead to ADA formation. The results of our studies suggest that ozoralizumab, which exhibited low immunogenicity in the animal model used and has a different antibody structure from that of IgGs, is a promising candidate for the treatment of RA patients not only at the onset of RA but also during secondary failure of anti-TNFα treatment.
Collapse
Affiliation(s)
| | - Masanao Kyuuma
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Hitoshi Ogata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Machi Yamakawa
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Katsuya Iwata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Motoki Ochi
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Miyuki Hori
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Noriyuki Miyata
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yasuyuki Fujii
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| |
Collapse
|
9
|
Suh K, Kyei I, Hage DS. Approaches for the detection and analysis of anti-drug antibodies to biopharmaceuticals: A review. J Sep Sci 2022; 45:2077-2092. [PMID: 35230731 DOI: 10.1002/jssc.202200112] [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: 02/10/2022] [Revised: 02/10/2022] [Accepted: 02/26/2022] [Indexed: 11/10/2022]
Abstract
Antibody-based therapeutic agents and other biopharmaceuticals are now used in the treatment of many diseases. However, when these biopharmaceuticals are administrated to patients, an immune reaction may occur that can reduce the drug's efficacy and lead to adverse side effects. The immunogenicity of biopharmaceuticals can be evaluated by detecting and measuring antibodies that have been produced against these drugs, or anti-drug antibodies (ADAs). Methods for ADA detection and analysis can be important during the selection of a therapeutic approach based on such drugs and is crucial when developing and testing new biopharmaceuticals. This review examines approaches that have been used for ADA detection, measurement, and characterization. Many of these approaches are based on immunoassays and antigen binding tests, including homogeneous mobility shift assays. Other techniques that have been used for the analysis of ADAs are capillary electrophoresis, reporter gene assays, surface plasmon resonance spectroscopy, and liquid chromatography-mass spectrometry. The general principles of each approach will be discussed, along with their recent applications with regards to ADA analysis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kyungah Suh
- Department of Chemistry, University of Nebraska-Lincoln
| | - Isaac Kyei
- Department of Chemistry, University of Nebraska-Lincoln
| | - David S Hage
- Department of Chemistry, University of Nebraska-Lincoln
| |
Collapse
|
10
|
Impact of anti-PEG antibody affinity on accelerated blood clearance of pegylated epoetin beta in mice. Biomed Pharmacother 2021; 146:112502. [PMID: 34891120 DOI: 10.1016/j.biopha.2021.112502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022] Open
Abstract
Antibodies that bind polyethylene glycol (PEG) can be induced by pegylated biomolecules and also exist in a significant fraction of healthy individuals who have never received pegylated medicines. The binding affinity of antibodies against PEG (anti-PEG antibodies) likely varies depending on if they are induced or naturally occurring. Anti-PEG antibodies can accelerate the clearance of pegylated medicines from the circulation, resulting in loss of drug efficacy, but it is unknown how accelerated blood clearance is affected by anti-PEG antibody affinity. We identified a panel of anti-PEG IgG and IgM antibodies with binding avidities ranging over several orders of magnitude to methoxy polyethylene glycol-epoetin beta (PEG-EPO), which is used to treat patients suffering from anemia. Formation of in vitro immune complexes between PEG-EPO and anti-PEG IgG or IgM antibodies was more obvious as antibody affinity increased. Likewise, high affinity anti-PEG antibodies produced greater accelerated blood clearance of PEG-EPO as compared to low affinity antibodies. The molar ratio of anti-PEG antibody to PEG-EPO that accelerates drug clearance in mice correlates with antibody binding avidity. Our study indicates that the bioactivity of PEG-EPO may be reduced due to rapid clearance in patients with either high concentrations of low affinity or low concentrations of high affinity anti-PEG IgG and IgM antibodies.
Collapse
|
11
|
Opolka-Hoffmann E, Jordan G, Otteneder M, Kieferle R, Lechmann M, Winter G, Staack RF. The impact of immunogenicity on therapeutic antibody pharmacokinetics: A preclinical evaluation of the effect of immune complex formation and antibody effector function on clearance. MAbs 2021; 13:1995929. [PMID: 34763611 PMCID: PMC8726625 DOI: 10.1080/19420862.2021.1995929] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The occurrence of an immune response against therapeutic proteins poses a major risk for the development of biologics and for successful treatment of patients. Generation of anti-drug antibodies (ADAs) can lead to formation of immune complexes (ICs), consisting of drug and ADAs, with potential impact on safety, efficacy and exposure. Here, we focus on the effects of IC formation, i.e., specific IC sizes, ADA and drug properties, on drug pharmacokinetics. Pre-formed IC preparations of an IgG1 drug (with wild type or with an ablated effector function at the Fc domain) and different ADA surrogates (directed against the complementarity-determining regions or Fc domain of the drug) were administered to rats and collected serum was analyzed to determine the total drug concentration. A combination of size-exclusion chromatography and ELISA enabled a size-specific evaluation of IC profiles in serum and their changes over time. Within five minutes, total drug concentration decreased by ~20–60% when the drug was complexed. Independent of the ADA surrogate and drug variant used, increasing IC size led to increased clearance. Comparing ICs formed with the same ADA surrogate but different IgG1 variants, we observed that complexed drug with a wildtype Fc domain showed faster clearance compared to immune effector function modified drug. Data generated in this study indicated that clearance of drug due to ADA generation is driven by size and structure of the formed ICs, but also by the immune effector functions of the Fc domains of IgGs. Abbreviations Ab: antibody, ADA: anti-drug antibody, AUC: area under the curve, Bi: biotin, CDR: complementary-determining region, cmax: maximal concentration, Dig: digoxigenin, ELISA: enzyme-linked immunosorbent assay, Fc: fragment crystallizable, FcRn: neonatal Fc receptor, HMW: high molecular weight, IC: immune complex, IC-QC: immune complex quality control, IgG: immunoglobulin G, mAb: monoclonal antibody, mADA: monoclonal ADA, pAb: polyclonal antibody, pADA: polyclonal ADA, PD: pharmacodynamics; PK: pharmacokinetic, QC: quality control, SEC: size-exclusion chromatography, WT: wildtype
Collapse
Affiliation(s)
- Eugenia Opolka-Hoffmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| | - Gregor Jordan
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| | - Michael Otteneder
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Robin Kieferle
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| | - Martin Lechmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-University, Munich, Germany
| | - Roland F Staack
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
12
|
Zheng S, Niu J, Geist B, Fink D, Xu Z, Zhou H, Wang W. A minimal physiologically based pharmacokinetic model to characterize colon TNF suppression and treatment effects of an anti-TNF monoclonal antibody in a mouse inflammatory bowel disease model. MAbs 2021; 12:1813962. [PMID: 32967523 PMCID: PMC7531524 DOI: 10.1080/19420862.2020.1813962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Biotherapeutic drugs against tumor necrosis factor (TNF) are effective treatments for moderate to severe inflammatory bowel disease (IBD). Here, we evaluated CNTO 5048, an antimurine TNF surrogate monoclonal antibody (mAb), in a CD45RBhigh adoptive T cell transfer mouse colitis model, which allows examination of the early immunological events associated with gut inflammation and the therapeutic effects. The study was designed to quantitatively understand the effects of IBD on CNTO 5048 disposition, the ability of CNTO 5048 to neutralize pathogenic TNF at the colon under disease conditions, and the impact of dosing regimen on CNTO 5048 treatment effect. CNTO 5048 and TNF concentrations in both mice serum and colon homogenate were also measured. Free TNF concentrations in colon, but not in serum, were shown to correlate well with the colon pharmacodynamic readout, such as the summed histopathology score and neutrophil score. A minimal physiologically based pharmacokinetic (mPBPK) model was developed to characterize CNTO 5048 PK and disposition, as well as colon soluble TNF target engagement (TE). The mPBPK/TE model reasonably captured the observed data and provided a quantitative understanding of an anti-TNF mAb on its colon TNF suppression and therapeutic effect in a physiologically relevant IBD animal model. These results also provided insights into the potential benefits of using induction doses for the treatment of IBD patients.
Collapse
Affiliation(s)
- Songmao Zheng
- Biologics Development Sciences, Janssen BioTherapeutics (JBIO) , Spring House, PA, USA
| | - Jin Niu
- Biologics Development Sciences, Janssen BioTherapeutics (JBIO) , Spring House, PA, USA.,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA
| | - Brian Geist
- Biologics Development Sciences, Janssen BioTherapeutics (JBIO) , Spring House, PA, USA
| | - Damien Fink
- Biologics Development Sciences, Janssen BioTherapeutics (JBIO) , Spring House, PA, USA
| | - Zhenhua Xu
- Clinical Pharmacology and Pharmacometrics, Quantitative Sciences , Spring House, PA, USA
| | - Honghui Zhou
- Clinical Pharmacology and Pharmacometrics, Quantitative Sciences , Spring House, PA, USA
| | - Weirong Wang
- Clinical Pharmacology and Pharmacometrics, Quantitative Sciences , Spring House, PA, USA
| |
Collapse
|
13
|
Doo C, Bao L, Shen K, Yang JF, Shen RR, Chan LS. Diacerein Alone and in Combination with Infliximab Suppresses the Combined Proinflammatory Effects of IL-17A, IL-22, Oncostatin M, IL-1A, and TNF-alpha in Keratinocytes: A Potential Therapeutic Option in Psoriasis. J Interferon Cytokine Res 2021; 41:302-306. [PMID: 34410877 DOI: 10.1089/jir.2021.0036] [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: 11/12/2022] Open
Abstract
Psoriasis is a chronic disorder characterized by a complex interplay between keratinocytes and inflammatory mediators. In a previous study, we evaluated diacerein's ability to diminish interleukin (IL)-1's proinflammatory effects on cultured primary human keratinocytes. In this study, we evaluated diacerein's ability to diminish the inflammatory effects of a cytokine mixture (CM) consisting of IL-17A, IL-22, oncostatin M, IL-1A, and tumor necrosis factor (TNF)-alpha on cultured primary human keratinocytes. These five cytokines have been previously shown to induce an in vivo-equivalent cell culture psoriasis model. We also evaluated diacerein's anti-inflammatory effects in comparison to and in combination with infliximab, a TNF-alpha inhibitor currently used in the treatment of psoriasis. We found 81 genes that were significantly (P < 0.05) dysregulated by CM compared to medium control. All three treatment groups (diacerein alone, infliximab alone, and diacerein plus infliximab) diminished the effects of CM on these genes, with the greatest effect seen with diacerein plus infliximab. Using enzyme-linked immunosorbent assay method on cell culture supernatant, we determined the protein concentration for five genes (IL-19, IL-6, CSF3, S100A8, and NAP-2) significantly (P < 0.05) upregulated by CM at the gene level. Diacerein alone diminished the effect of CM on the protein concentration of two genes, whereas diacerein plus infliximab diminished the effect of CM on the protein concentration of all the five genes. Based on these results, we conclude that diacerein alone or in combination with infliximab may have a therapeutic role in psoriasis by downregulating key inflammatory mediators.
Collapse
Affiliation(s)
- Caroline Doo
- Department of Dermatology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lei Bao
- Department of Dermatology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kui Shen
- Department of Dermatology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jessica F Yang
- Department of Dermatology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Rachel R Shen
- Department of Dermatology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lawrence S Chan
- Department of Dermatology, University of Illinois at Chicago, Chicago, Illinois, USA
- Medicine Service, Captain James Lovell FHCC, North Chicago, Illinois, USA
| |
Collapse
|
14
|
Gao Y, Chen Z, Yang C, Zhong D. Liquid chromatography-mass spectrometry method for the quantification of an anti-sclerostin monoclonal antibody in cynomolgus monkey serum. J Pharm Anal 2021; 11:472-479. [PMID: 34513123 PMCID: PMC8424367 DOI: 10.1016/j.jpha.2020.08.005] [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] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/01/2020] [Accepted: 08/09/2020] [Indexed: 11/20/2022] Open
Abstract
Liquid chromatography tandem mass spectrometry (LC-MS/MS) has gradually become a promising alternative to ligand binding assay for the bioanalysis of biotherapeutic molecules, due to its rapid method development and high accuracy. In this study, we established a new LC-MS/MS method for the determination of the anti-sclerostin monoclonal antibody (SHR-1222) in cynomolgus monkey serum, and compared it to a previous electrochemiluminescence method. The antibody was quantified by detecting the surrogate peptide obtained by trypsin digestion. The surrogate peptide was carefully selected by investigating its uniqueness, stability and MS response. The quantitative range of the proposed method was 2.00-500 μg/mL, and this verified method was successfully applied to the toxicokinetic assessment of SHR-1222 in cynomolgus monkey serum. It was found that the concentrations of SHR-1222 in cynomolgus monkeys displayed an excellent agreement between the LC-MS/MS and electrochemiluminescence methods (ratios of drug exposure, 0.8-1.0). Notably, two monkeys in the 60 mg/kg dose group had abnormal profiles with a low detection value of SHR-1222 in their individual sample. Combining the high-level anti-drug antibodies (ADAs) in these samples and the consistent quantitative results of the two methods, we found that the decreased concentration of SHR-1222 was due to the accelerated clearance mediated by ADAs rather than the interference of ADAs to the detection platform. Taken together, we successfully developed an accurate, efficient and cost-effective LC-MS/MS method for the quantification of SHR-1222 in serum samples, which could serve as a powerful tool to improve the preclinical development of antibody drugs.
Collapse
Affiliation(s)
- Yuxiong Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhendong Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Changyong Yang
- Preclinical Department, Hengrui Medicine Co., Ltd., Lianyungang, Jiangsu Province, 222047, China
| | - Dafang Zhong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
15
|
Mc Gettigan N, Afridi AS, Harkin G, Lardner C, Patchett S, Cheriyan D, Harewood G, Boland K, O'Toole A. The optimal management of anti-drug antibodies to infliximab and identification of anti-drug antibody values for clinical outcomes in patients with inflammatory bowel disease. Int J Colorectal Dis 2021; 36:1231-1241. [PMID: 33515082 DOI: 10.1007/s00384-021-03855-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE Secondary loss of response (LOR) to infliximab (IFX) commonly occurs. One cause is the development of anti-drug antibodies (ADAs). Evidence regarding the optimal management of ADAs is lacking. We aim to identify the best practice of management of ADAs to IFX to avoid discontinuation of therapy and to determine specific ADA cut-off values to determine pre-specified clinical outcomes. METHODS This is a 3-year study of patients receiving IFX who developed ADAs > 8μg/ml. We reviewed the management strategies and subsequent outcomes in patients who developed ADAs. RESULTS A total of 132 patients are included. Baseline characteristics include 54% male patients and mean age of 39.4 years. Fifty-two percent (n = 69) of patients discontinued IFX following the development of ADAs, 33.3% (n = 44) sited as secondary to LOR. Both an increase in IFX and adjustments to combination therapy were associated with lower rates of discontinuation of IFX vs no intervention (p value < 0.001, p value < 0.001). An increase in IFX resulted in a significant difference in ADAs/IFX trough levels pre- and post-intervention (p value < 0.001, p value = 0.032). ROC curve analysis yielded significant cut-off values for ADAs and treatment failure (ADA >16μg/ml, AUC 0.642, p value 0.003), steroid use (ADA >19 μg/ml, AUC 0.61, p value 0.048) development of infusion reactions (ADA> 37 μg/ml, AUC 0.68, p value 0.045) and switch to another biologic (ADA >45 μg/ml, AUC 0.739, p value <0.001). CONCLUSION Both escalation of IFX and combination therapy resulted in lower rates of LOR. ROC curve analysis identified significant cut-off values for ADA trough levels and important clinical outcomes.
Collapse
|
16
|
A cell based assay for evaluating binding and uptake of an antibody using hepatic nonparenchymal cells. Sci Rep 2021; 11:8383. [PMID: 33863984 PMCID: PMC8052349 DOI: 10.1038/s41598-021-87912-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/06/2021] [Indexed: 11/08/2022] Open
Abstract
Evaluation of the binding and uptake of an antibody in liver non-parenchymal cells (NPC), including liver sinusoidal endothelial cells, is important for revealing its pharmacokinetic (PK) behavior, since NPC has important roles in eliminating an antibody from the blood via the Fc fragment of IgG receptor IIB (FcγRIIB). However, there is currently no in vitro quantitative assay using NPC. This study reports on the development of a cell-based assay for evaluating the binding and uptake of such an antibody using liver NPC of mice and monkeys. In mice, the FcγRIIB-expressing cells were identified in the CD146-positive and CD45-negative fraction by flow cytometry. A titration assay was performed to determine the PK parameters, and the obtained parameter was comparable to that determined by the fitting of the in vivo PK. This approach was also extended to NPC from monkeys. The concentration-dependent binding and uptake was measured to determine the PK parameters using monkey NPC, the FcγRIIB-expressing fraction of which was identified by CD31 and CD45. The findings presented herein demonstrate that the in vitro liver NPC assay using flow cytometry is a useful tool to determine the binding and uptake of biologics and to predict the PK.
Collapse
|
17
|
Nagayasu M, Ozeki K. Combination of cassette-dosing and microsampling for reduced animal usage for antibody pharmacokinetics in cynomolgus monkeys, wild-type mice, and human FcRn transgenic mice. Pharm Res 2021; 38:583-592. [PMID: 33782838 DOI: 10.1007/s11095-021-03028-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/03/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE The aim of this study was to develop a useful antibody PK evaluation tool using a combination of cassette-dosing and microsampling in mice and monkeys in order to reduce the number of animals used. METHODS Cetuximab, denosumab, infliximab, and a mixture of the three antibodies, i.e., cassette-dosing, were administered intravenously to cynomolgus monkeys, C57BL/6J mice, and homozygous human neonatal Fc-receptor transgenic (Tg32) mice. Mouse blood was collected from one animal continuously via the jugular vein at nine points. RESULTS In cynomolgus monkeys, infliximab showed faster elimination in the cassette-dosing group than in the single-dose group. Anti-drug antibody production was observed, but the PK parameters of the clearance and distribution volume were similar in both groups. In C57BL/6J and Tg32 mice, each of the plasma concentrations-time profiles after cassette-dosing were similar to those after single dosing. PK evaluation using a combination of cassette-dosing and microsampling in mice may reduce the number of mice used by approximately 90% compared with the conventional method. CONCLUSIONS The combination of antibody cassette-dosing and microsampling is a promising PK evaluation method as a high-throughput and reliable with reduced numbers of mice and cynomolgus monkeys.
Collapse
Affiliation(s)
- Miho Nagayasu
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka, 412-8513, Japan
| | - Kazuhisa Ozeki
- Research Division, Chugai Pharmaceutical Co. Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka, 412-8513, Japan.
| |
Collapse
|
18
|
2020 White Paper on Recent Issues in Bioanalysis: Vaccine Assay Validation, qPCR Assay Validation, QC for CAR-T Flow Cytometry, NAb Assay Harmonization and ELISpot Validation ( Part 3 - Recommendations on Immunogenicity Assay Strategies, NAb Assays, Biosimilars and FDA/EMA Immunogenicity Guidance/Guideline, Gene & Cell Therapy and Vaccine Assays). Bioanalysis 2021; 13:415-463. [PMID: 33533276 DOI: 10.4155/bio-2021-0007] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The 14th edition of the Workshop on Recent Issues in Bioanalysis (14th WRIB) was held virtually on June 15-29, 2020 with an attendance of over 1000 representatives from pharmaceutical/biopharmaceutical companies, biotechnology companies, contract research organizations, and regulatory agencies worldwide. The 14th WRIB included three Main Workshops, seven Specialized Workshops that together spanned 11 days in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy and vaccine. Moreover, a comprehensive vaccine assays track; an enhanced cytometry track and updated Industry/Regulators consensus on BMV of biotherapeutics by LCMS were special features in 2020. As in previous years, this year's WRIB continued to gather a wide diversity of international industry opinion leaders and regulatory authority experts working on both small and large molecules to facilitate sharing and discussions focused on improving quality, increasing regulatory compliance and achieving scientific excellence on bioanalytical issues. This 2020 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop and is aimed to provide the Global 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 2020 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations on Vaccine, Gene/Cell Therapy, NAb Harmonization and Immunogenicity). Part 1 (Innovation in Small Molecules, Hybrid LBA/LCMS & Regulated Bioanalysis), Part 2A (BAV, PK LBA, Flow Cytometry Validation and Cytometry Innovation) and Part 2B (Regulatory Input) are published in volume 13 of Bioanalysis, issues 4 and 5 (2020), respectively.
Collapse
|
19
|
Alten R, Markland C, Boyce M, Kawakami K, Muniz R, Genovese MC. Immunogenicity of an adalimumab biosimilar, FKB327, and its reference product in patients with rheumatoid arthritis. Int J Rheum Dis 2020; 23:1514-1525. [PMID: 32852139 PMCID: PMC7754138 DOI: 10.1111/1756-185x.13951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022]
Abstract
Aim This study, FKB327‐003, is a phase 3, open‐label extension (OLE) study comparing the long‐term immunogenicity of an adalimumab biosimilar, FKB327 (F), with the reference product (RP). Methods In the OLE, patients completing 24 weeks of an initial randomized, double‐blind (DB) study (Period 1) with clinical response and no safety concerns were rerandomized to F or RP, so that two‐thirds of patients remained on the same treatment and one‐third switched to the alternate treatment for weeks 24 through 54 (OLE weeks 0‐30; Period 2), then all received F through week 100 (OLE week 76; Period 3). Treatment sequences were F‐F‐F (no switch), RP‐F‐F and RP‐RP‐F (single switch), and F‐RP‐F (double switch). Patients who entered the OLE study were evaluated for immunogenicity across switching sequences. Results The proportion of patients with positive antidrug antibody (ADA) status at the end of Period 1 was 61.7% and 60.0% for F and RP, respectively. The proportion of patients with positive ADA status did not increase throughout Period 1, and was similar for F and RP at all time points. At the end of Period 3, the proportion of patients with positive ADA status was lower in all treatment sequences, at 51.1%, 54.4%, 48.1%, and 42.5% for F‐F‐F, F‐RP‐F, RP‐F‐F, and RP‐RP‐F, respectively. Conclusion The RP and F showed comparable immunogenicity characteristics after long‐term administration. Development of ADAs with the RP and F was similar, and was not impacted by switching and double switching between F and RP treatment.
Collapse
|
20
|
A method combining blue native polyacrylamide gel electrophoresis with liquid chromatography tandem-mass spectrometry to detect circulating immune complexes between therapeutic monoclonal antibodies and anti-drug antibodies in animals. J Pharm Biomed Anal 2020; 186:113329. [PMID: 32371323 DOI: 10.1016/j.jpba.2020.113329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 11/24/2022]
Abstract
Therapeutic monoclonal antibodies can potentially induce unwanted immune responses, resulting in the production of anti-drug antibodies (ADAs). The binding of ADAs to drugs and subsequent formation of immune complexes (ICs) can trigger various responses, dependent on the size, concentration, and subclass of ADAs. To better understand the impact of ADAs on pharmacokinetics, pharmacodynamics, and toxicological profiles, a bioanalytical method was developed for the detection of ICs between human monoclonal immunoglobulin G (IgG) and ADAs in biological samples. Regarding the experimental procedure, in brief, the human antibody-specific ICs and unbound human antibody in biological samples are separated through blue native polyacrylamide gel electrophoresis (BN-PAGE). The target fractions are then cut from the gel, followed by in-gel trypsin digestion and subsequent liquid chromatography tandem-mass spectrometry (LC-MS/MS) to monitor the human IgG-specific peptide. This method was able to detect various types of human antibodies with a lower limit of detection of 10 μg/mL in monkey serum. The assay performance for the detection of ICs was demonstrated using spiked samples, and pre-incubated ICs in monkey serum were clearly detected. Taken together, these findings indicate that our method enables a semi-quantitative analysis for estimating the ratio of human antibody included ICs in comparison to the total antibody. This method was successfully applied to an in vivo study using mice, and the data helped explain the unexpectedly rapid clearance of a humanized antibody due to the formation of large ICs. The combination of the separation of ICs by BN-PAGE and the detection of the human IgG-specific peptide by LC-MS/MS is a useful general bioanalytical approach for the detection of ICs in animals.
Collapse
|
21
|
Boysen L, Viuff BM, Landsy LH, Lykkesfeldt J, Raymond JT, Price SA, Pelzer H, Lauritzen B. Formation and Glomerular Deposition of Immune Complexes in Mice Administered Human Antibodies: Evaluation of Dose, Frequency, and Biomarkers. Toxicol Pathol 2020; 48:570-585. [PMID: 32319353 DOI: 10.1177/0192623320919121] [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]
Abstract
Administration of human protein-based drugs to animals often leads to formation of antidrug antibodies (ADAs) that may form circulating immune complexes (CICs) with the dosed protein. Circulating immune complexes can activate and bind complement (cCICs), and if large amount of CICs or cCICs is formed, the clearance mechanism potentially becomes saturated, which can lead to immune complex (IC) deposition and inflammation. To obtain a better understanding of the underlying factors, including the relationship between different dose regimes on IC formation and deposition and identification of possible biomarkers of IC deposition and IC-related pathological changes in kidneys, BALB/c and C57BL/6J mice were administered with human anti-tumor necrosis factor α (aTNFα, adalimumab) or a humanized anti-TNP (aTNP) antibody for 13 weeks. Particularly, ADA, CIC, cCIC formation, IC deposition, and glomerulonephritis were observed in C57BL/6J administered with aTNFα, whereas the immunologic response was minor in BALB/c mice administered with aTNFα and in BALB/c and C57BL/6J mice administered aTNP. Changing dose levels or increasing dosing frequency of aTNFα on top of an already-established CIC and cCIC response did not lead to substantial changes in CIC, cCIC formation, or IC deposition. Finally, no association between the presence of CICs or cCIC in plasma and glomerular IC deposition and/or glomerulonephritis was observed.
Collapse
Affiliation(s)
- Lykke Boysen
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark.,Faculty of Health & Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Birgitte M Viuff
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| | - Lone H Landsy
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| | - Jens Lykkesfeldt
- Faculty of Health & Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - James T Raymond
- Pathology Associates, Charles River Laboratories Inc, Frederick, Maryland, USA
| | - Shari A Price
- Pathology Associates, Charles River Laboratories Inc, Frederick, Maryland, USA
| | - Hermann Pelzer
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| | - Brian Lauritzen
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
22
|
Doeleman MJH, van Maarseveen EM, Swart JF. Immunogenicity of biologic agents in juvenile idiopathic arthritis: a systematic review and meta-analysis. Rheumatology (Oxford) 2020; 58:1839-1849. [PMID: 30809664 PMCID: PMC6758589 DOI: 10.1093/rheumatology/kez030] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The clinical impact of anti-drug antibodies (ADAbs) in paediatric patients with JIA remains unknown. This systematic review and meta-analysis aimed to summarize the prevalence of ADAbs in JIA studies; investigate the effect of ADAbs on treatment efficacy and adverse events; and explore the effect of immunosuppressive therapy on antibody formation. METHODS PubMed, Embase and the Cochrane Library were systematically searched to identify relevant clinical trials and observational studies that reported prevalence of ADAbs. Studies were systematically reviewed in accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses and appropriate proportional and pairwise meta-analyses were performed. RESULTS A total of 5183 references were screened; 28 articles, involving 26 studies and 2354 JIA patients, met eligibility criteria. Prevalence of ADAbs ranged from 0% to 82% across nine biologic agents. Overall pooled prevalence of ADAbs was 16.9% (95% CI, 9.5, 25.9). Qualitative analysis of included studies indicated that antibodies to infliximab, adalimumab, anakinra and tocilizumab were associated with treatment failure and/or hypersensitivity reactions. Concomitant MTX uniformly reduced the risk of antibody formation during adalimumab treatment (risk ratio 0.33; 95% CI 0.21, 0.52). CONCLUSION The association of ADAbs with treatment failure and hypersensitivity reactions indicates their clinical relevance in paediatric patients with JIA. Based on our findings, we recommend a preliminary course of action regarding immunogenicity of biologic agents in patients with JIA. Further strategies to predict, prevent, detect and manage immunogenicity could optimize treatment outcomes and personalize treatment with biologic therapies.
Collapse
Affiliation(s)
- Martijn J H Doeleman
- Department of Paediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Centre Utrecht.,Faculty of Medicine, Utrecht University
| | - Erik M van Maarseveen
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Joost F Swart
- Department of Paediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Centre Utrecht.,Faculty of Medicine, Utrecht University
| |
Collapse
|
23
|
Atiqi S, Hooijberg F, Loeff FC, Rispens T, Wolbink GJ. Immunogenicity of TNF-Inhibitors. Front Immunol 2020; 11:312. [PMID: 32174918 PMCID: PMC7055461 DOI: 10.3389/fimmu.2020.00312] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/07/2020] [Indexed: 01/19/2023] Open
Abstract
Tumor necrosis factor inhibitors (TNFi) have significantly improved treatment outcome of rheumatic diseases since their incorporation into treatment protocols two decades ago. Nevertheless, a substantial fraction of patients experiences either primary or secondary failure to TNFi due to ineffectiveness of the drug or adverse reactions. Secondary failure and adverse events can be related to the development of anti-drug antibodies (ADA). The earliest studies that reported ADA toward TNFi mainly used drug-sensitive assays. Retrospectively, we recognize this has led to an underestimation of the amount of ADA produced due to drug interference. Drug-tolerant ADA assays also detect ADA in the presence of drug, which has contributed to the currently reported higher incidence of ADA development. Comprehension and awareness of the assay format used for ADA detection is thus essential to interpret ADA measurements correctly. In addition, a concurrent drug level measurement is informative as it may provide insight in the extent of underestimation of ADA levels and improves understanding the clinical consequences of ADA formation. The clinical effects are dependent on the ratio between the amount of drug that is neutralized by ADA and the amount of unbound drug. Pharmacokinetic modeling might be useful in this context. The ADA response generally gives rise to high affinity IgG antibodies, but this response will differ between patients. Some patients will not reach the phase of affinity maturation while others generate an enduring high titer high affinity IgG response. This response can be transient in some patients, indicating a mechanism of tolerance induction or B-cell anergy. In this review several different aspects of the ADA response toward TNFi will be discussed. It will highlight the ADA assays, characteristics and regulation of the ADA response, impact of immunogenicity on the pharmacokinetics of TNFi, clinical implications of ADA formation, and possible mitigation strategies.
Collapse
Affiliation(s)
- Sadaf Atiqi
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, Netherlands
| | - Femke Hooijberg
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, Netherlands
| | - Floris C Loeff
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Amsterdam, Netherlands
| | - Gerrit J Wolbink
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology, Reade, Amsterdam, Netherlands.,Department of Immunopathology, Sanquin Research and Landsteiner Laboratory Academic Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
24
|
Thomas PWA, Chin PKL, Barclay ML. A nationwide survey on therapeutic drug monitoring of anti-tumour necrosis factor agents for inflammatory bowel disease. Intern Med J 2020; 51:341-347. [PMID: 32043746 DOI: 10.1111/imj.14778] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Routine therapeutic drug monitoring (TDM) during treatment with anti-tumour necrosis factor (anti-TNF) agents in inflammatory bowel disease may increase treatment efficacy and cost-effectiveness, and reduce the risk of loss of response. AIMS To assess the current use of anti-TNF agent TDM, including trough concentration and anti-drug antibodies, among gastroenterology practitioners in New Zealand. METHODS A web-based survey was delivered to gastroenterologists and advanced trainees in New Zealand, identified by the New Zealand Society of Gastroenterology. RESULTS The response rate was 36% (48/134). Adalimumab was the most common initial anti-TNF agent used (78%, infliximab 22%). Ninety-three percent of those who completed the survey used TDM, mainly in cases of non-response or loss or response. Most respondents (93% and 83% for adalimumab and infliximab, respectively) measured trough concentrations within 24 h prior to the next administration. In patients in clinical remission but with endoscopic inflammation on anti-TNF agents, 72% would measure drug concentrations. In the presence of anti-drug antibodies, 45% would add an immunomodulator in patients with active disease and 47% would add an immunomodulator in patients in remission. With low trough concentrations, 77% would make no changes if the patient was in remission, and 75% would increase the dose in case of active disease. CONCLUSION TDM was routinely used among inflammatory bowel disease gastroenterology clinicians who responded to this survey. However, interpretation of results and decision-making is variable, suggesting more guidance is required.
Collapse
Affiliation(s)
| | - Paul K L Chin
- Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, New Zealand
| | - Murray L Barclay
- Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, New Zealand.,Department of Gastroenterology, Christchurch Hospital, Christchurch, New Zealand
| |
Collapse
|
25
|
Hanauer SB, Sandborn WJ, Feagan BG, Gasink C, Jacobstein D, Zou B, Johanns J, Adedokun OJ, Sands BE, Rutgeerts P, de Villiers WJS, Colombel JF, Ghosh S. IM-UNITI: Three-year Efficacy, Safety, and Immunogenicity of Ustekinumab Treatment of Crohn's Disease. J Crohns Colitis 2020; 14:23-32. [PMID: 31158271 DOI: 10.1093/ecco-jcc/jjz110] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Following induction/maintenance treatment in the UNITI/IM-UNITI studies of ustekinumab for Crohn's disease, patients entered a long-term extension for up to 5 years from induction. Efficacy through 152 and safety through 156 weeks are reported. METHODS At IM-UNITI Week 44, 567 ustekinumab-treated patients entered the long-term extension and continued to receive blinded subcutaneous ustekinumab on their assigned dose interval, without any subsequent dose adjustment. Placebo-treated patients discontinued after study unblinding [after IM-UNITI Week 44 analyses]. Efficacy data in the long-term extension [LTE] were collected every 12 weeks [q12w] before unblinding and then at q12w/q8w dosing visits. RESULTS Through Week 156, 29.6% of ustekinumab-treated patients discontinued. In an intent-to-treat analysis of randomised patients from IM-UNITI Weeks 0-152, 38.0% of ustekinumab induction responders receiving the drug q12w and 43.0% q8w were in remission at Week 152. Among patients entering the long-term extension in their original randomised groups, 61.9% of q12w and 69.5% of q8w patients were in remission at Week 152. Across all ustekinumab-treated patients [randomised and non-randomised] entering the long-term extension, remission rates at Week 152 were 56.3% and 55.1% for q12w and q8w, respectively. Safety events [per 100 patient-years] were similar among all ustekinumab-treated patients entering the long-term extension and placebo [overall adverse events 389.70 vs 444.17; serious adverse events, 18.97 vs 19.54; serious infections, 4.21 vs 3.97]. Rates of antibodies to ustekinumab through Week 156 remained low, 4.6% in all randomised ustekinumab-treated patients; lowest among patients in the original randomised q8w group [2/82, 2.4%]. CONCLUSIONS Continued treatment with subcutaneous ustekinumab maintained clinical response and remission through 3 years in a majority of patients who responded to induction therapy and was well-tolerated. ClinicalTrials.gov number NCT01369355.
Collapse
Affiliation(s)
- Stephen B Hanauer
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Brian G Feagan
- Robarts Clinical Trials, Western University, London, ON, Canada
| | | | | | - Bin Zou
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Jewel Johanns
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | | - Bruce E Sands
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Subrata Ghosh
- NIHR Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| |
Collapse
|
26
|
Failure of anti-TNF treatment in patients with rheumatoid arthritis: The pros and cons of the early use of alternative biological agents. Autoimmun Rev 2019; 18:102398. [DOI: 10.1016/j.autrev.2019.102398] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
|
27
|
Boysen L, Viuff BM, Landsy LH, Price SA, Raymond JT, Lykkesfeldt J, Lauritzen B. Formation and glomerular deposition of immune complexes in mice administered bovine serum albumin: Evaluation of dose, frequency, and biomarkers. J Immunotoxicol 2019; 16:191-200. [DOI: 10.1080/1547691x.2019.1680776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Lykke Boysen
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
- Faculty of Health & Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Birgitte M. Viuff
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| | - Lone H. Landsy
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| | | | | | - Jens Lykkesfeldt
- Faculty of Health & Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Brian Lauritzen
- Global Discovery & Development Sciences, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
28
|
Malik PRV, Edginton AN. Physiologically-Based Pharmacokinetic Modeling vs. Allometric Scaling for the Prediction of Infliximab Pharmacokinetics in Pediatric Patients. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:835-844. [PMID: 31343836 PMCID: PMC6875711 DOI: 10.1002/psp4.12456] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
Abstract
The comparative performances of physiologically‐based pharmacokinetic (PBPK) modeling and allometric scaling for predicting the pharmacokinetics (PKs) of large molecules in pediatrics are unknown. Therefore, both methods were evaluated for accuracy in translating knowledge of infliximab PKs from adults to children. PBPK modeling was performed using the base model for large molecules in PK‐Sim version 7.4 with modifications in Mobi. Eight population PK models from literature were reconstructed and scaled by allometry to pediatrics. Evaluation data included seven pediatric studies (~4–18 years). Both methods performed comparably with 66.7% and 68.6% of model‐predicted concentrations falling within twofold of the observed concentrations for PBPK modeling and allometry, respectively. Considerable variability was noted among the allometric models. Therefore, pediatric clinical trial planning would benefit from using approaches that require predictions depending on the specific question i.e., PBPK modeling and allometry.
Collapse
Affiliation(s)
- Paul R V Malik
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| |
Collapse
|
29
|
Generation, Characterization, and Quantitative Bioanalysis of Drug/Anti-drug Antibody Immune Complexes to Facilitate Dedicated In Vivo Studies. Pharm Res 2019; 36:129. [PMID: 31254106 PMCID: PMC6598957 DOI: 10.1007/s11095-019-2661-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Immunogenicity against biotherapeutics can lead to the formation of drug/anti-drug-antibody (ADA) immune complexes (ICs) with potential impact on safety and drug pharmacokinetics (PK). This work aimed to generate defined drug/ADA ICs, characterized by quantitative (bio) analytical methods for dedicated determination of IC sizes and IC profile changes in serum to facilitate future in vivo studies. METHODS Defined ICs were generated and extensively characterized with chromatographic, biophysical and imaging methods. Quantification of drug fully complexed with ADAs (drug in ICs) was performed with an acid dissociation ELISA. Sequential coupling of SEC and ELISA enabled the reconstruction of IC patterns and thus analysis of IC species in serum. RESULTS Characterization of generated ICs identified cyclic dimers, tetramers, hexamers, and larger ICs of drug and ADA as main IC species. The developed acid dissociation ELISA enabled a total quantification of drug fully complexed with ADAs. Multiplexing of SEC and ELISA allowed unbiased reconstruction of IC oligomeric states in serum. CONCLUSIONS The developed in vitro IC model system has been properly characterized by biophysical and bioanalytical methods. The specificity of the developed methods enable discrimination between different oligomeric states of ICs and can be bench marking for future in vivo studies with ICs.
Collapse
|
30
|
Martínez-Romero GJ, Alvariño A, Hinojosa E, Mora M, Oltra L, Maroto N, Ferrer I, Hinojosa MD, Hinojosa JE. Validation of a population pharmacokinetic model of adalimumab in a cohort of patients with inflammatory bowel disease. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2019; 111:431-436. [PMID: 31021170 DOI: 10.17235/reed.2019.5600/2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
BACKGROUND therapeutic monitoring of anti-TNF drugs and anti-drug antibody levels are useful for clinical decision-making, via the rationalization and optimization of the use of anti-TNF treatments. The objective of the present study was to validate the model of Ternant et al., in a cohort of patients with inflammatory bowel diseases (IBD). This model was originally established for patients with rheumatoid arthritis and was used in this study to optimize the adalimumab (ADA) dose and predict ADA trough levels (ATL). METHODS this study used concentration data points from 30 IBD patients who received ADA treatment between 2014 and 2015. A goodness-of-fit of the model was determined by evaluating the relationship between the observed ATL values and population model-predicted values (PRED) or individual model-predicted values (IPRED). RESULTS a total of 51 ADA concentration points were analyzed. The bias of the model was 2.39 (95% CI, 1.63-3.15) for PRED and 0.63 (95% CI, 0.23-1.03) for IPRED. The precision was 3.57 (95% CI, 2.90-4.13) and 1.53 (95% CI, 1.22-1.80), respectively. CONCLUSIONS therapeutic drug monitoring involving ATL may allow the optimization of the treatment of IBD patients. The validation results of the phamacokinectic (PK) model for ADA in IBD patients are inadequate. However, additional studies will strengthen the bias and precision of the model.
Collapse
|
31
|
Dong Y, Li P, Xu T, Bi L. Effective serum level of etanercept biosimilar and effect of antidrug antibodies on drug levels and clinical efficacy in Chinese patients with ankylosing spondylitis. Clin Rheumatol 2019; 38:1587-1594. [PMID: 30747393 DOI: 10.1007/s10067-018-04424-x] [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: 09/20/2018] [Revised: 12/03/2018] [Accepted: 12/28/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To investigate the effective serum level of etanercept biosimilar in Chinese patients with ankylosing spondylitis (AS) who achieve AS Disease Activity Score-C-reactive protein (ASDAS-CRP) < 2.1, and the effect of antidrug antibodies on drug levels and clinical efficacy. METHODS Our study enrolled 60 patients with AS who were treated with etanercept biosimilar. Serum and clinical data were collected at baseline and treatment weeks 4, 12, and 24. Drug levels and antidrug antibody levels were measured using an enzyme-linked immunosorbent assay while tumour necrosis factor (TNF)-α levels were measured using cytometric bead array. A receiver operating characteristic (ROC) curve was used to analyse effective serum level of etanercept biosimilar. RESULTS Patients with ASDAS-CRP ≥ 2.1 exhibited significantly lower drug levels than those with ASDAS-CRP < 2.1 did. The cut-off values of effective serum level of patients with AS who achieved ASDAS-CRP < 2.1 at weeks 4, 12, and 24 were 2.32, 2.12, and 2.36 μg/mL, respectively. Patients with drug levels above the cut-off value had lower Bath AS Disease Activity Index (BASDAI) and TNF-α levels. Antidrug antibodies had no effect on the Assessment of Spondylosis Arthritis International Society (ASAS) remission rates, but patients with antidrug antibodies had lower drug levels and higher TNF-α levels. CONCLUSIONS Detecting serum drug levels and antidrug antibody levels might facilitate estimation of the clinical efficacy and adjustment of medication regimen during etanercept biosimilar therapy in Chinese patients with AS.
Collapse
Affiliation(s)
- Yidian Dong
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Tingshuang Xu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China.
| |
Collapse
|
32
|
Manickam C, Shah SV, Lucar O, Ram DR, Reeves RK. Cytokine-Mediated Tissue Injury in Non-human Primate Models of Viral Infections. Front Immunol 2018; 9:2862. [PMID: 30568659 PMCID: PMC6290327 DOI: 10.3389/fimmu.2018.02862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Viral infections trigger robust secretion of interferons and other antiviral cytokines by infected and bystander cells, which in turn can tune the immune response and may lead to viral clearance or immune suppression. However, aberrant or unrestricted cytokine responses can damage host tissues, leading to organ dysfunction, and even death. To understand the cytokine milieu and immune responses in infected host tissues, non-human primate (NHP) models have emerged as important tools. NHP have been used for decades to study human infections and have played significant roles in the development of vaccines, drug therapies and other immune treatment modalities, aided by an ability to control disease parameters, and unrestricted tissue access. In addition to the genetic and physiological similarities with humans, NHP have conserved immunologic properties with over 90% amino acid similarity for most cytokines. For example, human-like symptomology and acute respiratory syndrome is found in cynomolgus macaques infected with highly pathogenic avian influenza virus, antibody enhanced dengue disease is common in neotropical primates, and in NHP models of viral hepatitis cytokine-induced inflammation induces severe liver damage, fibrosis, and hepatocellular carcinoma recapitulates human disease. To regulate inflammation, anti-cytokine therapy studies in NHP are underway and will provide important insights for future human interventions. This review will provide a comprehensive outline of the cytokine-mediated exacerbation of disease and tissue damage in NHP models of viral infections and therapeutic strategies that can aid in prevention/treatment of the disease syndromes.
Collapse
Affiliation(s)
- Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Spandan V. Shah
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Olivier Lucar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Daniel R. Ram
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - R. Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
33
|
van Schie KA, Kruithof S, Ooijevaar-de Heer P, Derksen NIL, van de Bovenkamp FS, Saris A, Vidarsson G, Bentlage AEH, Jiskoot W, Romeijn S, Koning RI, Bos E, Stork EM, Koeleman CAM, Wuhrer M, Wolbink G, Rispens T. Restricted immune activation and internalisation of anti-idiotype complexes between drug and antidrug antibodies. Ann Rheum Dis 2018; 77:1471-1479. [PMID: 29945923 DOI: 10.1136/annrheumdis-2018-213299] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/29/2018] [Accepted: 06/06/2018] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Therapeutic antibodies can provoke an antidrug antibody (ADA) response, which can form soluble immune complexes with the drug in potentially high amounts. Nevertheless, ADA-associated adverse events are usually rare, although with notable exceptions including infliximab. The immune activating effects and the eventual fate of these 'anti-idiotype' complexes are poorly studied, hampering assessment of ADA-associated risk of adverse events. We investigated the in vitro formation and biological activities of ADA-drug anti-idiotype immune complexes using patient-derived monoclonal anti-infliximab antibodies. METHODS Size distribution and conformation of ADA-drug complexes were characterised by size-exclusion chromatography and electron microscopy. Internalisation of and immune activation by complexes of defined size was visualised with flow imaging, whole blood cell assay and C4b/c ELISA. RESULTS Size and conformation of immune complexes depended on the concentrations and ratio of drug and ADA; large complexes (>6 IgGs) formed only with high ADA titres. Macrophages efficiently internalised tetrameric and bigger complexes in vitro, but not dimers. Corroborating these results, ex vivo analysis of patient sera demonstrated only dimeric complexes in circulation.No activation of immune cells by anti-idiotype complexes was observed, and only very large complexes activated complement. Unlike Fc-linked hexamers, anti-idiotype hexamers did not activate complement, demonstrating that besides size, conformation governs immune complex potential for triggering effector functions. CONCLUSIONS Anti-idiotype ADA-drug complexes generally have restricted immune activation capacity. Large, irregularly shaped complexes only form at high concentrations of both drug and ADA, as may be achieved during intravenous infusion of infliximab, explaining the rarity of serious ADA-associated adverse events.
Collapse
Affiliation(s)
- Karin A van Schie
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone Kruithof
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Pleuni Ooijevaar-de Heer
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ninotska I L Derksen
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Fleur S van de Bovenkamp
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anno Saris
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arthur E H Bentlage
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Wim Jiskoot
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Stefan Romeijn
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Roman I Koning
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik Bos
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eva Maria Stork
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolien A M Koeleman
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gertjan Wolbink
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Vahle JL. Immunogenicity and Immune Complex Disease in Preclinical Safety Studies. Toxicol Pathol 2018; 46:1013-1019. [DOI: 10.1177/0192623318797070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article summarizes a continuing education presentation on immunogenicity that was part of a continuing education course entitled, “Clinical Pathology of Biotherapeutics.” Immunogenicity of a biotherapeutic can have diverse impacts including altered systemic exposure and pharmacologic responses and, in a fraction of the cases, safety concerns including cross-reactive neutralization of endogenous proteins or sequela related to immune complex disease (ICD). In most cases, immune complexes are readily cleared from circulation; however, based on physiochemical properties, insoluble complexes form, activate complement, and deposit in tissues. Using published information and personal experience, a set of repeat-dose monkey toxicity studies with manifestations suggestive of ICD was reviewed to summarize the spectrum of clinical and pathology findings. The most common live-phase observation linked to ICD was an acute postdosing reaction following multiple dose administrations characterized by generalized collapse and attributed to acute complement activation. Less common live-phase observations were related to syndromes such as a consumptive coagulopathy or a protein losing nephropathy. The most common histologic change attributed to ICD was multi-organ vascular/perivascular inflammation followed by glomerulonephritis. The presentation concluded with a description of the challenges in assessing the relevance of immunogenicity-related reaction in monkey to human clinical use.
Collapse
Affiliation(s)
- John L. Vahle
- Lilly Research Laboratories, Indianapolis, Indiana, USA
| |
Collapse
|
35
|
Beaumont M, Tomazela D, Hodges D, Ermakov G, Hsieh E, Figueroa I, So OY, Song Y, Ma H, Antonenko S, Mengesha W, Zhang YW, Zhang S, Hseih S, Ayanoglu G, Du X, Rimmer E, Judo M, Vives F, Yearley JH, Moon C, Manibusan A, Knudsen N, Beck A, Bresson D, Gately D, Neupane D, Escandón E. Antibody-drug conjugates: integrated bioanalytical and biodisposition assessments in lead optimization and selection. AAPS OPEN 2018. [DOI: 10.1186/s41120-018-0026-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
36
|
Frazier KS, Obert LA. Drug-induced Glomerulonephritis: The Spectre of Biotherapeutic and Antisense Oligonucleotide Immune Activation in the Kidney. Toxicol Pathol 2018; 46:904-917. [PMID: 30089413 DOI: 10.1177/0192623318789399] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prevalence of immune-mediated glomerulonephritis has increased in preclinical toxicity studies, with more frequent use of biotherapeutic agents (especially antigenic humanized molecules) and antisense oligonucleotide (ASO) therapies. Immune complex disease affects a small number of study monkeys, often correlates with antidrug antibody (ADA) titers, and occurs at a dose that favors immune complex formation or impedes clearance. While preclinical glomerulonephritis often fails to correlate with evidence of glomerular or vascular injury in human clinical trials and is not considered predictive, additional animal investigative immunohistochemical work may be performed to substantiate evidence for immune complex pathogenesis. While ADA is most commonly encountered as a predisposing factor with biotherapeutic agents, complement activation may occur without circulating complexes, and other mechanisms of non-ADA immune-mediated glomerulonephritis have been observed including nonendogenous immune aggregates and immunoregulatory pharmacology. Although glomerulonephritis associated with oligonucleotide therapies has been noted occasionally in preclinical studies and more rarely with human patients, pathophysiologic mechanisms involved appear to be different between species and preclinical cases are not considered predictive for humans. ADA is not involved in oligonucleotide-associated cases, and complement fixation plays a more important role in monkeys. Recent screening of ASOs for proinflammatory activity appears to have decreased glomerulonephritis incidence preclinically.
Collapse
|
37
|
Sensitivity and drug tolerance of antidrug antibody assays in relation to positive control characteristics. Bioanalysis 2018; 10:1289-1306. [DOI: 10.4155/bio-2018-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Detection and characterization of antidrug antibodies (ADA) play a key role in understanding the relation of ADA with safety and efficacy. Positive controls (PCs) are used to estimate the sensitivity and assay sensitivity in the presence of drug (drug tolerance) of assays to detect ADA. We investigated a number of factors that may drive sensitivity and drug tolerance. Results: We found no correlation between affinity and sensitivity and sensitivity in the presence of drug in multiple assays with two antibody–drug conjugates. Multiple factors that influenced sensitivity and drug tolerance were observed, yet these had limited overall predictive value for all investigated assay formats, PCs and compounds.Conclusion: Assay sensitivity and drug tolerance as studied here, are likely driven by multiple factors such as cut point confidence level, stoichiometry of PC–drug complexes and presentation of epitopes.
Collapse
|
38
|
Sanchez-Hernandez JG, Rebollo N, Munoz F, Martin-Suarez A, Calvo MV. Therapeutic drug monitoring of tumour necrosis factor inhibitors in the management of chronic inflammatory diseases. Ann Clin Biochem 2018; 56:28-41. [PMID: 29807436 DOI: 10.1177/0004563218782286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumour necrosis factor inhibitor therapy has drastically changed the management of chronic inflammatory diseases. Some important drawbacks that can cause loss of response during treatment with these drugs are related to their large individual variability, the disease burden and the formation of antidrug antibodies that increase its clearance. Therapeutic drug monitoring of these drugs is not yet recommended by all scientific societies, and if so, only in patients with inflammatory symptoms. Proactive therapeutic drug monitoring represents a new strategy with many potential clinical benefits, including the prevention of immunogenicity, a reduction in the need for rescue therapy and greater durability of tumour necrosis factor inhibitor treatment. The review is based on a systematic search of the literature for controlled trials, systematic reviews, experimental studies, guideline papers and cohort studies addressing the best practice in tumour necrosis factor inhibitor therapeutic drug monitoring. Although there is ample evidence supporting the use of therapeutic drug monitoring in clinical practice to achieve better outcomes, some challenges have been detected. Many studies are focused on finding solutions for the lack of standardization of analytical methods to measure tumour necrosis factor inhibitor and antidrug antibodies concentrations. Other challenges are development of effective cost-saving proactive algorithms to identify optimal drug concentrations and the research on the role of antidrug antibodies, especially in the management and prevention of loss of response. Therapeutic drug monitoring of tumour necrosis factor inhibitor offers a rational approach to the optimization of the treatment of chronic inflammatory disease. Although prospective controlled trials yield little conclusive evidence of its benefits, there is growing acceptance of its value in clinical practice.
Collapse
Affiliation(s)
- J G Sanchez-Hernandez
- 1 Pharmacy Service, University Hospital of Salamanca, Salamanca, Spain.,2 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain.,3 Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
| | - N Rebollo
- 1 Pharmacy Service, University Hospital of Salamanca, Salamanca, Spain.,2 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain.,3 Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
| | - F Munoz
- 3 Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain.,4 Gastroenterology Service, University Hospital of Salamanca, Salamanca, Spain
| | - A Martin-Suarez
- 2 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain.,3 Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
| | - M V Calvo
- 1 Pharmacy Service, University Hospital of Salamanca, Salamanca, Spain.,2 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain.,3 Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
| |
Collapse
|
39
|
McSweeney MD, Wessler T, Price LSL, Ciociola EC, Herity LB, Piscitelli JA, Zamboni WC, Forest MG, Cao Y, Lai SK. A minimal physiologically based pharmacokinetic model that predicts anti-PEG IgG-mediated clearance of PEGylated drugs in human and mouse. J Control Release 2018; 284:171-178. [PMID: 29879519 DOI: 10.1016/j.jconrel.2018.06.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/18/2018] [Accepted: 06/02/2018] [Indexed: 10/14/2022]
Abstract
Circulating antibodies that specifically bind polyethylene glycol (PEG), a polymer routinely used in protein and nanoparticle therapeutics, have been associated with reduced efficacy and increased adverse reactions to some PEGylated therapeutics. In addition to acute induction of anti-PEG antibodies (APA) by PEGylated drugs, typically low but detectable levels of APA are also found in up to 70% of the general population. Despite the broad implications of APA, the dynamics of APA-mediated clearance of PEGylated drugs, and why many patients continue to respond to PEGylated drugs despite the presence of pre-existing APA, remains not well understood. Here, we developed a minimal physiologically based pharmacokinetic (mPBPK) model that incorporates various properties of APA and PEGylated drugs. Our mPBPK model reproduced clinical PK data of APA-mediated accelerated blood clearance of pegloticase, as well as APA-dependent elimination of PEGyated liposomes in mice. Our model predicts that the prolonged circulation of PEGylated drugs will be compromised only at APA concentrations greater than ~500 ng/mL, providing a quantitative explanation to why the effects of APA on PEGylated treatments appear to be limited in most patients. This mPBPK model is readily adaptable to other PEGylated drugs and particles to predict the precise levels of APA that could render them ineffective, providing a powerful tool to support the development and interpretation of preclinical and clinical studies of various PEGylated therapeutics.
Collapse
Affiliation(s)
- M D McSweeney
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - T Wessler
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA.
| | - L S L Price
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - E C Ciociola
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - L B Herity
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - J A Piscitelli
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - W C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - M G Forest
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA.
| | - Y Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - S K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
40
|
Husar E, Solonets M, Kuhlmann O, Schick E, Piper-Lepoutre H, Singer T, Tyagi G. Hypersensitivity Reactions to Obinutuzumab in Cynomolgus Monkeys and Relevance to Humans. Toxicol Pathol 2018; 45:676-686. [PMID: 28830332 DOI: 10.1177/0192623317723539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Obinutuzumab (GA101, Gazyva™, Gazyvaro®, F. Hoffmann-La Roche AG, Basel, Switzerland) is a humanized, glycoengineered type II antibody targeted against CD20. The preclinical safety evaluation required to support clinical development and marketing authorization of obinutuzumab included repeat-dose toxicity studies in cynomolgus monkeys for up to 6-month dosing with a 9-month recovery period. Results from those studies showed decreases in circulating B cells and corresponding B-cell depletion in lymphoid tissues, consistent with the desired pharmacology of obinutuzumab. Hypersensitivity reactions were noted at all doses in the 6-month study and were attributed to the foreign recognition of the drug construct in cynomolgus monkeys. Findings in monkeys were classified as acute hypersensitivity reactions that were evident immediately after dosing, such as excessive salivation, erythema, pruritus, irregular respiration, or ataxia, or chronic hypersensitivity reactions characterized by glomerulonephritis, arteritis/periarteritis, and inflammation in several tissues including serosal/adventitial inflammation. Immune complex deposits were demonstrated in tissues by immunohistochemistry, immunofluorescence, and electron microscopy. Some of, but not all, the animals that developed these reactions had detectable antidrug antibodies or circulating immune complexes accompanied by loss of drug exposure and pharmacodynamic effect. On the basis of clinical evidence to date, hypersensitivity reactions following obinutuzumab are rare, further supporting the general view that incidence and manifestation of immunogenicity in nonclinical species are generally not predictive for humans.
Collapse
Affiliation(s)
- Elisabeth Husar
- 1 Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Maria Solonets
- 2 Safety Science, F. Hoffmann-La Roche, Basel, Switzerland
| | - Olaf Kuhlmann
- 1 Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland.,3 Boehringer Ingelheim, Ingelheim, Germany
| | - Eginhard Schick
- 1 Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | | | - Thomas Singer
- 1 Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Gaurav Tyagi
- 4 Pharmaceutical Sciences, Roche Innovation Center New York, New York, New York, USA
| |
Collapse
|
41
|
Wang W, Leu J, Watson R, Xu Z, Zhou H. Investigation of the Mechanism of Therapeutic Protein-Drug Interaction Between Methotrexate and Golimumab, an Anti-TNFα Monoclonal Antibody. AAPS JOURNAL 2018; 20:63. [PMID: 29667047 DOI: 10.1208/s12248-018-0219-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/12/2018] [Indexed: 12/31/2022]
Abstract
A prominent example of human therapeutic protein-drug interaction (TP-DI) is between methotrexate (MTX) and anti-TNFα mAbs. One plausible mechanism for this TP-DI is through the pharmacodynamic effect of MTX on immunogenicity. However, there is no definitive evidence to substantiate this mechanism, and other competing hypotheses, such as MTX suppressing FcγRI expression thereby affecting mAb PK, have also been proposed. In order to understand this mechanism, a cynomolgus monkey study was conducted using golimumab as a model compound. Golimumab elicited high incidences of immunogenicity in healthy cynomolgus monkeys. Concomitant dosing of MTX delayed the onset and reduced the magnitude of anti-drug antibody (ADA) formation. The impact of MTX on golimumab PK correlated with the ADA status. Prior to ADA formation, MTX has no discernable effect on golimumab PK. Additionally, no alteration in FcγRI expression was observed following MTX treatment. The impact of MTX on golimumab immunogenicity and PK has been observed in patients with rheumatoid arthritis, psoriatic arthritis (PsA), and ankylosing spondylitis. In a representative phase 3 study of golimumab in patients with PsA, patients not receiving concomitant MTX was reported to have ~ 30% lower steady-state trough golimumab levels compared to those who received MTX. However, further analysis showed that PsA patients who were negative for ADA in both treatment groups had comparable trough levels of golimumab. Taken together, our results suggest that the mechanism of TP-DI between MTX and golimumab can mostly be attributed to the pharmacodynamic effect of MTX, i.e., the lowering of immunogenicity and immunogenicity-mediated clearance of mAbs.
Collapse
Affiliation(s)
- Weirong Wang
- Biologics Development Sciences, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Jocelyn Leu
- Global Clinical Pharmacology, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Rebecca Watson
- Biologics Development Sciences, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Zhenhua Xu
- Global Clinical Pharmacology, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Honghui Zhou
- Global Clinical Pharmacology, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA.
| |
Collapse
|
42
|
Abstract
During the development of monoclonal antibodies (mAbs) and other therapeutic proteins, immunogenicity, in particular the induction of anti-drug antibodies (ADAs), is an important concern, and thus immunogenicity assessment is a requirement for their approval. Establishment of appropriate methods for detecting and characterizing ADAs is necessary for immunogenicity assessment, but the lack of commonly available reference standards makes it difficult to compare and evaluate the methods. It is also difficult to compare the data with those obtained by other methods or facilities without reference standards. Here, we developed a panel of ADAs against anti-CD20 rituximab (Rituxan®, MabThera®); the panel consisted of eight clones of recombinant human-rat chimeric mAbs that target rituximab. The anti-rituximab mAbs showed different binding properties (specificity, epitope and affinity), and different neutralization potencies for CD20 binding, complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity. The molecular size of the immune complex consisting of rituximab and the anti-rituximab mAb differed among the clones, and was well correlated with their level of Fcγ-receptor activation. These results suggest that the ADAs chosen for the newly developed panel are suitable surrogates for human ADAs, which exhibit different potential to affect the efficacy and safety of rituximab. Next, we used this panel to compare several ADA-detecting assays and revealed that the assays had different abilities to detect the ADAs with different binding characteristics. We conclude that our panel of ADAs against rituximab will be useful for the future development and characterization of assays for immunogenicity assessment.
Collapse
Affiliation(s)
- Minoru Tada
- a Division of Biological Chemistry and Biologicals , National Institute of Health Sciences , Kawasaki, Kanagawa , Japan
| | - Takuo Suzuki
- a Division of Biological Chemistry and Biologicals , National Institute of Health Sciences , Kawasaki, Kanagawa , Japan
| | - Akiko Ishii-Watabe
- a Division of Biological Chemistry and Biologicals , National Institute of Health Sciences , Kawasaki, Kanagawa , Japan
| |
Collapse
|
43
|
van Hoeve K, Hoffman I, Vermeire S. Therapeutic drug monitoring of anti-TNF therapy in children with inflammatory bowel disease. Expert Opin Drug Saf 2017; 17:185-196. [PMID: 29202588 DOI: 10.1080/14740338.2018.1413090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Karen van Hoeve
- Department of Pediatric Gastroenterology, University Hospitals Leuven, Leuven, Belgium
| | - Ilse Hoffman
- Department of Pediatric Gastroenterology, University Hospitals Leuven, Leuven, Belgium
| | - Severine Vermeire
- Department of Gastroenterology & Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
44
|
Mitrev N, Vande Casteele N, Seow CH, Andrews JM, Connor SJ, Moore GT, Barclay M, Begun J, Bryant R, Chan W, Corte C, Ghaly S, Lemberg DA, Kariyawasam V, Lewindon P, Martin J, Mountifield R, Radford-Smith G, Slobodian P, Sparrow M, Toong C, van Langenberg D, Ward MG, Leong RW. Review article: consensus statements on therapeutic drug monitoring of anti-tumour necrosis factor therapy in inflammatory bowel diseases. Aliment Pharmacol Ther 2017; 46:1037-1053. [PMID: 29027257 DOI: 10.1111/apt.14368] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 08/06/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) in inflammatory bowel disease (IBD) patients receiving anti-tumour necrosis factor (TNF) agents can help optimise outcomes. Consensus statements based on current evidence will help the development of treatment guidelines. AIM To develop evidence-based consensus statements for TDM-guided anti-TNF therapy in IBD. METHODS A committee of 25 Australian and international experts was assembled. The initial draft statements were produced following a systematic literature search. A modified Delphi technique was used with 3 iterations. Statements were modified according to anonymous voting and feedback at each iteration. Statements with 80% agreement without or with minor reservation were accepted. RESULTS 22/24 statements met criteria for consensus. For anti-TNF agents, TDM should be performed upon treatment failure, following successful induction, when contemplating a drug holiday and periodically in clinical remission only when results would change management. To achieve clinical remission in luminal IBD, infliximab and adalimumab trough concentrations in the range of 3-8 and 5-12 μg/mL, respectively, were deemed appropriate. The range may differ for different disease phenotypes or treatment endpoints-such as fistulising disease or to achieve mucosal healing. In treatment failure, TDM may identify mechanisms to guide subsequent decision-making. In stable clinical response, TDM-guided dosing may avoid future relapse. Data indicate drug-tolerant anti-drug antibody assays do not offer an advantage over drug-sensitive assays. Further data are required prior to recommending TDM for non-anti-TNF biological agents. CONCLUSION Consensus statements support the role of TDM in optimising anti-TNF agents to treat IBD, especially in situations of treatment failure.
Collapse
|
45
|
Wong U, Cross RK. Primary and secondary nonresponse to infliximab: mechanisms and countermeasures. Expert Opin Drug Metab Toxicol 2017; 13:1039-1046. [PMID: 28876147 DOI: 10.1080/17425255.2017.1377180] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Primary and secondary non-response to infliximab are common in patients with inflammatory bowel disease and remain a management challenge in clinical practice. Areas covered: This article describes the epidemiology, mechanisms and risk factors for primary and secondary nonresponse to infliximab in patients with inflammatory bowel disease. Data on proactive and reactive therapeutic drug monitoring are examined in this review. An algorithm for evaluation and management of non-response to infliximab is provided. Preventative measures are also discussed. Relevant articles were identified after a literature search using PubMed. Search terms included 'infliximab', 'loss of response', 'immunogenicity', and 'drug monitoring'. References of identified articles were also reviewed to identify additional references. Expert opinion: A common cause for primary and secondary non-response include inadequate dosing of infliximab; inadequate dosing can be identified through assessment of drug and anti-drug antibody levels. Therapeutic drug monitoring should be done in patients losing response to infliximab. Use of drug monitoring proactively is still under debate.
Collapse
Affiliation(s)
- Uni Wong
- a Department of Medicine, Division of Gastroenterology and Hepatology , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Raymond K Cross
- a Department of Medicine, Division of Gastroenterology and Hepatology , University of Maryland School of Medicine , Baltimore , MD , USA
| |
Collapse
|
46
|
Kronenberg S, Husar E, Schubert C, Freichel C, Emrich T, Lechmann M, Giusti AM, Regenass F. Comparative assessment of immune complex-mediated hypersensitivity reactions with biotherapeutics in the non-human primate: Critical parameters, safety and lessons for future studies. Regul Toxicol Pharmacol 2017. [DOI: 10.1016/j.yrtph.2017.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Liefferinckx C, Minsart C, Toubeau JF, Cremer A, Amininejad L, Quertinmont E, Devière J, Gils A, van Gossum A, Franchimont D. Infliximab Trough Levels at Induction to Predict Treatment Failure During Maintenance. Inflamm Bowel Dis 2017; 23:1371-1381. [PMID: 28498153 DOI: 10.1097/mib.0000000000001120] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Infliximab (IFX) is indicated for the treatment of inflammatory bowel diseases (IBD). Nevertheless, loss of response (LOR) to IFX is reported in up to 10% to 30% of patients within the first year of treatment. Our objective was to evaluate the impact of the pharmacokinetics of IFX at induction on treatment failure. METHODS This is a longitudinal cohort study on 269 patients with IBD treated with IFX in a single center. A total of 2331 blood samples were prospectively collected from 2007 until March 2015 with a retrospective analysis of clinical data. IFX trough levels (TLs) were measured by enzyme-linked immunosorbent assay. Antibodies to IFX were measured by drug-sensitive bridging assay. RESULTS During follow-up, patients were defined according to treatment outcome. At week 6, median IFX TL in patients requiring a switch to another treatment due to LOR (LOR switched group) (2.32 μg/mL [0.12-19.93 μg/mL]) was lower than in patients with long-term response (long-term responders) (8.66 μg/mL [0.12-12.09 μg/mL], P = 0.007) and in patients responding to optimization (LOR optimized group) (7.28 μg/mL [0.17-14.91 μg/mL], P = 0.021). At week 2, median IFX TL was lower in the LOR switched group (5.7 μg/mL [0.15-12.09 μg/mL]) compared with the long-term responders (11.92 μg/mL [0.14-19.93 μg/mL], P = 0.041) but no significant difference was reached with the LOR optimized group (11.91 μg/mL [0.23-12.09 μg/mL], P = 0.065). In the LOR switched group, median IFX TL at induction (weeks 2 and 6) was significantly lower when patients had been previously exposed to anti-tumor necrosis factor compared with naive patients (0.91 μg/mL [0.12-4.4 μg/mL] versus 6.6 μg/mL [0.15-19.93 μg/mL], P = 0.044). CONCLUSIONS This study suggests that patients who do not respond to any optimization strategy have lower IFX TLs during induction at week 6. IFX TLs measured early on at induction might predict treatment failure to IFX during maintenance.
Collapse
Affiliation(s)
- Claire Liefferinckx
- *Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium; †Electrical Power Engineering Unit, Université de Mons, Mons, Belgium; ‡Department of Gastroenterology, Erasme Hospital, Brussels, Belgium; and §Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mitrev N, Leong RW. Therapeutic drug monitoring of anti-tumour necrosis factor-α agents in inflammatory bowel disease. Expert Opin Drug Saf 2016; 16:303-317. [PMID: 27922765 DOI: 10.1080/14740338.2017.1269169] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Anti-TNFα therapy has revolutionised treatment of inflammatory bowel disease, however primary non-response and secondary loss of response are a significant problem. Therapeutic drug monitoring (TDM) has recently emerged as a means of optimising use of anti-TNFα agents. Areas covered: TDM of anti-TNFα agents can guide clinical decisions during treatment failure events, prevent treatment failure events, and potentially result in significant healthcare cost saving. TDM for anti-TNFα agent involves measurement of drug levels and anti-drug antibodies, and can be performed reactively or proactively. Reactive TDM reserves testing for treatment failure events, while proactive TDM also consists of periodic TDM for patients responding to anti-TNFα therapy to allow treatment optimisation. Generation of anti-drug antibodies is recognised as one important mechanism of treatment failure and adverse events. Expert opinion: Evidence strongly supports TDM at time of treatment failure, while studies employing proactive TDM have demonstrated conflicting results. TDM can also help better select patients likely to remain in clinical remission on anti-TNFα treatment interruption. Currently TDM is used to optimise anti-TNFα treatment, but it is not used by most clinicians to prevent adverse reactions to anti-TNFα agents.
Collapse
Affiliation(s)
- Nikola Mitrev
- a Concord Hospital IBD Service, Department of Gastroenterology , Concord Repatriation General Hospital , Sydney , Australia
| | - Rupert W Leong
- a Concord Hospital IBD Service, Department of Gastroenterology , Concord Repatriation General Hospital , Sydney , Australia
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW There are an expanding number of therapies available to treat pediatric inflammatory bowel disease (IBD). As pediatric gastroenterologists attempt to achieve complete intestinal mucosal healing for their patients, it has become more important to gain an understanding of how to maximize the efficacy of our medications while minimizing their toxicities. We aim to provide an overview of therapeutic drug monitoring in IBD with an emphasis on the biologic therapies (antitumor necrosis factor and anti-integrin monoclonal antibodies). RECENT FINDINGS Recent findings do support optimized drug dosing for infliximab based on early trough levels, but question the utility of checking these values in patients doing well in maintenance therapy. Patients with severe colonic inflammation may be at increased risk for needing optimization with dose escalation because of medication loss in the stool. Dose escalation can recapture response in some patients with a secondary loss of response, including those with low level antibody formation. The monitoring of nontrough drug levels to allow timelier dose adjustment as well as the role of drug monitoring with anti-integrin therapy are areas of active research. SUMMARY Therapeutic drug monitoring is an effective strategy in the management of pediatric IBD that can help patients achieve mucosal healing and aid the clinical decision-making of the pediatric gastroenterologist.
Collapse
|
50
|
Myzithras M, Bigwarfe T, Li H, Waltz E, Ahlberg J, Giragossian C, Roberts S. Utility of immunodeficient mouse models for characterizing the preclinical pharmacokinetics of immunogenic antibody therapeutics. MAbs 2016; 8:1606-1611. [PMID: 27598372 DOI: 10.1080/19420862.2016.1229721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Prior to clinical studies, the pharmacokinetics (PK) of antibody-based therapeutics are characterized in preclinical species; however, those species can elicit immunogenic responses that can lead to an inaccurate estimation of PK parameters. Immunodeficient (SCID) transgenic hFcRn and C57BL/6 mice were used to characterize the PK of three antibodies that were previously shown to be immunogenic in mice and cynomolgus monkeys. Four mouse strains, Tg32 hFcRn SCID, Tg32 hFcRn, SCID and C57BL/6, were administered adalimumab (Humira®), mAbX and mAbX-YTE at 1 mg/kg, and in SCID strains there was no incidence of immunogenicity. In non-SCID strains, drug-clearing ADAs appeared after 4-7 days, which affected the ability to accurately calculate PK parameters. Single species allometric scaling of PK data for Humira® in SCID and hFcRn SCID mice resulted in improved human PK predictions compared to C57BL/6 mice. Thus, the SCID mouse model was demonstrated to be a useful tool for assessing the preclinical PK of immunogenic therapeutics.
Collapse
Affiliation(s)
- Maria Myzithras
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| | - Tammy Bigwarfe
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| | - Hua Li
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| | - Erica Waltz
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| | - Jennifer Ahlberg
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| | - Craig Giragossian
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| | - Simon Roberts
- a Immune Modulation and Biotherapeutics Discovery, Research, Boehringer Ingelheim , Ridgefield , CT , USA
| |
Collapse
|