1
|
Pezeshkpoor B, Sereda N, Becker-Gotot J, Berkemeier AC, Matuschek I, Müller J, Ramaraje Urs SU, Singh S, Klein C, Marquardt N, Oldenburg J. Comprehensive evaluation of anti-emicizumab antibodies in acquired hemophilia A: a detailed case study and methodological evaluation. J Thromb Haemost 2024:S1538-7836(24)00581-6. [PMID: 39401737 DOI: 10.1016/j.jtha.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/19/2024] [Accepted: 10/03/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Acquired hemophilia A (AHA) is a rare and severe bleeding disorder characterized by autoantibodies inhibiting coagulation factor (F)VIII. Current treatment of AHA involves bypassing agents or FVIII replacement therapy, yet their efficacy is limited in cases of high inhibitor titers. Emicizumab, a humanized bispecific monoclonal antibody, has shown promising hemostatic effectiveness in persons with congenital hemophilia A (HA) and AHA, but a minority of patients developed anti-drug antibodies (ADAs), compromising its efficacy. OBJECTIVES This study aims to characterize the development and impact of anti-emicizumab antibodies in a patient with acquired hemophilia A (AHA) who experienced reduced efficacy of emicizumab treatment. METHODS We present a comprehensive characterization of anti-emicizumab antibodies in a patient with AHA experiencing diminished emicizumab efficacy from week 10 of treatment. We developed a method for analysis of anti-emicizumab antibodies, distinguishing immunoglobulin (Ig) subclasses and IgG subtype profiling. ADAs' neutralizing activity was evaluated using a modified clotting assay. RESULTS The Luminex-based assay demonstrated the presence of anti-emicizumab antibodies, confirmed through competitive analysis. We detected anti-emicizumab IgG antibodies in the patient's plasma between week 16 and 29. IgG1 and IgG2 subclasses were identified. Longitudinal analysis showed IgG isotype antibodies against both emicizumab and FVIII. Anti-emicizumab antibodies exhibited noninhibitory activity, confirmed by a modified Bethesda assay. Moreover, no accelerated clearance of emicizumab was observed in plasma samples from the patient. CONCLUSION This study presents the first documented case of anti-emicizumab antibodies in AHA. Our study provides insights into ADA development against emicizumab, emphasizing the need for monitoring tools. The developed method enables comprehensive evaluation of ADAs, aiding in personalized treatment strategies. These findings contribute to understanding emicizumab's immunogenicity profile in AHA, facilitating its optimized clinical use.
Collapse
Affiliation(s)
- Behnaz Pezeshkpoor
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany.
| | - Nadja Sereda
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Janine Becker-Gotot
- Institute of Molecular Medicine and Experimental Immunology (IMMEI), University of Bonn, Bonn, Germany
| | - Ann-Cristin Berkemeier
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Isabell Matuschek
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Samhitha Urs Ramaraje Urs
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Sneha Singh
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany
| | - Claudia Klein
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany; Center for Rare Diseases Bonn (Zentrum für seltene Erkrankungen der Universität Bonn), University Clinic Bonn, Bonn, Germany
| | - Natascha Marquardt
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany; Center for Rare Diseases Bonn (Zentrum für seltene Erkrankungen der Universität Bonn), University Clinic Bonn, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Medical Faculty, University of Bonn, Germany; Center for Rare Diseases Bonn (Zentrum für seltene Erkrankungen der Universität Bonn), University Clinic Bonn, Bonn, Germany.
| |
Collapse
|
2
|
Aoki K, Higashi K, Oda S, Manabe A, Maeda K, Morise J, Oka S, Inuki S, Ohno H, Oishi S, Nonaka M. Engineering a Low-Immunogenic Mirror-Image VHH against Vascular Endothelial Growth Factor. ACS Chem Biol 2024; 19:1194-1205. [PMID: 38695546 DOI: 10.1021/acschembio.4c00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Immunogenicity is a major caveat of protein therapeutics. In particular, the long-term administration of protein therapeutic agents leads to the generation of antidrug antibodies (ADAs), which reduce drug efficacy while eliciting adverse events. One promising solution to this issue is the use of mirror-image proteins consisting of d-amino acids, which are resistant to proteolytic degradation in immune cells. We have recently reported the chemical synthesis of the enantiomeric form of the variable domain of the antibody heavy chain (d-VHH). However, identifying mirror-image antibodies capable of binding to natural ligands remains challenging. In this study, we developed a novel screening platform to identify a d-VHH specific for vascular endothelial growth factor A (VEGF-A). We performed mirror-image screening of two newly constructed synthetic VHH libraries displayed on T7 phage and identified VHH sequences that effectively bound to the mirror-image VEGF-A target (d-VEGF-A). We subsequently synthesized a d-VHH candidate that preferentially bound the native VEGF-A (l-VEGF-A) with submicromolar affinity. Furthermore, immunization studies in mice demonstrated that this d-VHH elicited no ADAs, unlike its corresponding l-VHH. Our findings highlight the utility of this novel d-VHH screening platform in the development of protein therapeutics exhibiting both reduced immunogenicity and improved efficacy.
Collapse
Affiliation(s)
- Keisuke Aoki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| | - Katsuaki Higashi
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Sakiho Oda
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Asako Manabe
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kayuu Maeda
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Jyoji Morise
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shogo Oka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Medicinal Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan
| | - Motohiro Nonaka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
3
|
Spreafico A, Couselo EM, Irmisch A, Bessa J, Au-Yeung G, Bechter O, Svane IM, Sanmamed MF, Gambardella V, McKean M, Callahan M, Dummer R, Klein C, Umaña P, Justies N, Heil F, Fahrni L, Opolka-Hoffmann E, Waldhauer I, Bleul C, Staack RF, Karanikas V, Fowler S. Phase 1, first-in-human study of TYRP1-TCB (RO7293583), a novel TYRP1-targeting CD3 T-cell engager, in metastatic melanoma: active drug monitoring to assess the impact of immune response on drug exposure. Front Oncol 2024; 14:1346502. [PMID: 38577337 PMCID: PMC10991832 DOI: 10.3389/fonc.2024.1346502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Although checkpoint inhibitors (CPIs) have improved outcomes for patients with metastatic melanoma, those progressing on CPIs have limited therapeutic options. To address this unmet need and overcome CPI resistance mechanisms, novel immunotherapies, such as T-cell engaging agents, are being developed. The use of these agents has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs), which is challenging to predict preclinically and can lead to neutralization of the drug and loss of efficacy. Methods TYRP1-TCB (RO7293583; RG6232) is a T-cell engaging bispecific (TCB) antibody that targets tyrosinase-related protein 1 (TYRP1), which is expressed in many melanomas, thereby directing T cells to kill TYRP1-expressing tumor cells. Preclinical studies show TYRP1-TCB to have potent anti-tumor activity. This first-in-human (FIH) phase 1 dose-escalation study characterized the safety, tolerability, maximum tolerated dose/optimal biological dose, and pharmacokinetics (PK) of TYRP1-TCB in patients with metastatic melanoma (NCT04551352). Results Twenty participants with cutaneous, uveal, or mucosal TYRP1-positive melanoma received TYRP1-TCB in escalating doses (0.045 to 0.4 mg). All participants experienced ≥1 treatment-related adverse event (TRAE); two participants experienced grade 3 TRAEs. The most common toxicities were grade 1-2 cytokine release syndrome (CRS) and rash. Fractionated dosing mitigated CRS and was associated with lower levels of interleukin-6 and tumor necrosis factor-alpha. Measurement of active drug (dual TYPR1- and CD3-binding) PK rapidly identified loss of active drug exposure in all participants treated with 0.4 mg in a flat dosing schedule for ≥3 cycles. Loss of exposure was associated with development of ADAs towards both the TYRP1 and CD3 domains. A total drug PK assay, measuring free and ADA-bound forms, demonstrated that TYRP1-TCB-ADA immune complexes were present in participant samples, but showed no drug activity in vitro. Discussion This study provides important insights into how the use of active drug PK assays, coupled with mechanistic follow-up, can inform and enable ongoing benefit/risk assessment for individuals participating in FIH dose-escalation trials. Translational studies that lead to a better understanding of the underlying biology of cognate T- and B-cell interactions, ultimately resulting in ADA development to novel biotherapeutics, are needed.
Collapse
Affiliation(s)
- Anna Spreafico
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Eva Muñoz Couselo
- Department of Medical Oncology, Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Anja Irmisch
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Juliana Bessa
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - George Au-Yeung
- Department of Medical Oncology, Peter MacCallum Cancer Center and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Oliver Bechter
- Department of General Medical Oncology, Universitair Ziekenhuis (UZ), Leuven, Leuven, Belgium
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy and Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Miguel F. Sanmamed
- Department of Medical Oncology, Clínica Universidad de Navarra and Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Valentina Gambardella
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Meredith McKean
- Sarah Cannon Research Institute at Tennessee Oncology, Nashville, TN, United States
| | - Margaret Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Pablo Umaña
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Nicole Justies
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Florian Heil
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Linda Fahrni
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Eugenia Opolka-Hoffmann
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Inja Waldhauer
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Conrad Bleul
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Roland F. Staack
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Vaios Karanikas
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Stephen Fowler
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
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
|
Yu X, Li T, Shen Z, Jing H, Xie X, Zhou X, Shen Y, Yang Y. The establishment of B cell-deficient Igh-J KO mouse model by gene editing and efficacy evaluation. Int Immunopharmacol 2023; 116:109704. [PMID: 36689847 DOI: 10.1016/j.intimp.2023.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Over the last few years, immunotherapy has made significant progress in treating various cancers with therapeutic antibodies. However, therapeutic antibodies have been validated for inducing an unintended immune response in human and animal models, which leads to the emergence of anti-drug antibodies (ADAs) and affects their effectiveness and safety. In preclinical research, ADAs production by B cells may accelerate antibody metabolism and result in missing potential candidate molecules. Thus, it is urgent to develop preclinical models that remove only B cells without affecting the function of T and NK cells. Rearrangement of immunoglobulin heavy chain J gene fragment (Igh-J) is the first link in B cell development, and immunotherapies are currently leaning toward combination treatments with PD-1/PD-L1 antibodies, here we created humanized PD-1, PD-L1 and Igh-J knockout (hPD-1/hPD-L1, Igh-J KO) mice and validated by using the reported high immunogenicity drug M7824 (a protein designed to simultaneously block PD-L1 and TGF-β pathways, poorly anti-tumor efficacy in immunocompetent mice). Phenotypic analysis revealed that human PD-1 and PD-L1 were detectable in hPD-1/hPD-L1, Igh-J KO mice, but not mouse IgM and IgD. Igh-J KO depleted B cells while increased the percentage of other immune cell types. Meanwhile, the humanization of PD-1/PD-L1 and Igh-J KO had neither effect on the overall development, differentiation, or distribution of T cell subtypes, nor on the activation of NK and T cells, indicating that mice can be used for T and NK-related immunotherapies. Furthermore, M7824 treatment of these B cell-deficient mice inhibited tumor growth significantly, with higher M7824 analog concentrations and lower ADA-positive rates. These findings demonstrate that Igh-J KO mice are an effective and stable preclinical model for testing drugs based on T and NK cells with high immunogenicity in vivo.
Collapse
Affiliation(s)
- Xingyan Yu
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Tao Li
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Zhiyuan Shen
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Hongyan Jing
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Xiulong Xie
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Xiaofei Zhou
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Yuelei Shen
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Yi Yang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China.
| |
Collapse
|
6
|
Pre-Clinical In-Vitro Studies on Parameters Governing Immune Complex Formation. Pharmaceutics 2022; 14:pharmaceutics14061254. [PMID: 35745826 PMCID: PMC9227392 DOI: 10.3390/pharmaceutics14061254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
The success of biotherapeutics is often challenged by the undesirable events of immunogenicity in patients, characterized by the formation of anti-drug antibodies (ADA). Under specific conditions, the ADAs recognizing the biotherapeutic can trigger the formation of immune complexes (ICs), followed by cascades of subsequent effects on various cell types. Hereby, the connection between the characteristics of ICs and their downstream impact is still not well understood. Factors governing the formation of ICs and the characteristics of these IC species were assessed systematically in vitro. Classic analytical methodologies such as SEC-MALS and SV-AUC, and the state-of-the-art technology mass photometry were applied for the characterization. The study demonstrates a clear interplay between (1) the absolute concentration of the involved components, (2) their molar ratios, (3) structural features of the biologic, (4) and of its endogenous target. This surrogate study design and the associated analytical tool-box is readily applicable to most biotherapeutics and provides valuable insights into mechanisms of IC formation prior to FIH studies. The applicability is versatile—from the detection of candidates with immunogenicity risks during developability assessment to evaluation of the impact of degraded or post-translationally modified biotherapeutics on the formation of ICs.
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: 17] [Impact Index Per Article: 5.7] [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
|
Egli J, Heiler S, Weber F, Steiner G, Schwandt T, Bray-French K, Klein C, Fenn S, Lotz GP, Opolka-Hoffmann E, Kraft TE, Petersen L, Moser R, DeGeer J, Siegel M, Finke D, Bessa J, Iglesias A. Enhanced immunogenic potential of cancer immunotherapy antibodies in human IgG1 transgenic mice. MAbs 2022; 14:2143009. [PMID: 36394299 PMCID: PMC9673943 DOI: 10.1080/19420862.2022.2143009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
ABBREVIATIONS ADA Anti-Drug Antibodies; BCR B Cell Receptor; BId Idiotype-specific B Cell; BiTE Bispecific T cell Engager; BMC Bone Marrow Chimeric Mice; BSA Bovine Serum Albumin; CDR Complementary Determining Region; CEA Carcinoembryonic Antigen; CIT Cancer Immunotherapy; CitAbs Cancer Immunotherapy Antibodies; DC Dendritic Cell; ELISA Enzyme-Linked Immunosorbent Assay; FcRn Neonatal Fc Receptor; FcyR Fc gamma Receptor; GM-CSF Granulocyte-Macrophage Colony Stimulating Factor; gMFI Geometric Mean Fluorescence Intensity; H Heavy Chain; IC Immune Complex; Id Idiotype; IgA Immunoglobulin alpha; IgG1 Immunoglobulin gamma 1; IL-2 Interleukin 2; IL-2R Interleukin 2 Receptor; IL2v Interleukin 2 Variant; IVIG1 Intravenous Immunoglobulin 1; KLH Keyhole Limpet Hemocyanin; L Light Chain; MAPPs MHC-associated Peptide Proteomics; MHC Major Histocompatibility Complex; PBMC Peripheral Blood Mononuclear Cells; PBS Phosphate Buffered Saline; SHM Somatic Hypermutation; scFv Single-chain Variable Fragment; TCR T cell Receptor; TFc Fc-specific T cell; TId Id-specific T cell; UV Ultraviolet; V Variable.
Collapse
Affiliation(s)
- Jerome Egli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Stefan Heiler
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Felix Weber
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Guido Steiner
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Timo Schwandt
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Katharine Bray-French
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Roche Glycart AG, Schlieren, Switzerland
| | - Sebastian Fenn
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Gregor P. Lotz
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Eugenia Opolka-Hoffmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Thomas E. Kraft
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Laetitia Petersen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Rebecca Moser
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jonathan DeGeer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Michel Siegel
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Daniela Finke
- Department of Biomedicine and University Children’s Hospital of Basel, University of Basel, Basel, Switzerland
| | - Juliana Bessa
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland,CONTACT Juliana Bessa Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070Basel, Switzerland
| | - Antonio Iglesias
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|