1
|
Sarsarshahi S, Bhattacharya S, Zacharias ZR, Kamel ES, Houtman JCD, Nejadnik R. Highly variable aggregation and glycosylation profiles and their roles in immunogenicity to protein-based therapeutics. J Pharm Sci 2025; 114:103771. [PMID: 40139530 DOI: 10.1016/j.xphs.2025.103771] [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: 02/11/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Production of antibodies against protein-based therapeutics (e.g., monoclonal antibodies (mAbs)) by a recipient's immune system can vary from benign symptoms to chronic neutralization of the compound, and in rare cases, a lethal cytokine storm. One critical factor that can induce or contribute to an anti-drug antibody (ADA) response is believed to be the presence of aggregated proteins in protein-based therapeutics. There is a high level of variability in the aggregation of different proteins, which adds to the complexity in understanding the immune response to these drugs. Furthermore, the level of glycosylation of proteins, which increases drug stability, functionality, and serum half-life, is highly variable and may influence their immunogenicity. Considering the abundance of literature on the effect of aggregation and glycosylation on the immunogenicity of protein-based therapeutics, this review aims to summarize the current knowledge and clarify the immunogenic effects of different protein-based therapeutics such as mAbs. This review focuses on the properties of aggregated proteins and elucidates their relationship with immunogenicity. The contribution of different immune cell subsets and the mechanisms in aggregation-induced immunogenicity are also reviewed. Finally, the potential effects of each glycan, such as sialic acid, mannose, and fucose, on protein-based therapeutics' immunogenicity and stability is discussed.
Collapse
Affiliation(s)
- Sina Sarsarshahi
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Sanghati Bhattacharya
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Zeb R Zacharias
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States
| | - Eman S Kamel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Jon C D Houtman
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Reza Nejadnik
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
2
|
Zhang X, Zhuo J, Wang D, Zhu X. Supramolecular Polymers for Drug Delivery. Chemistry 2025; 31:e202404617. [PMID: 39961052 DOI: 10.1002/chem.202404617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Indexed: 03/21/2025]
Abstract
Supramolecular polymers are constructed through highly reversible and directionally specific non-covalent interactions between monomer units. This unique feature enables supramolecular polymers to undergo controlled structural reconfiguration and functional transformation in response to external stimuli, imparting them with high environmental responsiveness and self-healing properties. In particular, supramolecular polymers exhibit several specific advantages compared to conventional polymers, such as inherent degradability, the ease of preparation and the incorporation of functional units, and smart responsiveness to various biological stimuli. These characters make supramolecular polymers promising candidates for intelligent drug delivery systems in complex biological environments. In this review, we comprehensively summarize the latest developments and representative achievements of supramolecular polymers in drug delivery fields, focusing primarily on the design and synthesis, the properties and functionalities, and the practical applications of supramolecular polymers in small molecule drug delivery, gene therapy, and protein delivery. Finally, we highlight future research directions, focusing on multifunctionality, adaptability, and personalized therapy. We focus on recent studies that address key challenges in the field, providing rational polymer design, important properties, functionality, and understanding delivery strategies. These developments are expected to advance supramolecular polymers as new platforms of intelligent drug delivery systems, offering innovative solutions for the treatment of complex diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiaxin Zhuo
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Dali Wang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
3
|
Sharma K, da Silva BC, Hanauer SB. The role of immunogenicity in optimizing biological therapies for inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2025:1-16. [PMID: 39964309 DOI: 10.1080/17474124.2025.2468302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Immunogenicity of biologic agents for inflammatory bowel disease (IBD) is a critical issue, especially for tumor necrosis factor (TNF) inhibitors, where anti-drug antibodies (ADAs) significantly impact drug clearance, efficacy, and safety. Studies have demonstrated that non-TNF biologics tend to have lower susceptibility to immunogenicity, potentially offering advantages, especially in long-term management. Understanding these differences is important for optimizing IBD treatment outcomes. AREAS COVERED This review examines immunogenicity associated with different classes and individual biologic agents used in IBD; including TNF inhibitors and biologics targeting integrins and interleukins. We discuss key factors influencing ADAs formation, including drug structure, route of administration, and patient-specific factors. The literature reviewed includes recent clinical studies and long-term trials focusing on strategies to reduce immunogenicity such as therapeutic drug monitoring (TDM) and advanced combination. EXPERT OPINION While newer biologics demonstrate lower immunogenicity compared to anti-TNF agents, challenges remain in management to overcome existing ADAs responses while advances in genetic profiling, point-of-care TDM, and combination therapies offer promising pathways to reduce immunogenicity and enhance treatment durability. Continued research and innovation in biologic delivery methods, such as oral and subcutaneous formulations, will be critical in the next decade to further mitigate immunogenic risks and improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Stephen B Hanauer
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
4
|
Garg S, Singla P, Kaur S, Canfarotta F, Velliou E, Dawson JA, Kapur N, Warren NJ, Amarnath S, Peeters M. Future Perspectives on the Automation and Biocompatibility of Molecularly Imprinted Polymers for Healthcare Applications. Macromolecules 2025; 58:1157-1168. [PMID: 39958488 PMCID: PMC11823616 DOI: 10.1021/acs.macromol.4c01621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 02/18/2025]
Abstract
Molecular recognition is of crucial importance in several healthcare applications, such as sensing, drug delivery, and therapeutics. Molecularly imprinted polymers (MIPs) present an interesting alternative to biological receptors (e.g., antibodies, enzymes) for this purpose since synthetic receptors overcome the limited robustness, flexibility, high-cost, and potential for inhibition that comes with natural recognition elements. However, off the shelf MIP products remain limited, which is likely due to the lack of a scalable production approach that can manufacture these materials in high yields and narrow and defined size distributions to have full control over their properties. In this Perspective, we will confer how breakthroughs in the automation of MIP design, manufacturing, and evaluation of performance will accelerate the (commercial) implementation of MIPs in healthcare technology. In addition, we will discuss how prediction of the in vivo behavior of MIPs with animal-free technologies (e.g., 3D tissue models) will be critical to assess their clinical potential.
Collapse
Affiliation(s)
- Saweta Garg
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Pankaj Singla
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
| | - Sarbjeet Kaur
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Francesco Canfarotta
- MIP
Discovery, Colworth Park, Sharnbrook, MK44 1LQ, Bedfordshire, United Kingdom
| | - Eirini Velliou
- University
College London, Centre for 3D Models
of Health and Disease, Charles Bell House, London, W1W 7TY, United Kingdom
| | - James A. Dawson
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Nikil Kapur
- University
of Leeds, School of Mechanical Engineering, Woodhouse Lane, Leeds, LS2 9JT, United Kingdom
| | - Nicholas J. Warren
- School of
Chemical, Materials and Biological Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, S1 3JD, United Kingdom
| | - Shoba Amarnath
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Marloes Peeters
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
| |
Collapse
|
5
|
Hale G. Living in LALA land? Forty years of attenuating Fc effector functions. Immunol Rev 2024; 328:422-437. [PMID: 39158044 PMCID: PMC11659930 DOI: 10.1111/imr.13379] [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: 08/20/2024]
Abstract
The Fc region of antibodies is vital for most of their physiological functions, many of which are engaged through binding to a range of Fc receptors. However, these same interactions are not always helpful or wanted when therapeutic antibodies are directed against self-antigens, and can sometimes cause catastrophic adverse reactions. Over the past 40 years, there have been intensive efforts to "silence" unwanted binding to Fc-gamma receptors, resulting in at least 45 different variants which have entered clinical trials. One of the best known is "LALA" (L234A/L235A). However, neither this, nor most of the other variants in clinical use are completely silenced, and in addition, the biophysical properties of many of them are compromised. I review the development of different variants to see what we can learn from their biological properties and use in the clinic. With the rise of powerful new uses of antibody therapy such as bispecific T-cell engagers, antibody-drug conjugates, and checkpoint inhibitors, it is increasingly important to optimize the Fc region as well as the antibody binding site in order to achieve the best combination of safety and efficacy.
Collapse
|
6
|
Malinczak CA, Burns Naas LA, Clark A, Conze D, DiNovi M, Kaminski N, Kruger C, Lönnerdal B, Lukacs NW, Merker R, Peterson R. Workshop report: A study roadmap to evaluate the safety of recombinant human lactoferrin expressed in Komagataella phaffii intended as an ingredient in conventional foods - Recommendations of a scientific expert panel. Food Chem Toxicol 2024; 190:114817. [PMID: 38880466 DOI: 10.1016/j.fct.2024.114817] [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: 04/09/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
Human milk lactoferrin (hmLF) is a glycoprotein with well-known effects on immune function. Helaina Inc. has used a glycoengineered yeast, Komatagaella phaffii, to produce recombinant human lactoferrin (Helaina rhLF, Effera™) that is structurally similar to hmLF with intended uses as a food ingredient. However, earlier FDA reviews of rhLF were withdrawn due to insufficient safety data and unanswered safety questions the experts and FDA raised about the immunogenicity/immunotoxicity risks of orally ingested rhLF. Helaina organized a panel of leading scientists to build and vet a safety study roadmap containing the studies and safety endpoints needed to address these questions. Panelists participated in a one-day virtual workshop in June 2023 and ensuing discussions through July 2023. Relevant workshop topics included physicochemical properties of LF, regulatory history of bovine LF and rhLF as food ingredients in the FDA's generally recognized as safe (GRAS) program, and synopses of publicly available studies on the immunogenicity/alloimmunization, immunotoxicology, iron homeostasis, and absorption, distribution, metabolism, and excretion of rhLF. Panelists concluded that the safety study roadmap addresses the unanswered safety questions and the intended safe use of rhLF as a food ingredient for adults and agreed on broad applications of the roadmap to assess the safety and support GRAS of other recombinant milk proteins with immunomodulatory functions.
Collapse
Affiliation(s)
| | | | | | | | | | - Norbert Kaminski
- Department of Pharmacology & Toxicology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
| | | | - Bo Lönnerdal
- Department of Internal Medicine, University of California, Davis, CA, USA.
| | - Nicholas W Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA.
| | | | | |
Collapse
|
7
|
Agrohia DK, Goswami R, Jantarat T, Çiçek YA, Thongsukh K, Jeon T, Bell JM, Rotello VM, Vachet RW. Suborgan Level Quantitation of Proteins in Tissues Delivered by Polymeric Nanocarriers. ACS NANO 2024; 18:16808-16818. [PMID: 38870478 PMCID: PMC11497159 DOI: 10.1021/acsnano.4c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Amidst the rapid growth of protein therapeutics as a drug class, there is an increased focus on designing systems to effectively deliver proteins to target organs. Quantitative monitoring of protein distributions in tissues is essential for optimal development of delivery systems; however, existing strategies can have limited accuracy, making it difficult to assess suborgan dosing. Here, we describe a quantitative imaging approach that utilizes metal-coded mass tags and laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to quantify the suborgan distributions of proteins in tissues that have been delivered by polymeric nanocarriers. Using this approach, we measure nanomole per gram levels of proteins as delivered by guanidinium-functionalized poly(oxanorborneneimide) (PONI) polymers to various tissues, including the alveolar region of the lung. Due to the multiplexing capability of the LA-ICP-MS imaging, we are also able to simultaneously quantify protein and polymer distributions, obtaining valuable information about the relative excretion pathways of the protein cargo and carrier. This imaging approach will facilitate quantitative correlations between nanocarrier properties and protein cargo biodistributions.
Collapse
Affiliation(s)
- Dheeraj K. Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Teerapong Jantarat
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Yağız Anil Çiçek
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Korndanai Thongsukh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jonathan M. Bell
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
8
|
Liu F, Hutchinson R. Visible particles in parenteral drug products: A review of current safety assessment practice. Curr Res Toxicol 2024; 7:100175. [PMID: 38975062 PMCID: PMC11223083 DOI: 10.1016/j.crtox.2024.100175] [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: 03/29/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Parenteral drug products (PDPs) are administered extensively to treat various diseases. Product quality plays a critical role in ensuring patient safety and product efficacy. One important quality challenge is the contamination of particles in PDPs. Particle presence in PDPs represents potential safety risk to patients. Differential guidance and practice have been in place for visible (VPs) and subvisible particles (SVPs) in PDPs. For SVPs, the amount limits have been harmonized in multiple Pharmacopeias. The pharmaceutical industry follows the guided limits for regulatory and quality compliance. However, for VPs, no such acceptable limit has been set. This results in not only quality but also safety challenges for manufacturers and drug developers in managing and evaluating VPs. It is important to understand the potential safety risk of VPs so these can be weighed against the benefit of the PDPs. To evaluate their potential risk(s), it is necessary to understand their nature, origin, frequency of their occurrence, safety risk, the risk mitigation measures, and the method to evaluate their safety. The current paper reviews the critical literature on these aspects and provides insight into considerations when performing safety assessment and managing the risk(s) for VPs in PDPs.
Collapse
Affiliation(s)
- Frank Liu
- Safe Product Services LLC, Pittsfield, MA, USA
| | | |
Collapse
|
9
|
Abu Serhan H, Taha MJJ, Abuawwad MT, Abdelaal A, Irshaidat S, Abu Serhan L, Abu Salim QF, Awamleh N, Abdelazeem B, Elnahry AG. Safety and Efficacy of Brolucizumab in the Treatment of Diabetic Macular Edema and Diabetic Retinopathy: A Systematic Review and Meta-Analysis. Semin Ophthalmol 2024; 39:251-260. [PMID: 37849309 DOI: 10.1080/08820538.2023.2271095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023]
Abstract
PURPOSE To investigate the efficacy and safety of brolucizumab in diabetic macular edema (DME) and diabetic retinopathy (DR). METHODS In this systematic review and meta-analysis, an electronic search was done to acquire all articles describing brolucizumab use in patients with DME and DR. The review was prospectively registered on PROSPERO (CRD42022382625). Collected articles were filtered through two stages by independent reviewers. Data were extracted from the included articles and then analyzed accordingly. RESULTS Brolucizumab induced significant improvement in best-corrected visual acuity and was either better or non-inferior to other types of anti-VEGF (MD -0.64 mu, 95% CI [-1.15, -0.13], P = .01); the same observation was noted with regards to central subfield macular thickness (CSMT) (MD -138.6 mu, 95% CI [-151.9, -125.3], P = .00001). Brolucizumab was reported to be relatively safe for use in diabetic patients, with few adverse events observed, with a higher frequency of adverse events in relation to the 3 mg dose compared to the 6 mg dose. CONCLUSION Brolucizumab is a new drug that has potential advantages in efficacy over other anti-VEGF agents in the treatment of DME and DR. It showed significant improvement in BCVA and CSMT with the possibility of a lower dosing schedule compared to other agents. Although observed in low frequency, sight-threatening adverse effects appear to occur more frequently compared to other anti-VEGF agents. The main observed adverse event was retinal vasculitis which was seen more commonly with the 3 mg dose versus the 6 mg dose.
Collapse
Affiliation(s)
- Hashem Abu Serhan
- Department of Ophthalmology, Hamad Medical Corporations, Doha, Qatar
| | | | | | - Abdelaziz Abdelaal
- Harvard Medical School, Postgraduate Medical Education, Boston, USA
- Doheny Eye Institute, University of California, Los Angeles, USA
| | - Sara Irshaidat
- Department of Pediatrics, King Hussein Cancer Centre, Amman, Jordan
| | | | | | - Nour Awamleh
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Basel Abdelazeem
- Department of Internal Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Ayman G Elnahry
- Department of Ophthalmology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| |
Collapse
|
10
|
Harris CT, Cohen S. Reducing Immunogenicity by Design: Approaches to Minimize Immunogenicity of Monoclonal Antibodies. BioDrugs 2024; 38:205-226. [PMID: 38261155 PMCID: PMC10912315 DOI: 10.1007/s40259-023-00641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/24/2024]
Abstract
Monoclonal antibodies (mAbs) have transformed therapeutic strategies for various diseases. Their high specificity to target antigens makes them ideal therapeutic agents for certain diseases. However, a challenge to their application in clinical practice is their potential risk to induce unwanted immune response, termed immunogenicity. This challenge drives the continued efforts to deimmunize these protein therapeutics while maintaining their pharmacokinetic properties and therapeutic efficacy. Because mAbs hold a central position in therapeutic strategies against an array of diseases, the importance of conducting comprehensive immunogenicity risk assessment during the drug development process cannot be overstated. Such assessment necessitates the employment of in silico, in vitro, and in vivo strategies to evaluate the immunogenicity risk of mAbs. Understanding the intricacies of the mechanisms that drive mAb immunogenicity is crucial to improving their therapeutic efficacy and safety and developing the most effective strategies to determine and mitigate their immunogenic risk. This review highlights recent advances in immunogenicity prediction strategies, with a focus on protein engineering strategies used throughout development to reduce immunogenicity.
Collapse
Affiliation(s)
- Chantal T Harris
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA
| | - Sivan Cohen
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA.
| |
Collapse
|
11
|
Di Ianni A, Fraone T, Balestra P, Cowan K, Riccardi Sirtori F, Barbero L. Assessing MAPPs assay as a tool to predict the immunogenicity potential of protein therapeutics. Life Sci Alliance 2024; 7:e202302095. [PMID: 37833075 PMCID: PMC10576005 DOI: 10.26508/lsa.202302095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
MHC-II-associated peptide proteomics (MAPPs) is a mass spectrometry-based (MS) method to identify naturally presented MHC-II-associated peptides that could elicit CD4+T cell activation. MAPPs assay is considered one of the assays that better characterize the safety of biotherapeutics by driving the selection of the best candidates concerning their immunogenicity risk. However, there is little knowledge about the impact of bead material on the recovery of MHC-II MS-eluted ligands in MAPPs assays. Here, we firstly describe a robust MAPPs protocol by implementing streptavidin magnetic beads for the isolation of these peptides instead of commonly used NHS-activated beads. Moreover, we assessed the impact of the cell medium used for cell cultures on the morphology and recovery of the in vitro-generated APCs, and its potential implications in the amount of MHC-II isolated peptides. We also described an example of a MAPPs assay application to investigate drug-induced immunogenicity of two bispecific antibodies and compared them with monospecific trastuzumab IgG1 control. This work highlighted the importance of MAPPs in the preclinical in vitro strategy to mitigate the immunogenicity risk of biotherapeutics.
Collapse
Affiliation(s)
- Andrea Di Ianni
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., An Affiliate of Merck KGaA, Darmstadt, Germany
| | - Tiziana Fraone
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., An Affiliate of Merck KGaA, Darmstadt, Germany
| | - Piercesare Balestra
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., An Affiliate of Merck KGaA, Darmstadt, Germany
| | - Kyra Cowan
- New Biological Entities, Drug Metabolism and Pharmacokinetics (NBE-DMPK), Research and Development, Merck KGaA, Darmstadt, Germany
| | - Federico Riccardi Sirtori
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., An Affiliate of Merck KGaA, Darmstadt, Germany
| | - Luca Barbero
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., An Affiliate of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
12
|
Vuong CN, Reynolds KM, Rivera GS, Zeng B, Karimpourkalou Z, Norng M, Zhang Y, Chowdhury R, Pedersen D, Pantoja M, Collarini E, Garimalla S, Izquierdo S, Vajda EG, Antonio B, Srivastava DB, van de Lavoir MC, Abdiche Y, Harriman W, Leighton PA. Heavy chain-only antibodies with a stabilized human VH in transgenic chickens for therapeutic antibody discovery. MAbs 2024; 16:2435476. [PMID: 39607037 PMCID: PMC11610561 DOI: 10.1080/19420862.2024.2435476] [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: 09/21/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
Heavy chain-only antibodies have found many applications where conventional heavy-light heterodimeric antibodies are not favorable. Heavy chain-only antibodies with their single antigen-binding domain offer the advantage of a smaller size and higher stability relative to conventional antibodies, and thus, the potential for novel targeting modalities. Domain antibodies have commonly been sourced from camelids with ex-vivo humanization or transgenic rodents expressing heavy chains without light chains, but these host species are all mammalian, limiting their capacity to elicit robust immune responses to conserved mammalian targets. We have developed transgenic chickens expressing heavy chain-only antibodies with a human variable region to combine the superior target recognition advantages of a divergent, non-mammalian host with the ability to discover single-domain binders. These birds produce robust immune responses, consisting of antigen-specific antibodies targeting diverse epitopes with a range of affinities. Biophysical attributes are favorable, with good developability profiles and low predicted immunogenicity.
Collapse
|
13
|
Lee MV, Saad OM, Wong S, LaMar J, Kamen L, Ordonia B, Melendez R, Hassanzadeh A, Chung S, Kaur S. Development of a semi-automated MHC-associated peptide proteomics (MAPPs) method using streptavidin bead-based immunoaffinity capture and nano LC-MS/MS to support immunogenicity risk assessment in drug development. Front Immunol 2023; 14:1295285. [PMID: 38022649 PMCID: PMC10667718 DOI: 10.3389/fimmu.2023.1295285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Major histocompatibility complex (MHC)-Associated Peptide Proteomics (MAPPs) is an ex vivo method used to assess the immunogenicity risk of biotherapeutics. MAPPs can identify potential T-cell epitopes within the biotherapeutic molecule. Using adalimumab treated human monocyte derived dendritic cells (DCs) and a pan anti-HLA-DR antibody (Ab), we systematically automated and optimized biotin/streptavidin (SA)-capture antibody coupling, lysate incubation with capture antibody, as well as the washing and elution steps of a MAPPs method using functionalized magnetic beads and a KingFisher Magnetic Particle processor. Automation of these steps, combined with capturing using biotinylated-Ab/SA magnetic beads rather than covalently bound antibody, improved reproducibility as measured by minimal inter-and intra-day variability, as well as minimal analyst-to-analyst variability. The semi-automated MAPPs workflow improved sensitivity, allowing for a lower number of cells per analysis. The method was assessed using five different biotherapeutics with varying immunogenicity rates ranging from 0.1 to 48% ADA incidence in the clinic. Biotherapeutics with ≥10%immunogenicity incidence consistently presented more peptides (1.8-28 fold) and clusters (10-21 fold) compared to those with <10% immunogenicity incidence. Our semi-automated MAPPs method provided two main advantages over a manual workflow- the robustness and reproducibility affords confidence in the epitopes identified from as few as 5 to 10 donors and the method workflow can be readily adapted to incorporate different capture Abs in addition to anti-HLA-DR. The incorporation of semi-automated MAPPs with biotinylated-Ab/SA bead-based capture in immunogenicity screening strategies allows the generation of more consistent and reliable data, helping to improve immunogenicity prediction capabilities in drug development. MHC associated peptide proteomics (MAPPs), Immunogenicity risk assessment, in vitro/ex vivo, biotherapeutics, Major Histocompatibility Complex Class II (MHC II), LC-MS, Immunoaffinity Capture, streptavidin magnetic beads.
Collapse
Affiliation(s)
| | - Ola M. Saad
- *Correspondence: M. Violet Lee, ; Ola M. Saad,
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
De Groot AS, Khan S, Mattei AE, Lelias S, Martin WD. Does human homology reduce the potential immunogenicity of non-antibody scaffolds? Front Immunol 2023; 14:1215939. [PMID: 38022550 PMCID: PMC10664710 DOI: 10.3389/fimmu.2023.1215939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Biologics developers are moving beyond antibodies for delivery of a wide range of therapeutic interventions. These non-antibody modalities are often based on 'natural' protein scaffolds that are modified to deliver bioactive sequences. Both human-derived and non-human-sourced scaffold proteins have been developed. New types of "non-antibody" scaffolds are still being discovered, as they offer attractive alternatives to monoclonals due to their smaller size, improved stability, and ease of synthesis. They are believed to have low immunogenic potential. However, while several human-sourced protein scaffolds have not been immunogenic in clinical studies, this may not predict their overall performance in other therapeutic applications. A preliminary evaluation of their potential for immunogenicity is warranted. Immunogenicity risk potential has been clearly linked to the presence of T "helper" epitopes in the sequence of biologic therapeutics. In addition, tolerogenic epitopes are present in some human proteins and may decrease their immunogenic potential. While the detailed sequences of many non-antibody scaffold therapeutic candidates remain unpublished, their backbone sequences are available for review and analysis. We assessed 12 example non-antibody scaffold backbone sequences using our epitope-mapping tools (EpiMatrix) for this perspective. Based on EpiMatrix scoring, their HLA DRB1-restricted T cell epitope content appears to be lower than the average protein, and sequences that may act as tolerogenic epitopes are present in selected human-derived scaffolds. Assessing the potential immunogenicity of scaffold proteins regarding self and non-self T cell epitopes may be of use for drug developers and clinicians, as these exciting new non-antibody molecules begin to emerge from the preclinical pipeline into clinical use.
Collapse
Affiliation(s)
- Anne S. De Groot
- EpiVax, Providence, RI, United States
- University of Georgia, Center for Vaccines and Immunology, Athens, GA, United States
| | | | | | | | | |
Collapse
|
15
|
Jankowski W, Kidchob C, Bunce C, Cloake E, Resende R, Sauna ZE. The MHC Associated Peptide Proteomics assay is a useful tool for the non-clinical assessment of immunogenicity. Front Immunol 2023; 14:1271120. [PMID: 37915568 PMCID: PMC10616951 DOI: 10.3389/fimmu.2023.1271120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/13/2023] [Indexed: 11/03/2023] Open
Abstract
The propensity of therapeutic proteins to elicit an immune response, poses a significant challenge in clinical development and safety of the patients. Assessment of immunogenicity is crucial to predict potential adverse events and design safer biologics. In this study, we employed MHC Associated Peptide Proteomics (MAPPS) to comprehensively evaluate the immunogenic potential of re-engineered variants of immunogenic FVIIa analog (Vatreptacog Alfa). Our finding revealed the correlation between the protein sequence affinity for MHCII and the number of peptides identified in a MAPPS assay and this further correlates with the reduced T-cell responses. Moreover, MAPPS enable the identification of "relevant" T cell epitopes and may contribute to the development of biologics with lower immunogenic potential.
Collapse
Affiliation(s)
- Wojciech Jankowski
- Hemostasis Branch 1, Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Christopher Kidchob
- Hemostasis Branch 1, Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | | | | | | | - Zuben E. Sauna
- Hemostasis Branch 1, Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
16
|
Baghini SS, Razeghian E, Malayer SK, Pecho RDC, Obaid M, Awfi ZS, Zainab HA, Shamsara M. Recent advances in the application of genetic and epigenetic modalities in the improvement of antibody-producing cell lines. Int Immunopharmacol 2023; 123:110724. [PMID: 37582312 DOI: 10.1016/j.intimp.2023.110724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023]
Abstract
There are numerous applications for recombinant antibodies (rAbs) in biological and toxicological research. Monoclonal antibodies are synthesized using genetic engineering and other related processes involved in the generation of rAbs. Because they can identify specific antigenic sites on practically any molecule, including medicines, hormones, microbial antigens, and cell receptors, rAbs are particularly useful in scientific research. The key benefits of rAbs are improved repeatability, control, and consistency, shorter manufacturing times than with hybridoma technology, an easier transition from one format of antibody to another, and an animal-free process. The engineering of the host cell has recently been developed method for enhancing the production efficiency and improving the quality of antibodies from mammalian cell lines. In this light, genetic engineering is mostly utilized to manage cellular chaperones, decrease cell death, increase cell viability, change the microRNAs (miRNAs) pattern in mammalian cells, and glycoengineered cell lines. Here, we shed light on how genetic engineering can be used therapeutically to produce antibodies at higher levels with greater potency and effectiveness.
Collapse
Affiliation(s)
- Sadegh Shojaei Baghini
- Plant Biotechnology Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Setare Kakavand Malayer
- Department of Biology, Faculty of Biological Science, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | | | | | - Zinah Salem Awfi
- Department of Dental Industry Techniques, Al-Noor University College, Nineveh, Iraq.
| | - H A Zainab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | - Mehdi Shamsara
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
17
|
De Groot AS, Roberts BJ, Mattei A, Lelias S, Boyle C, Martin WD. Immunogenicity risk assessment of synthetic peptide drugs and their impurities. Drug Discov Today 2023; 28:103714. [PMID: 37467878 DOI: 10.1016/j.drudis.2023.103714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Peptide drugs play an important part in medicine owing to their many therapeutic applications. Of the 80 peptide drugs approved for use in humans, at least five are now off-patent and are consequently being developed as generic alternatives to the originator products. To accelerate access to generic products, the FDA has proposed new regulatory pathways that do not require direct comparisons of generics to originators in clinical trials. The 'Abbreviated New Drug Application' (ANDA) pathway recommends that sponsors provide information on any new impurities in the generic drug, compared with the originator product, because the impurities can have potential to elicit unwanted immune responses owing to the introduction of T-cell epitopes. This review describes how peptide drug impurities can elicit unexpected immunogenicity and describes a framework for performing immunogenicity risk assessment of all types of bioactive peptide products. Although this report primarily focuses on generic peptides and their impurities, the approach might also be of interest for developers of novel peptide drugs who are preparing their products for an initial regulatory review.
Collapse
Affiliation(s)
- Anne S De Groot
- EpiVax, 188 Valley Street, Suite 424, Providence, RI, USA; University of Georgia, Center for Vaccines and Immunology, Athens, GA USA.
| | | | - Aimee Mattei
- EpiVax, 188 Valley Street, Suite 424, Providence, RI, USA
| | - Sandra Lelias
- EpiVax, 188 Valley Street, Suite 424, Providence, RI, USA
| | | | | |
Collapse
|
18
|
Di Ianni A, Barbero L, Fraone T, Cowan K, Sirtori FR. Preclinical risk assessment strategy to mitigate the T-cell dependent immunogenicity of protein biotherapeutics: State of the art, challenges and future perspectives. J Pharm Biomed Anal 2023; 234:115500. [PMID: 37311374 DOI: 10.1016/j.jpba.2023.115500] [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: 03/27/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023]
Abstract
Protein therapeutics hold a prominent role and have brought significant diversity in efficacious medicinal products. Not just monoclonal antibodies and different antibody formats (pegylated antigen-binding fragments, bispecifics, antibody-drug conjugates, single chain variable fragments, nanobodies, dia-, tria- and tetrabodies), but also purified blood products, growth factors, recombinant cytokines, enzyme replacement factors, fusion proteins are all good instances of therapeutic proteins that have been developed in the past decades and approved for their value in oncology, immune-oncology, and autoimmune diseases discovery programs. Although there was an ingrained belief that fully humanized proteins were expected to have limited immunogenicity, adverse effects associated with immune responses to biological therapies raised some concern in biotech companies. Consequently, drug developers are designing strategies to assess potential immune responses to protein therapeutics during both the preclinical and clinical phases of development. Despite the many factors that can contribute to protein immunogenicity, T cell- (thymus-) dependent (Td) immunogenicity seems to play a crucial role in the development of anti-drug antibodies (ADAs) to biologics. A broad range of methodologies to predict and rationally assess Td immune responses to protein drugs has been developed. This review aims to briefly summarize the preclinical immunogenicity risk assessment strategy to mitigate the risk of potential immunogenic candidates coming towards clinical phases, discussing the advantages and limitations of these technologies, and suggesting a rational approach for assessing and mitigating Td immunogenicity.
Collapse
Affiliation(s)
- Andrea Di Ianni
- University of Turin, Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Via Ribes 1, 10010 Colleretto Giacosa (TO), Italy
| | - Luca Barbero
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Via Ribes 1, 10010 Colleretto Giacosa (TO), Italy
| | - Tiziana Fraone
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Via Ribes 1, 10010 Colleretto Giacosa (TO), Italy
| | - Kyra Cowan
- New Biological Entities, Drug Metabolism and Pharmacokinetics (NBE-DMPK), Research and Development, Merck KGaA, Frankfurterstrasse 250, 64293 Darmstadt, Germany
| | - Federico Riccardi Sirtori
- NBE-DMPK Innovative BioAnalytics, Merck Serono RBM S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Via Ribes 1, 10010 Colleretto Giacosa (TO), Italy.
| |
Collapse
|
19
|
Nguyen NH, Jarvi NL, Balu-Iyer SV. Immunogenicity of Therapeutic Biological Modalities - Lessons from Hemophilia A Therapies. J Pharm Sci 2023; 112:2347-2370. [PMID: 37220828 DOI: 10.1016/j.xphs.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/25/2023]
Abstract
The introduction and development of biologics such as therapeutic proteins, gene-, and cell-based therapy have revolutionized the scope of treatment for many diseases. However, a significant portion of the patients develop unwanted immune reactions against these novel biological modalities, referred to as immunogenicity, and no longer benefit from the treatments. In the current review, using Hemophilia A (HA) therapy as an example, we will discuss the immunogenicity issue of multiple biological modalities. Currently, the number of therapeutic modalities that are approved or recently explored to treat HA, a hereditary bleeding disorder, is increasing rapidly. These include, but are not limited to, recombinant factor VIII proteins, PEGylated FVIII, FVIII Fc fusion protein, bispecific monoclonal antibodies, gene replacement therapy, gene editing therapy, and cell-based therapy. They offer the patients a broader range of more advanced and effective treatment options, yet immunogenicity remains the most critical complication in the management of this disorder. Recent advances in strategies to manage and mitigate immunogenicity will also be reviewed.
Collapse
Affiliation(s)
- Nhan H Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA; Currently at Truvai Biosciences, Buffalo, NY, USA
| | - Nicole L Jarvi
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
20
|
Álamo P, Parladé E, Favaro MTP, Gallardo A, Mendoza R, Ferreira LC, Roher N, Mangues R, Villaverde A, Vázquez E. Probing the Biosafety of Implantable Artificial Secretory Granules for the Sustained Release of Bioactive Proteins. ACS APPLIED MATERIALS & INTERFACES 2023; 15:39167-39175. [PMID: 37614001 PMCID: PMC10450642 DOI: 10.1021/acsami.3c08643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/27/2023] [Indexed: 08/25/2023]
Abstract
Among bio-inspired protein materials, secretory protein microparticles are of clinical interest as self-contained, slow protein delivery platforms that mimic secretory granules of the human endocrine system, in which the protein is both the drug and the scaffold. Upon subcutaneous injection, their progressive disintegration results in the sustained release of the building block polypeptides, which reach the bloodstream for systemic distribution and subsequent biological effects. Such entities are easily fabricated in vitro by Zn-assisted cross-molecular coordination of histidine residues. Using cationic Zn for the assembly of selected pure protein species and in the absence of any heterologous holding material, these granules are expected to be nontoxic and therefore adequate for different clinical uses. However, such presumed biosafety has not been so far confirmed and the potential protein dosage threshold not probed yet. By selecting the receptor binding domain (RBD) from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein as a model protein and using a mouse lab model, we have explored the toxicity of RBD-made secretory granules at increasing doses up to ∼100 mg/kg of animal weight. By monitoring body weight and biochemical blood markers and through the histological scrutiny of main tissues and organs, we have not observed systemic toxicity. Otherwise, the bioavailability of the material was demonstrated by the induction of specific antibody responses. The presented data confirm the intrinsic biosafety of artificial secretory granules made by recombinant proteins and prompt their further clinical development as self-contained and dynamic protein reservoirs.
Collapse
Affiliation(s)
- Patricia Álamo
- Institut
d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Josep
Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
| | - Eloi Parladé
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Marianna T. P. Favaro
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
- Instituto
de Ciências Biomédicas, Universidade
de São Paulo, São
Paulo 05508-000, Brazil
| | - Alberto Gallardo
- Institut
d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Department
of Pathology, Hospital de la Santa Creu
i Sant Pau, 08025 Barcelona, Spain
| | - Rosa Mendoza
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Luís C.
S. Ferreira
- Instituto
de Ciências Biomédicas, Universidade
de São Paulo, São
Paulo 05508-000, Brazil
| | - Nerea Roher
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
- Department
of Cell Biology, Animal Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ramón Mangues
- Institut
d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Josep
Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
| | - Antonio Villaverde
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Esther Vázquez
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN,
ISCIII), Universitat Autònoma de
Barcelona, 08193 Bellaterra, Spain
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament
de Genètica i de Microbiologia, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
21
|
Gokemeijer J, Wen Y, Jawa V, Mitra-Kaushik S, Chung S, Goggins A, Kumar S, Lamberth K, Liao K, Lill J, Phung Q, Walsh R, Roberts BJ, Swanson M, Singh I, Tourdot S, Kroenke MA, Rup B, Goletz TJ, Gupta S, Malherbe L, Pattijn S. Survey Outcome on Immunogenicity Risk Assessment Tools for Biotherapeutics: an Insight into Consensus on Methods, Application, and Utility in Drug Development. AAPS J 2023; 25:55. [PMID: 37266912 DOI: 10.1208/s12248-023-00820-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/12/2023] [Indexed: 06/03/2023] Open
Abstract
A survey conducted by the Therapeutic Product Immunogenicity (TPI) community within the American Association of Pharmaceutical Scientists (AAPS) posed questions to the participants on their immunogenicity risk assessment strategies prior to clinical development. The survey was conducted in 2 phases spanning 5 years, and queried information about in silico algorithms and in vitro assay formats for immunogenicity risk assessments and how the data were used to inform early developability effort in discovery, chemistry, manufacturing and control (CMC), and non-clinical stages of development. The key findings representing the trends from a majority of the participants included the use of high throughput in silico algorithms, human immune cell-based assays, and proteomics based outputs, as well as specialized assays when therapeutic mechanism of action could impact risk assessment. Additional insights into the CMC-related risks could also be gathered with the same tools to inform future process development and de-risk critical quality attributes with uncertain and unknown risks. The use of the outputs beyond supporting early development activities was also noted with participants utilizing the risk assessments to drive their clinical strategy and streamline bioanalysis.
Collapse
Affiliation(s)
- Jochem Gokemeijer
- Bristol Myers Squibb, 100 Binney Street, Cambridge, Massachusetts, 02143, USA.
| | - Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, USA
| | - Vibha Jawa
- Bristol Myers Squibb, Lawrenceville, New Jersey, 08540, USA
| | | | - Shan Chung
- Genentech Inc., South San Francisco, California, 94080, USA
| | - Alan Goggins
- Merck & Co., Inc., South San Francisco, California, 94080, USA
| | - Seema Kumar
- EMD Serono Research & Development Institute, (A Business of Merck KGaA, Darmstadt, Germany), Billerica, Massachusetts, 01826, USA
| | | | - Karen Liao
- Merck & Co., Inc., West Point, Pennsylvania, 19486, USA
| | - Jennie Lill
- Genentech Inc., South San Francisco, California, 94080, USA
| | - Qui Phung
- Genentech Inc., South San Francisco, California, 94080, USA
| | - Robin Walsh
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, USA
| | | | - Michael Swanson
- Janssen R&D LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA
| | | | - Sophie Tourdot
- BioMedicine Design, Pfizer Inc., Andover, Massachusetts, 01810, USA
| | - Mark A Kroenke
- Clinical Immunology, Amgen, Thousand Oaks, California, 91320, USA
| | - Bonita Rup
- Bonnie Rup Consulting, LLC, 42 Commonwealth Ave, Boston, Massachusetts, 02116, USA
| | | | | | - Laurent Malherbe
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, 46285, USA
| | | |
Collapse
|
22
|
Funeh CN, Bridoux J, Ertveldt T, De Groof TWM, Chigoho DM, Asiabi P, Covens P, D'Huyvetter M, Devoogdt N. Optimizing the Safety and Efficacy of Bio-Radiopharmaceuticals for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051378. [PMID: 37242621 DOI: 10.3390/pharmaceutics15051378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The precise delivery of cytotoxic radiation to cancer cells through the combination of a specific targeting vector with a radionuclide for targeted radionuclide therapy (TRT) has proven valuable for cancer care. TRT is increasingly being considered a relevant treatment method in fighting micro-metastases in the case of relapsed and disseminated disease. While antibodies were the first vectors applied in TRT, increasing research data has cited antibody fragments and peptides with superior properties and thus a growing interest in application. As further studies are completed and the need for novel radiopharmaceuticals nurtures, rigorous considerations in the design, laboratory analysis, pre-clinical evaluation, and clinical translation must be considered to ensure improved safety and effectiveness. Here, we assess the status and recent development of biological-based radiopharmaceuticals, with a focus on peptides and antibody fragments. Challenges in radiopharmaceutical design range from target selection, vector design, choice of radionuclides and associated radiochemistry. Dosimetry estimation, and the assessment of mechanisms to increase tumor uptake while reducing off-target exposure are discussed.
Collapse
Affiliation(s)
- Cyprine Neba Funeh
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Jessica Bridoux
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Timo W M De Groof
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Dora Mugoli Chigoho
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Parinaz Asiabi
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Peter Covens
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Matthias D'Huyvetter
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Nick Devoogdt
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| |
Collapse
|
23
|
Wang C, Xiao C, Chen Y, Li Y, Zhang Q, Shan W, Li Y, Bi S, Wang Y, Wang X, Ren L. Sequential administration of virus-like particle-based nanomedicine to elicit enhanced tumor chemotherapy. J Mater Chem B 2023; 11:2674-2683. [PMID: 36857702 DOI: 10.1039/d2tb02163c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Protein cages have played a long-standing role in biomedicine applications, especially in tumor chemotherapy. Among protein cages, virus like particles (VLPs) have received attention for their potential applications in vaccine development and targeted drug delivery. However, most of the existing protein-based platform technologies are plagued with immunological problems that may limit their systemic delivery efficiency as drug carriers. Here, we show that using immune-orthogonal protein cages sequentially and modifying the dominant loop epitope can circumvent adaptive immune responses and enable effective drug delivery using repeated dosing. We genetically modified three different hepadnavirus core protein derived VLPs as delivery vectors for doxorubicin (DOX). These engineered VLPs have similar assembly characteristics, particle sizes, and immunological properties. Our results indicated that there was negligible antibody cross-reactivity in either direction between these three RGD-VLPs in mice that were previously immunized against HBc VLPs. Moreover, the sequential administration of multiple RGD-VLP-based nanomedicine (DOX@RGD-VLPs) could effectively reduce immune clearance and inhibited tumor growth. Hence, this study could provide an attractive protein cage-based platform for therapeutic drug delivery.
Collapse
Affiliation(s)
- Chufan Wang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Cheng Xiao
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Yurong Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Yao Li
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Qiang Zhang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Wenjun Shan
- Department of Pharmacology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing 400038, P. R. China
| | - Yulin Li
- Henan Bioengineering Research Center, Zhengdong New District, Zhengzhou, China
| | - Shengli Bi
- Chinese Center for Disease Control & Prevention, Institute Viral Disease Control & Prevention, Beijing, P. R. China
| | - Yunlong Wang
- Henan Bioengineering Research Center, Zhengdong New District, Zhengzhou, China
| | - Xiumin Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Lei Ren
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
- State Key Lab of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, P. R. China
| |
Collapse
|
24
|
Brun MK, Bjørlykke KH, Viken MK, Stenvik GE, Klaasen RA, Gehin JE, Warren DJ, Sexton J, Sandanger Ø, Kvien TK, Mørk C, Haavardsholm EA, Jahnsen J, Goll GL, Lie BA, Bolstad N, Jørgensen KK, Syversen SW. HLA-DQ2 is associated with anti-drug antibody formation to infliximab in patients with immune-mediated inflammatory diseases. J Intern Med 2023; 293:648-655. [PMID: 36843323 DOI: 10.1111/joim.13616] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
BACKGROUND Immunogenicity to tumour necrosis factor inhibitors is a significant clinical problem leading to treatment failure and adverse events. The study aimed to assess human leukocyte antigen (HLA) associations with anti-drug antibody (ADAb) formation to infliximab. METHODS Immune-mediated inflammatory disease patients on infliximab therapy (n = 612) were included. Neutralising ADAb were assessed with a drug-sensitive assay. Next generation sequencing-based HLA typing was performed. RESULTS Overall, 147 (24%) patients developed ADAb. Conditional analyses indicated HLA-DQB1 (p = 1.4 × 10-6 ) as a primary risk locus. Highest risk of ADAb was seen when carrying at least one of the HLA-DQ2 haplotypes; DQB1*02:01-DQA1*05:01 or DQB1*02:02-DQA1*02:01 (OR 3.18, 95% CI 2.15-4.69 and p = 5.9 × 10-9 ). Results were consistent across diseases and when adjusting for concomitant immunomodulator. Computational predictions indicated that these HLA-DQ2 haplotypes bind to peptide motifs from infliximab light chain. CONCLUSION A genome-wide significant association between two HLA-DQ2 haplotypes and the risk of ADAb formation to infliximab was identified, suggesting that HLA-DQ2 testing may facilitate personalised treatment decisions.
Collapse
Affiliation(s)
- Marthe Kirkesaether Brun
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kristin Hammersbøen Bjørlykke
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Marte K Viken
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Grethe-Elisabeth Stenvik
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Rolf A Klaasen
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Johanna E Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - David John Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Joseph Sexton
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | | | - Tore K Kvien
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Cato Mørk
- Akershus Dermatology Center, Lørenskog, Norway
| | - Espen A Haavardsholm
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Guro Løvik Goll
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Benedicte A Lie
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Medical Genetics, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Nils Bolstad
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | | | - Silje Watterdal Syversen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
25
|
A Review on Forced Degradation Strategies to Establish the Stability of Therapeutic Peptide Formulations. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
26
|
Karle AC, Wrobel MB, Koepke S, Gutknecht M, Gottlieb S, Christen B, Rubic-Schneider T, Pruimboom-Brees I, Leber XC, Scharenberg M, Maciejewski B, Turner O, Saravanan C, Huet F, Littlewood-Evans A, Clemens A, Grosskreutz CL, Kearns JD, Mehan P, Schmouder RL, Sasseville V, Brees D. Anti-brolucizumab immune response as one prerequisite for rare retinal vasculitis/retinal vascular occlusion adverse events. Sci Transl Med 2023; 15:eabq5241. [PMID: 36724238 DOI: 10.1126/scitranslmed.abq5241] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In October 2019, Novartis launched brolucizumab, a single-chain variable fragment molecule targeting vascular endothelial growth factor A, for the treatment of neovascular age-related macular degeneration. In 2020, rare cases of retinal vasculitis and/or retinal vascular occlusion (RV/RO) were reported, often during the first few months after treatment initiation, consistent with a possible immunologic pathobiology. This finding was inconsistent with preclinical studies in cynomolgus monkeys that demonstrated no drug-related intraocular inflammation, or RV/RO, despite the presence of preexisting and treatment-emergent antidrug antibodies (ADAs) in some animals. In this study, the immune response against brolucizumab in humans was assessed using samples from clinical trials and clinical practice. In the brolucizumab-naïve population, anti-brolucizumab ADA responses were detected before any treatment, which was supported by the finding that healthy donors can harbor brolucizumab-specific B cells. This suggested prior exposure of the immune system to proteins with structural similarity. Experiments on samples showed that naïve and brolucizumab-treated ADA-positive patients developed a class-switched, high-affinity immune response, with several linear epitopes being recognized by ADAs. Only patients with RV/RO showed a meaningful T cell response upon recall with brolucizumab. Further studies in cynomolgus monkeys preimmunized against brolucizumab with adjuvant followed by intravitreal brolucizumab challenge demonstrated that high ADA titers were required to generate ocular inflammation and vasculitis/vascular thrombosis, comparable to RV/RO in humans. Immunogenicity therefore seems to be a prerequisite to develop RV/RO. However, because only 2.1% of patients with ADA develop RV/RO, additional factors must play a role in the development of RV/RO.
Collapse
Affiliation(s)
- Anette C Karle
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Matthias B Wrobel
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Stephan Koepke
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Michael Gutknecht
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Sascha Gottlieb
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Brigitte Christen
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | | | | | | | - Meike Scharenberg
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | | | - Oliver Turner
- Novartis Institutes for BioMedical Research, East Hanover, NJ 07960, USA
| | - Chandra Saravanan
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Francois Huet
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | | | - Andreas Clemens
- Medical Affairs Region Europe, Novartis Pharma AG, Basel CH-4056, Switzerland
| | | | - Jeffrey D Kearns
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Pawan Mehan
- TRD Biologics & CGT, Novartis Pharma AG, Basel CH-4056, Switzerland
| | - Robert L Schmouder
- Novartis Institutes for BioMedical Research, East Hanover, NJ 07960, USA
| | - Vito Sasseville
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Dominique Brees
- Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| |
Collapse
|
27
|
Desai M, Kundu A, Hageman M, Lou H, Boisvert D. Monoclonal antibody and protein therapeutic formulations for subcutaneous delivery: high-concentration, low-volume vs. low-concentration, high-volume. MAbs 2023; 15:2285277. [PMID: 38013454 DOI: 10.1080/19420862.2023.2285277] [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: 10/05/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Biologic drugs are used to treat a variety of cancers and chronic diseases. While most of these treatments are administered intravenously by trained healthcare professionals, a noticeable trend has emerged favoring subcutaneous (SC) administration. SC administration of biologics poses several challenges. Biologic drugs often require higher doses for optimal efficacy, surpassing the low volume capacity of traditional SC delivery methods like autoinjectors. Consequently, high concentrations of active ingredients are needed, creating time-consuming formulation obstacles. Alternatives to traditional SC delivery systems are therefore needed to support higher-volume biologic formulations and to reduce development time and other risks associated with high-concentration biologic formulations. Here, we outline key considerations for SC biologic drug formulations and delivery and explore a paradigm shift: the flexibility afforded by low-to-moderate-concentration drugs in high-volume formulations as an alternative to the traditionally difficult approach of high-concentration, low-volume SC formulation delivery.
Collapse
Affiliation(s)
- M Desai
- Medical Affairs, Enable Injections, Inc, Cincinnati, OH, USA
| | - A Kundu
- Manufacturing Sciences, Takeda Pharmaceuticals, Brooklyn Park, MN, USA
| | - M Hageman
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| | - H Lou
- Biopharmaceutical Innovation & Optimization Center, The University of Kansas, Lawrence, KS, USA
| | - D Boisvert
- Independent Chemistry Manufacturing & Controls (CMC) Consultant, El Cerrito, CA, USA
| |
Collapse
|
28
|
Liang S, Zhang C. PITHA: A Webtool to Predict Immunogenicity for Humanized and Fully Human Therapeutic Antibodies. Methods Mol Biol 2023; 2552:143-150. [PMID: 36346590 DOI: 10.1007/978-1-0716-2609-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Immunogenicity is an important concern to therapeutic antibodies during antibody design and development. Based on the co-crystal structures of idiotypic antibodies and their antibodies, one can see that anti-idiotypic antibodies usually bind the complementarity-determining regions (CDR) of idiotypic antibodies. Sequence and structural features, such as cavity volume at the CDR region and hydrophobicity of CDR-H3 loop region, were identified for distinguishing immunogenic antibodies from non-immunogenic antibodies. These features were integrated together with a machine learning platform to predict immunogenicity for humanized and fully human therapeutic antibodies (PITHA). This method achieved an accuracy of 83% in a leave-one-out experiment for 29 therapeutic antibodies with available crystal structures. The web server of this method is accessible at http://mabmedicine.com/PITHA or http://sysbio.unl.edu/PITHA . This method, as a step of computer-aided antibody design, helps evaluate the safety of new therapeutic antibody, which can save time and money during the therapeutic antibody development.
Collapse
Affiliation(s)
- Shide Liang
- Department of Research and Development, Bio-Thera Solutions, Guangzhou, P. R. China.
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA.
| |
Collapse
|
29
|
Han Q, Bao Z, Luo MZ, Zhang JY. Assessment of innate immune response modulating impurities in glucagon for injection. PLoS One 2022; 17:e0277922. [PMID: 36409759 PMCID: PMC9678265 DOI: 10.1371/journal.pone.0277922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/02/2022] [Indexed: 11/22/2022] Open
Abstract
Glucagon for Injection is a polypeptide hormone medication used to treat patients with severe hypoglycemia or low blood sugar. Only recently, was a generic version of glucagon for injection approved by the FDA. While the generic version was deemed equivalent to its brand-name counterpart, the two glucagon products were produced using different manufacturing processes. The brand-name glucagon is produced via recombinant DNA while the generic glucagon is produced by peptide synthesis. Different manufacturing processes can result in different levels of innate immune response modulating impurities (IIRMIs). This study utilized a cell-based assay method, which allows for detection of a broad spectrum of impurities, to investigate the IIRMI risks of the generic glucagon to make sure it has similar or less immunogenicity risks than the brand-name glucagon product. Three commercial cell lines (RAW-Blue™, HEK-Blue™-hNOD1 and HEK-Blue™-hNOD2) carrying a secreted embryonic alkaline phosphatase reporter construct were used to quantify the level of innate immune responses after being treated with the glucagon drugs. The study results showed that despite differences in manufacturing process, the innate immunogenicity risk in the synthetic (generic) glucagon was at negligible level and comparable to the recombinant (brand-name) glucagon product.
Collapse
Affiliation(s)
- Qingxia Han
- Amphastar Pharmaceuticals, Inc., Rancho Cucamonga, California, United States of America
| | - Zhongli Bao
- Amphastar Pharmaceuticals, Inc., Rancho Cucamonga, California, United States of America
| | - Mary Ziping Luo
- Amphastar Pharmaceuticals, Inc., Rancho Cucamonga, California, United States of America
| | - Jack Yongfeng Zhang
- Amphastar Pharmaceuticals, Inc., Rancho Cucamonga, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Gehin JE, Goll GL, Brun MK, Jani M, Bolstad N, Syversen SW. Assessing Immunogenicity of Biologic Drugs in Inflammatory Joint Diseases: Progress Towards Personalized Medicine. BioDrugs 2022; 36:731-748. [PMID: 36315391 PMCID: PMC9649489 DOI: 10.1007/s40259-022-00559-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 11/30/2022]
Abstract
Biologic drugs have greatly improved treatment outcomes of inflammatory joint diseases, but a substantial proportion of patients either do not respond to treatment or lose response over time. Drug immunogenicity, manifested as the formation of anti-drug antibodies (ADAb), constitute a significant clinical problem. Anti-drug antibodies influence the pharmacokinetics of the drug, are associated with reduced clinical efficacy, and an increased risk of adverse events such as infusion reactions. The prevalence of ADAb differs among drugs and diseases, and the detection of ADAb also depends on the assay format. Most data exist for the tumor necrosis factor-alpha inhibitors infliximab and adalimumab, with a frequency of ADAb that ranges from 10 to 60% across studies. Measurement of ADAb and serum drug concentrations, therapeutic drug monitoring, has been suggested as a strategy to optimize therapy with biologic drugs. Although the recent randomized clinical Norwegian Drug Monitoring (NOR-DRUM) trials show promise towards a personalized medicine prescribing approach by therapeutic drug monitoring, several challenges remain. A plethora of assay formats, with widely differing properties, is currently used for measuring ADAb. Comparing results between different assays and laboratories is difficult, which complicates the development of cut-offs necessary for guidelines and the implementation of ADAb measurements in clinical practice. With the possible exception of infliximab, limited data on clinical relevance and cost effectiveness exist to support therapeutic drug monitoring as a routine clinical strategy to monitor biologic drugs in inflammatory joint diseases. The aim of this review is to provide an overview of the characteristics and prevalence of ADAb, predisposing factors to ADAb formation, commonly used assessment methods, clinical consequences of ADAb, and the potential implications of ADAb assessments for everyday treatment of inflammatory joint diseases.
Collapse
Affiliation(s)
- Johanna Elin Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Radiumhospitalet, Nydalen, Box 4953, 0424, Oslo, Norway.
| | - Guro Løvik Goll
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Marthe Kirkesæther Brun
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Meghna Jani
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK
- Department of Rheumatology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Nils Bolstad
- Department of Medical Biochemistry, Oslo University Hospital, Radiumhospitalet, Nydalen, Box 4953, 0424, Oslo, Norway
| | - Silje Watterdal Syversen
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
31
|
A humanized minipig model for the toxicological testing of therapeutic recombinant antibodies. Nat Biomed Eng 2022; 6:1248-1256. [PMID: 36138193 DOI: 10.1038/s41551-022-00921-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 07/01/2022] [Indexed: 11/08/2022]
Abstract
The safety of most human recombinant proteins can be evaluated in transgenic mice tolerant to specific human proteins. However, owing to insufficient genetic diversity and to fundamental differences in immune mechanisms, small-animal models of human diseases are often unsuitable for immunogenicity testing and for predicting adverse outcomes in human patients. Most human therapeutic antibodies trigger xenogeneic responses in wild-type animals and thus rapid clearance of the drugs, which makes in vivo toxicological testing of human antibodies challenging. Here we report the generation of Göttingen minipigs carrying a mini-repertoire of human genes for the immunoglobulin heavy chains γ1 and γ4 and the immunoglobulin light chain κ. In line with observations in human patients, the genetically modified minipigs tolerated the clinically non-immunogenic IgG1κ-isotype monoclonal antibodies daratumumab and bevacizumab, and elicited antibodies against the checkpoint inhibitor atezolizumab and the engineered interleukin cergutuzumab amunaleukin. The humanized minipigs can facilitate the safety and efficacy testing of therapeutic antibodies.
Collapse
|
32
|
Colangelo GS, Di Ianni A, Cowan K, Riccardi Sirtori F, Barbero LM. Development of a Liquid Chromatography and High-Resolution and -Accuracy Mass Spectrometry Method to Evaluate New Biotherapeutic Entity Processing in Human Liver Lysosomes. Immunohorizons 2022; 7:467-479. [PMID: 37327020 PMCID: PMC10580112 DOI: 10.4049/immunohorizons.2300035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Biotherapeutic immunogenicity remains a great challenge for researchers because multiple factors trigger immune responses. Predicting and assessing the potential human immune response against biological drugs could represent an impressive breakthrough toward generating potentially safer and more efficacious therapeutic proteins. This article describes an in vitro assay that can contribute to evaluating the potential immunogenicity of biotherapeutics by focusing on lysosomal proteolysis. We selected human liver lysosomes (hLLs) from four different donors as a surrogate in vitro model instead of APC lysosomes because they are a ready-to-use lysosomal source. To assess the biological comparability of this surrogate to APC lysosomal extract, we compared the proteome content of hLLs with literature data of lysosomal fractions extracted from murine bone marrow and human blood-derived dendritic cells. Then we tested infliximab (IFX; Remicade) under different proteolytic conditions using liquid chromatography and high-resolution and -accuracy mass spectrometry to better define the degradation kinetics inside the lysosomes. hLLs revealed similar enzymatic content compared with human and murine dendritic cell lysosomes. Degradation assays demonstrated that our liquid chromatography and high-resolution and -accuracy mass spectrometry method could identify both the intact protein and the peptides resulting from proteolysis with high specificity and resolution. The rapid and easy assay described in this article can be extremely useful for evaluating the immunogenic risk associated with therapeutic proteins. In addition, this method can complement information from MHC class II-associated peptide proteomics assays and other in vitro and in silico techniques.
Collapse
Affiliation(s)
- Gabriele Sergio Colangelo
- University of Turin, Molecular Biotechnology Center, Turin, Italy
- NBE-DMPK Innovative BioAnalytics, RBM Merck S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Colleretto Giacosa, Torino, Italy
| | - Andrea Di Ianni
- University of Turin, Molecular Biotechnology Center, Turin, Italy
- NBE-DMPK Innovative BioAnalytics, RBM Merck S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Colleretto Giacosa, Torino, Italy
| | - Kyra Cowan
- New Biological Entities, Drug Metabolism and Pharmacokinetics, Research and Development, Merck KGaA, Darmstadt, Germany
| | - Federico Riccardi Sirtori
- NBE-DMPK Innovative BioAnalytics, RBM Merck S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Colleretto Giacosa, Torino, Italy
| | - Luca Maria Barbero
- NBE-DMPK Innovative BioAnalytics, RBM Merck S.p.A., an affiliate of Merck KGaA, Darmstadt, Germany, Colleretto Giacosa, Torino, Italy
| |
Collapse
|
33
|
Brinth AR, Svenson K, Mosyak L, Cunningham O, Hickling T, Lambert M. Crystal structure of ultra-humanized anti-pTau Fab reveals how germline substitutions humanize CDRs without loss of binding'. Sci Rep 2022; 12:8699. [PMID: 35610505 PMCID: PMC9130293 DOI: 10.1038/s41598-022-12838-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 05/10/2022] [Indexed: 11/08/2022] Open
Abstract
Administration of therapeutic antibodies can elicit adverse immune responses in patients through the generation of anti-drug antibodies that, in turn, reduce the efficacy of the therapeutic. Removal of foreign amino acid content by humanization can lower the immunogenic risk of the therapeutic mAb. We previously developed the ultra-humanization technology "Augmented Binary Substitution" (ABS) which enables single-step CDR germlining of antibodies. The application of ABS to a chicken anti-pTau antibody generated an ultra-humanized variant, anti-pTau C21-ABS, with increased human amino acid content in the CDRs and reduced in-silico predicted immunogenicity risk. Here, we report the high-resolution crystal structure of anti-pTau C21-ABS Fab in complex with the pTau peptide (7KQK). This study examines how ultra-humanization, via CDR germlining, is facilitated while maintaining near-identical antigen affinity (within 1.6-fold). The co-complex structure reveals that the ABS molecule targets the same antigenic epitope, accommodated by structurally-similar changes in the paratope. These findings confirm that ABS enables the germlining of amino acids within CDRs by exploiting CDR plasticity, to reduce non-human amino acid CDR content, with few alterations to the overall mechanism of binding.
Collapse
Affiliation(s)
- Alette R Brinth
- BioMedicine Design, Pfizer Worldwide R&D, Dublin, D22 V8F8, Ireland
| | - Kristine Svenson
- BioMedicine Design, Pfizer Worldwide R&D, Cambridge, MA, 02139, USA
| | - Lidia Mosyak
- BioMedicine Design, Pfizer Worldwide R&D, Cambridge, MA, 02139, USA
| | - Orla Cunningham
- Ultrahuman Ltd. Kreston Reeves LLP Innovation Hs, Ramsgate Rd, Sandwich, CT13 9FF, UK
| | - Timothy Hickling
- BioMedicine Design, Pfizer Worldwide R&D, Andover, MA, 01810, USA
| | - Matthew Lambert
- BioMedicine Design, Pfizer Worldwide R&D, Dublin, D22 V8F8, Ireland.
| |
Collapse
|
34
|
Introducing dendritic cell antibody internalization as an immunogenicity risk assessment tool. Bioanalysis 2022; 14:703-713. [PMID: 35593734 DOI: 10.4155/bio-2022-0024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Immunogenicity risk assessment assays are powerful tools that assess the relative immunogenicity of potential biotherapeutics. We detail here the development of a novel assay that measures the degree of antibody internalization by antigen-presenting cells as a predictor of immunogenicity. Results & methodology: The assay uses the fluorescence signal from the antibody bound to the outside of the cell as well as inside the cell to determine internalization. To calculate the amount of internalized antibody, the fluorescent signal from the outside was subtracted from the fluorescent signal from the inside, which is referred to as the internalization index. Conclusion: This assay format demonstrated that antibody-based biotherapeutics with higher clinical immunogenicity internalized to a higher degree than therapeutic antibodies with lower clinical immunogenicity.
Collapse
|
35
|
ElGamacy M. Accelerating therapeutic protein design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:85-118. [PMID: 35534117 DOI: 10.1016/bs.apcsb.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Protein structures provide for defined microenvironments that can support complex pharmacological functions, otherwise unachievable by small molecules. The advent of therapeutic proteins has thus greatly broadened the range of manageable disorders. Leveraging the knowledge and recent advances in de novo protein design methods has the prospect of revolutionizing how protein drugs are discovered and developed. This review lays out the main challenges facing therapeutic proteins discovery and development, and how present and future advancements of protein design can accelerate the protein drug pipelines.
Collapse
Affiliation(s)
- Mohammad ElGamacy
- University Hospital Tübingen, Division of Translational Oncology, Tübingen, Germany; Max Planck Institute for Biology, Tübingen, Germany.
| |
Collapse
|
36
|
Busch M, Pfeil JM, Dähmcke M, Brauckmann T, Großjohann R, Chisci V, Hunfeld E, Eilts S, Omran W, Morawiec‐Kisiel E, Schulz D, Paul S, Tayar A, Bründer M, Grundel B, Küstner M, Stahl A. Anti-drug antibodies to brolucizumab and ranibizumab in serum and vitreous of patients with ocular disease. Acta Ophthalmol 2022; 100:903-910. [PMID: 35225432 DOI: 10.1111/aos.15124] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/26/2022] [Accepted: 02/18/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Martin Busch
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Johanna M. Pfeil
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Merlin Dähmcke
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Tara Brauckmann
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Rico Großjohann
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Viola Chisci
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Elisabeth Hunfeld
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Sonja Eilts
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Wael Omran
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Ewa Morawiec‐Kisiel
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Daniel Schulz
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Sebastian Paul
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | - Allam Tayar
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | | | - Bastian Grundel
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| | | | - Andreas Stahl
- Department of Ophthalmology University Medical Center Greifswald Greifswald Germany
| |
Collapse
|
37
|
Jawa V, Maamary J, Swanson M, Zhang S, Montgomery D. Implementing a Clinical Immunogenicity Strategy using Preclinical Risk Assessment Outputs. J Pharm Sci 2022; 111:960-969. [PMID: 35122828 DOI: 10.1016/j.xphs.2022.01.032] [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: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/27/2022]
Abstract
Immunogenicity to biologics is often observed following dosing in human subjects during clinical trials. Both product and host specific factors may be implicated in contributing to a potential immune response. However, even if such risk factors are identified and eliminated as part of the rational quality by design approaches, the outcome in clinic can be uncertain and challenging to predict. Several tools have been employed to identify these risk factors and consequent mitigation approaches implemented prior to dosing in humans. However, the complexity of the immune system with an interplay of network of immune cells involved in driving a long- term immune response as well as patient characteristics, can make it challenging to predict the outcome in clinic. This perspective will provide an insight into recent advances in the risk assessment approaches that are utilized during preclinical stage of development of a biologic. The outputs from such tools can help to rank order and select the most optimal candidate with the least likelihood of an immune response and can further drive the development of a clinical bioanalytical and immunogenicity monitoring strategy. Such a strategy can be proactively shared with the regulators along with the proposal to streamline clinical immunogenicity and personalizing the outcome based on pharmacogenomics and other patient-related factors. This paper provides a roadmap on performing risk assessments through a systematic identification of risks and their mitigations wherever possible. Recommendations on incorporating the key components of such risk assessments as part of the new regulatory submissions are also provided. Shorter abstract Immunogenicity to biologics is common during clinical trials. Both product and host specific factors have been implicated. Several risk assessment tools can be used to identify and mitigate the risk factors responsible for immunogenicity. An insight into recent advances in the risk assessment approaches will be presented. The outputs can define a risk score and guide the clinical bioanalytical and immunogenicity monitoring strategy. A roadmap on performing risk assessments through a systematic identification of risks and their mitigations wherever possible is provided. Best practices for a risk assessment strategy and recommendations on the content for IND and the Integrated summary of Immunogenicity are also provided.
Collapse
Affiliation(s)
- Vibha Jawa
- Nonclinical Disposition and Bioanalysis, Bristol-Myers Squibb, Princeton, NJ 08648, USA.
| | - Jad Maamary
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| | - Michael Swanson
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| | - Shuli Zhang
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| | - Diana Montgomery
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| |
Collapse
|
38
|
Nihira K. [Non-clinical safety evaluations of next-generation therapeutic antibodies]. Nihon Yakurigaku Zasshi 2022; 157:458-463. [PMID: 36328561 DOI: 10.1254/fpj.22049] [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: 06/16/2023]
Abstract
Since the first monoclonal antibody was approved by FDA in 1986, numerous therapeutic antibodies have been developed along with advances in antibody engineering and finally, the number of approved therapeutic antibodies by FDA exceeded 100 in 2021. Although therapeutic antibodies were thought to be safer than conventional small molecule drugs, non-clinical safety evaluations of antibodies become more important because antibody-specific toxicity has been found. The depletion of target molecules for antibody drugs is a problem due to the limited number of promising targets. However, to overcome this, some technologies which can enhance the activity of antibodies and enable to add new functions to antibodies have been developed. In particular, bispecific antibodies and antibody-drug conjugates are considered to be attracting technologies as the next-generation antibody modalities. Several drugs applying these technologies have been approved in a recent decade. On the other hand, there is still plenty of room for improvement in these technologies which sometimes occur unexpected toxicity, so the safety of drugs applying these technologies should be carefully evaluated. Here some important considerations for the non-clinical safety evaluations of bispecific antibodies and antibody-drug conjugates are discussed based on a literature review of these types of antibody drugs (approved or terminated due to toxicity).
Collapse
Affiliation(s)
- Kaito Nihira
- Toxicological Research Laboratories, Translational Research Unit, Kyowa Kirin Co., Ltd
| |
Collapse
|
39
|
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: 5] [Impact Index Per Article: 1.7] [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
|
40
|
Wilkinson I, Anderson S, Fry J, Julien LA, Neville D, Qureshi O, Watts G, Hale G. Fc-engineered antibodies with immune effector functions completely abolished. PLoS One 2021; 16:e0260954. [PMID: 34932587 PMCID: PMC8691596 DOI: 10.1371/journal.pone.0260954] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/20/2021] [Indexed: 01/12/2023] Open
Abstract
Elimination of the binding of immunoglobulin Fc to Fc gamma receptors (FcγR) is highly desirable for the avoidance of unwanted inflammatory responses to therapeutic antibodies and fusion proteins. Many different approaches have been described in the literature but none of them completely eliminates binding to all of the Fcγ receptors. Here we describe a set of novel variants having specific amino acid substitutions in the Fc region at L234 and L235 combined with the substitution G236R. They show no detectable binding to Fcγ receptors or to C1q, are inactive in functional cell-based assays and do not elicit inflammatory cytokine responses. Meanwhile, binding to FcRn, manufacturability, stability and potential for immunogenicity are unaffected. These variants have the potential to improve the safety and efficacy of therapeutic antibodies and Fc fusion proteins.
Collapse
Affiliation(s)
- Ian Wilkinson
- Absolute Antibody Ltd, Wilton, United Kingdom
- mAbsolve Limited, Oxford, United Kingdom
| | | | - Jeremy Fry
- ProImmune Limited, Oxford, United Kingdom
| | | | - David Neville
- Reading Scientific Services Limited, Reading, United Kingdom
| | | | - Gary Watts
- Abzena Limited, Babraham, United Kingdom
| | - Geoff Hale
- mAbsolve Limited, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
41
|
Richard G, Princiotta MF, Bridon D, Martin WD, Steinberg GD, De Groot AS. Neoantigen-based personalized cancer vaccines: the emergence of precision cancer immunotherapy. Expert Rev Vaccines 2021; 21:173-184. [PMID: 34882038 DOI: 10.1080/14760584.2022.2012456] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The field of cancer therapy has undergone a major transformation in less than a decade due to the introduction of checkpoint inhibitors, the advent of next generation sequencing and the discovery of neoantigens. The key observation that the breadth of each patient's immune response to the unique mutations or neoantigens present in their tumor is directly related to their survival has led oncologists to focus on driving immune responses to neoantigens through vaccination. Oncology has entered the era of precision immunotherapy, and cancer vaccine development is undergoing a paradigm shift. AREAS COVERED Neoantigens are short peptide sequences found in tumors, but not noncancerous tissues, the vast majority of which are unique to each patient. In addition to providing a description of the distinguishing features of neoantigen discovery platforms, this review will address cross-cutting personalized cancer vaccine design themes and developmental stumbling blocks. EXPERT OPINION Immunoinformatic pipelines that can rapidly scan cancer genomes and identify 'the best' neoantigens are in high demand. Despite the need for such tools, immunoinformatic methods for identifying neoepitopes in cancer genomes are diverse and have not been well-validated. Validation of 'personalized vaccine design pipelines' will bring about a revolution in neoantigen-based vaccine design and delivery.
Collapse
Affiliation(s)
| | | | | | | | - Gary D Steinberg
- EpiVax Therapeutics, Inc., Providence, RI, USA.,Perlmutter Cancer Center, Department of Urology at NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI, USA.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| |
Collapse
|
42
|
An In Vitro Assessment of Immunostimulatory Responses to Ten Model Innate Immune Response Modulating Impurities (IIRMIs) and Peptide Drug Product, Teriparatide. Molecules 2021; 26:molecules26247461. [PMID: 34946542 PMCID: PMC8707785 DOI: 10.3390/molecules26247461] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding, predicting, and minimizing the immunogenicity of peptide-based therapeutics are of paramount importance for ensuring the safety and efficacy of these products. The so-called anti-drug antibodies (ADA) may have various clinical consequences, including but not limited to the alteration in the product's distribution, biological activity, and clearance profiles. The immunogenicity of biotherapeutics can be influenced by immunostimulation triggered by the presence of innate immune response modulating impurities (IIRMIs) inadvertently introduced during the manufacturing process. Herein, we evaluate the applicability of several in vitro assays (i.e., complement activation, leukocyte proliferation, and cytokine secretion) for the screening of innate immune responses induced by ten common IIRMIs (Bacillus subtilis flagellin, FSL-1, zymosan, ODN2006, poly(I:C) HMW, poly(I:C) LMW, CLO75, MDP, ODN2216, and Escherichia coli O111:B4 LPS), and a model biotherapeutic Forteo™ (teriparatide). Our study identifies cytokine secretion from healthy human donor peripheral blood mononuclear cells (PBMC) as a sensitive method for the in vitro monitoring of innate immune responses to individual IIRMIs and teriparatide (TP). We identify signature cytokines, evaluate both broad and narrow multiplex cytokine panels, and discuss how the assay logistics influence the performance of this in vitro assay.
Collapse
|
43
|
Innate Immunity Modulating Impurities and the Immunotoxicity of Nanobiotechnology-Based Drug Products. Molecules 2021; 26:molecules26237308. [PMID: 34885886 PMCID: PMC8658779 DOI: 10.3390/molecules26237308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 12/19/2022] Open
Abstract
Innate immunity can be triggered by the presence of microbial antigens and other contaminants inadvertently introduced during the manufacture and purification of bionanopharmaceutical products. Activation of these innate immune responses, including cytokine secretion, complement, and immune cell activation, can result in unexpected and undesirable host immune responses. These innate modulators can also potentially stimulate the activation of adaptive immune responses, including the formation of anti-drug antibodies which can impact drug effectiveness. To prevent induction of these adverse responses, it is important to detect and quantify levels of these innate immunity modulating impurities (IIMIs) that may be present in drug products. However, while it is universally agreed that removal of IIMIs from drug products is crucial for patient safety and to prevent long-term immunogenicity, there is no single assay capable of directly detecting all potential IIMIs or indirectly quantifying downstream biomarkers. Additionally, there is a lack of agreement as to which of the many analytical assays currently employed should be standardized for general IIMI screening. Herein, we review the available literature to highlight cellular and molecular mechanisms underlying IIMI-mediated inflammation and its relevance to the safety and efficacy of pharmaceutical products. We further discuss methodologies used for direct and indirect IIMI identification and quantification.
Collapse
|
44
|
The evolution of commercial drug delivery technologies. Nat Biomed Eng 2021; 5:951-967. [PMID: 33795852 DOI: 10.1038/s41551-021-00698-w] [Citation(s) in RCA: 637] [Impact Index Per Article: 159.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Drug delivery technologies have enabled the development of many pharmaceutical products that improve patient health by enhancing the delivery of a therapeutic to its target site, minimizing off-target accumulation and facilitating patient compliance. As therapeutic modalities expanded beyond small molecules to include nucleic acids, peptides, proteins and antibodies, drug delivery technologies were adapted to address the challenges that emerged. In this Review Article, we discuss seminal approaches that led to the development of successful therapeutic products involving small molecules and macromolecules, identify three drug delivery paradigms that form the basis of contemporary drug delivery and discuss how they have aided the initial clinical successes of each class of therapeutic. We also outline how the paradigms will contribute to the delivery of live-cell therapies.
Collapse
|
45
|
Lee SC, Ma JSY, Kim MS, Laborda E, Choi SH, Hampton EN, Yun H, Nunez V, Muldong MT, Wu CN, Ma W, Kulidjian AA, Kane CJ, Klyushnichenko V, Woods AK, Joseph SB, Petrassi M, Wisler J, Li J, Jamieson CAM, Schultz PG, Kim CH, Young TS. A PSMA-targeted bispecific antibody for prostate cancer driven by a small-molecule targeting ligand. SCIENCE ADVANCES 2021; 7:eabi8193. [PMID: 34380625 PMCID: PMC8357232 DOI: 10.1126/sciadv.abi8193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
Despite the development of next-generation antiandrogens, metastatic castration-resistant prostate cancer (mCRPC) remains incurable. Here, we describe a unique semisynthetic bispecific antibody that uses site-specific unnatural amino acid conjugation to combine the potency of a T cell-recruiting anti-CD3 antibody with the specificity of an imaging ligand (DUPA) for prostate-specific membrane antigen. This format enabled optimization of structure and function to produce a candidate (CCW702) with specific, potent in vitro cytotoxicity and improved stability compared with a bispecific single-chain variable fragment format. In vivo, CCW702 eliminated C4-2 xenografts with as few as three weekly subcutaneous doses and prevented growth of PCSD1 patient-derived xenograft tumors in mice. In cynomolgus monkeys, CCW702 was well tolerated up to 34.1 mg/kg per dose, with near-complete subcutaneous bioavailability and a PK profile supporting testing of a weekly dosing regimen in patients. CCW702 is being evaluated in a first in-human clinical trial for men with mCRPC who had progressed on prior therapies (NCT04077021).
Collapse
Affiliation(s)
- Sung Chang Lee
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jennifer S Y Ma
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Min Soo Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eduardo Laborda
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sei-Hyun Choi
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eric N Hampton
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hwayoung Yun
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vanessa Nunez
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michelle T Muldong
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina N Wu
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anna A Kulidjian
- Department of Orthopedic Surgery, Scripps MD Anderson Cancer Center, La Jolla, CA 92093, USA
| | - Christopher J Kane
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vadim Klyushnichenko
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ashley K Woods
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean B Joseph
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mike Petrassi
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John Wisler
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jing Li
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Christina A M Jamieson
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter G Schultz
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chan Hyuk Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Travis S Young
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
46
|
Mason A, Anver H, Lwin M, Holroyd C, Faust SN, Edwards CJ. Lupus, vaccinations and COVID-19: What we know now. Lupus 2021; 30:1541-1552. [PMID: 34134555 DOI: 10.1177/09612033211024355] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the virus causing Coronavirus disease 2019 (COVID-19), has had a huge impact on health services, with a high mortality associated with complications including pneumonia and acute respiratory distress syndrome. Patients with systemic lupus erythematosus (SLE) are at increased risk of viral infections, and recent data suggests they may be at an increased risk of poor outcomes with COVID-19. This may be particularly true for those on rituximab or high dose steroids. A huge international effort from the scientific community has so far resulted in the temporary authorisation of three vaccines which offer protection against SARS-CoV-2, with over 30 other vaccines being evaluated in ongoing trials. Although there has historically been concern that vaccines may trigger disease flares of SLE, there is little convincing evidence to show this. In general lupus patients appear to gain good protection from vaccination, although there may be reduced efficacy in those with high disease activity or those on immunosuppressive therapies, such as rituximab or high dose steroids. Recent concerns have been raised regarding rare clotting events with the AstraZeneca/Oxford vaccine and it is currently unknown whether this risk is higher for those patients with secondary antiphospholipid syndrome. With the possibility of annual COVID vaccination programmes in the future, prospective data collection and registries looking at the effect of vaccination on SLE disease control, the incidence of COVID-19 in SLE patients and severity of COVID-19 disease course would all be useful. As mass vaccination programmes begin to roll out across the world, we assess the evidence of the use of vaccines in SLE patients and in particular vaccines targeting SARS-CoV-2.
Collapse
Affiliation(s)
- Alice Mason
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Himashi Anver
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - May Lwin
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Christopher Holroyd
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christopher J Edwards
- Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
47
|
Dembele M, Tao S, Massoud AH, Miah SMS, Lelias S, De Groot AS, Mazer BD. Tregitopes Improve Asthma by Promoting Highly Suppressive and Antigen-Specific Tregs. Front Immunol 2021; 12:634509. [PMID: 33953711 PMCID: PMC8089381 DOI: 10.3389/fimmu.2021.634509] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/26/2021] [Indexed: 01/09/2023] Open
Abstract
Tregitopes (T regulatory epitopes) are IgG-derived peptides with high affinity to major histocompatibility complex class II (MHCII), that are known to promote tolerance by activating T regulatory cell (Treg) activity. Here we characterized the effect of IgG Tregitopes in a well-established murine model of allergic asthma, demonstrating in vivo antigen-specific tolerance via adoptive transfer of Tregitope-and-allergen-activated Tregs. Asthma is a heterogeneous chronic inflammatory condition affecting the airways and impacting over 300 million individuals worldwide. Treatment is suppressive, and no current therapy addresses immune regulation in severely affected asthmatics. Although high dose intra-venous immunoglobulin (IVIg) is not commonly used in the asthma clinic setting, it has been shown to improve severe asthma in children and in adults. In our laboratory, we previously demonstrated that IVIg abrogates airway hyperresponsiveness (AHR) in a murine model of asthma and induces suppressive antigen-specific T-regulatory cells. We hypothesized that IgG-derived Tregitopes would modulate allergic airway disease by inducing highly suppressive antigen-specific Tregs capable of diminishing T effector cell responses and establishing antigen-specific tolerance. Using ovalbumin (OVA-) and ragweed-driven murine models of allergic airway disease, we characterized the immunoregulatory properties of Tregitopes and performed Treg adoptive transfer to OVA- and ragweed-allergic mice to test for allergen specificity. Treatment with Tregitopes attenuated allergen-induced airway hyperresponsiveness and lung inflammation. We demonstrated that Tregitopes induce highly suppressive allergen-specific Tregs. The tolerogenic action of IgG Tregitopes in our model is very similar to that of IVIg, so we foresee that IgG Tregitopes could potentially replace steroid-based treatment and can offer a synthetic alternative to IVIg in a range of inflammatory and allergic conditions.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Animals, Genetically Modified
- Anti-Asthmatic Agents/pharmacology
- Antigens, Plant
- Asthma/drug therapy
- Asthma/immunology
- Asthma/metabolism
- Asthma/physiopathology
- Bronchoconstriction/drug effects
- Cells, Cultured
- Cytokines/metabolism
- Disease Models, Animal
- Epitopes, T-Lymphocyte/drug effects
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Humans
- Immunoglobulin Fab Fragments/pharmacology
- Immunoglobulin Fc Fragments/pharmacology
- Inflammation Mediators/metabolism
- Lung/drug effects
- Lung/immunology
- Lung/metabolism
- Lung/physiopathology
- Lymphocyte Activation/drug effects
- Mice, Inbred C57BL
- Ovalbumin
- Plant Extracts
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Mice
Collapse
Affiliation(s)
- Marieme Dembele
- The Research Institute of the McGill University Health Centre, Division of Pediatric Allergy Immunology and Dermatology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Shao Tao
- The Research Institute of the McGill University Health Centre, Division of Pediatric Allergy Immunology and Dermatology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Amir H. Massoud
- The Research Institute of the McGill University Health Centre, Division of Pediatric Allergy Immunology and Dermatology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC, Canada
| | | | | | - Anne S. De Groot
- EpiVax, Inc., Providence, RI, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Bruce D. Mazer
- The Research Institute of the McGill University Health Centre, Division of Pediatric Allergy Immunology and Dermatology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
48
|
Bray-French K, Hartman K, Steiner G, Marban-Doran C, Bessa J, Campbell N, Martin-Facklam M, Stubenrauch KG, Solier C, Singer T, Ducret A. Managing the Impact of Immunogenicity in an Era of Immunotherapy: From Bench to Bedside. J Pharm Sci 2021; 110:2575-2584. [PMID: 33812888 DOI: 10.1016/j.xphs.2021.03.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Biotherapeutics have revolutionized our ability to treat life-threatening diseases. Despite clinical success, the use of biotherapeutics has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs). The multifactorial nature of immunogenicity has prevented a standardized approach for assessing this and each of the assessment methods developed so far does not exhibit high enough reliability to be used alone, due to limited predictiveness. This prompted the Roche Pharma Research and Early Development (pRED) Immunogenicity Working Group to establish an internal preclinical immunogenicity toolbox of in vitro/in vivo approaches and accompanying guidelines for a harmonized assessment and management of immunogenicity in early development. In this article, the complex factors influencing immunogenicity and their associated clinical ramifications are discussed to highlight the importance of an end-to-end approach conducted from lead optimization to clinical candidate selection. We then examine the impact of the resulting lead candidate categorization on the design and implementation of a multi-tiered ADA/immunogenicity assay strategy prior to phase I (entry into human) through early clinical development. Ultimately, the Immunogenicity Toolbox ensures that Roche pRED teams are equipped to address immunogenicity in a standardized manner, paving the way for lifesaving products with improved safety and efficacy.
Collapse
Affiliation(s)
- Katharine Bray-French
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Katharina Hartman
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Guido Steiner
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Céline Marban-Doran
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Juliana Bessa
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Neil Campbell
- Global Product Strategy, Pharma Division, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Meret Martin-Facklam
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Kay-Gunnar Stubenrauch
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Munich, Germany
| | - Corinne Solier
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Thomas Singer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Axel Ducret
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland.
| |
Collapse
|
49
|
Rossotti MA, Bélanger K, Henry KA, Tanha J. Immunogenicity and humanization of single‐domain antibodies. FEBS J 2021; 289:4304-4327. [DOI: 10.1111/febs.15809] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/26/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Martin A. Rossotti
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
| | - Kasandra Bélanger
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
| | - Kevin A. Henry
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
- Department of Biochemistry, Microbiology and Immunology Faculty of Medicine University of Ottawa Canada
| | - Jamshid Tanha
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
- Department of Biochemistry, Microbiology and Immunology Faculty of Medicine University of Ottawa Canada
| |
Collapse
|
50
|
Azam A, Mallart S, Illiano S, Duclos O, Prades C, Maillère B. Introduction of Non-natural Amino Acids Into T-Cell Epitopes to Mitigate Peptide-Specific T-Cell Responses. Front Immunol 2021; 12:637963. [PMID: 33777029 PMCID: PMC7991740 DOI: 10.3389/fimmu.2021.637963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/17/2021] [Indexed: 11/18/2022] Open
Abstract
Non-natural modifications are widely introduced into peptides to improve their therapeutic efficacy, but their impact on immunogenicity remains largely unknown. As the CD4 T-cell response is a key factor in triggering immunogenicity, we investigated the effect of introducing D-amino acids (Daa), amino isobutyric acid (Aib), N-methylation, Cα-methylation, reduced amide, and peptoid bonds into an immunoprevalent T-cell epitope on binding to a set of HLA-DR molecules, recognition, and priming of human T cells. Modifications are differentially accepted at multiple positions, but are all tolerated in the flanking regions. Introduction of Aib and Daa in the binding core had the most deleterious effect on binding to HLA-DR molecules and T-cell activation. Their introduction at the positions close to the P1 anchor residue abolished T-cell priming, suggesting they might be sufficient to dampen peptide immunogenicity. Other modifications led to variable effects on binding to HLA-DR molecules and T-cell reactivity, but none exhibited an increased ability to stimulate T cells. Altogether, non-natural modifications appear generally to diminish binding to HLA-DR molecules and hence T-cell stimulation. These data might guide the design of therapeutic peptides to make them less immunogenic.
Collapse
Affiliation(s)
- Aurélien Azam
- Sanofi, Biologics Research, Vitry-sur-Seine, France.,Université de Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Sergio Mallart
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Stephane Illiano
- Sanofi R&D, Cardiovascular Diseases & Metabolism, Chilly-Mazarin, France
| | - Olivier Duclos
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | | | - Bernard Maillère
- Université de Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| |
Collapse
|