1
|
Mora J, Forman D, Hu J, Ijantkar A, Gokemeijer J, Kolaja KL, Picarillo C, Jawa V, Yue H, Lamy J, Denies S, Schockaert J, Ackaert C. Immunogenicity Risk Assessment of Process-Related Impurities in An Engineered T Cell Receptor Cellular Product. J Pharm Sci 2024; 113:2151-2160. [PMID: 38768755 DOI: 10.1016/j.xphs.2024.05.018] [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: 01/23/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
Cell therapies such as genetically modified T cells have emerged as a promising and viable treatment for hematologic cancers and are being aggressively pursued for a wide range of diseases and conditions that were previously difficult to treat or had no cure. The process development requires genetic modifications to T cells to express a receptor (engineered T cell receptor (eTCR)) of specific binding qualities to the desired target. Protein reagents utilized during the cell therapy manufacturing process, to facilitate these genetic modifications, are often present as process-related impurities at residual levels in the final drug product and can represent a potential immunogenicity risk upon infusion. This manuscript presents a framework for the qualification of an assay for assessing the immunogenicity risk of AA6 and Cas9 residuals. The same framework applies for other residuals; however, AAV6 and Cas9 were selected as they were residuals from the manufacturing of an engineered T cell receptor cellular product in development. The manuscript: 1) elucidates theoretical risks, 2) summarizes analytical data collected during process development, 3) describes the qualification of an in vitro human PBMC cytokine release assay to assess immunogenicity risk from cellular product associated process residuals; 4) identifies a multiplexed inflammatory innate and adaptive cytokine panel with pre-defined criteria using relevant positive controls; and 5) discusses qualification challenges and potential solutions for establishing meaningful thresholds. The assessment is not only relevant to establishing safe exposure levels of these residuals but also in guiding risk assessment and CMC strategy during the conduct of clinical trials.
Collapse
Affiliation(s)
- Johanna Mora
- Clinical Pharmacology Pharmacometrics and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States.
| | - Daron Forman
- Discovery Biotherapeutics, Bristol Myers Squibb, Cambridge MA, United States
| | - Jennifer Hu
- Current: Technical Operations, Analytical Development, Gentibio, Seattle, WA, United States
| | - Akshata Ijantkar
- Cell Therapy Product and Analytical Development, Bristol Myers Squibb, Seattle, WA, United States
| | - Jochem Gokemeijer
- Discovery Biotherapeutics, Bristol Myers Squibb, Cambridge MA, United States
| | - Kyle L Kolaja
- Nonclincial Safety, Bristol Meyers Squibb, Summit NJ, United States
| | - Caryn Picarillo
- Discovery Biotherapeutics, Bristol Myers Squibb, Cambridge MA, United States
| | - Vibha Jawa
- Clinical Pharmacology Pharmacometrics and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Hai Yue
- Cell Therapy Product and Analytical Development, Bristol Myers Squibb, Seattle, WA, United States
| | - Juliette Lamy
- ImmunXperts, a Q2 Solutions Company, Gosselies, Belgium
| | - Sofie Denies
- ImmunXperts, a Q2 Solutions Company, Gosselies, Belgium
| | | | - Chloé Ackaert
- ImmunXperts, a Q2 Solutions Company, Gosselies, Belgium
| |
Collapse
|
2
|
Kubheka MX, Ndlovu SI, Mkhwanazi NP. Anti-HIV Activity and Immunomodulatory Properties of Fractionated Crude Extracts of Alternaria alternata. Microorganisms 2024; 12:1150. [PMID: 38930532 PMCID: PMC11205553 DOI: 10.3390/microorganisms12061150] [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: 04/22/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Developing new anti-human immunodeficiency virus (HIV) drug candidates that target different sites in HIV-1 replication, with better resistance profiles and lower drug toxicity, is essential to eradicating HIV. This study investigated the potential of fractionated crude extracts of Alternaria alternata as immunomodulatory or anti-HIV drug candidates. Solid-phase extraction (SPE) was used to fractionate A. alternata PO4PR2 using three different columns: MAX (Mixed-mode, strong Anion-eXchange), MCX (Mixed-mode, strong Cation-eXchange), and HLB (Hydrophilic-Lipophilic Balance) with methanol gradient methods (5%, 45%, and 95%). An MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay was used to assess the cell viability and cytotoxicity of the fractionated crude extract A. alternata PO4PR2 in the TZM-bl cell lines. This was followed by a luciferase-based antiviral assay to assess the antiviral activity of A. alternata PO4PR2. A time of addition (TOA) assay was performed to ascertain the mechanism of inhibition employed by the fractionated crude extract of A. alternata PO4PR2 in the HIV life cycle. The p24 titer was determined using an ELISA, while a luciferase-based antiviral assay was used to evaluate the HIV percentage inhibition for different HIV-1 replication cycles. The TOA assay was established using antiviral drugs that target different sites in the HIV replication cycle. These included maraviroc, azidothymidine, raltegravir, and amprenavir. The immunomodulatory effect of the fractionated crude extracts on CD4+ T cells was measured by a flow cytometric analysis, for which fluorochrome-labelled monoclonal antibodies were used as markers for activation (CD38 and HLA-DR) and exhaustion (PD-1). The MCX fraction demonstrated a more significant anti-HIV inhibition than that of the fractions generated in other columns, with an IC50 of 0.3619 µg/mL, an HIV inhibition of 77%, 5% HLB (IC50: 0.7232 µg/mL; HIV inhibition of 64%), and 5% MAX (IC50: 5.240 µg/mL; HIV inhibition of 67%). It was evident from the time of addition data that the crude extract and the 5% MCX fraction inhibited viral binding (68%), reverse transcription (75%), integration (98%), and proteolysis (77%). It was shown that A. alternata (the MCX fraction) have a significant inhibitory effect on reverse transcription (75% HIV inhibition) and integration (100% HIV inhibition). The 5% MCX (p = 0.0062), 5% HLB (p = 0.0269), and 5% MAX (p = 0.0117) fractionated A. alternata crude extracts had low levels of CD4+ T cell (CD38 + HLA-DR+) activation compared to those of the AZT treatment, while CD4+ T cell activation was insignificant. The 5% MAX and HLB A. alternata fractions may possess immunomodulatory compounds with less anti-HIV-1 activity. A. alternata could be a key source of innovative anti-HIV drugs with immunomodulatory characteristics.
Collapse
Affiliation(s)
- Mbali X. Kubheka
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Sizwe I. Ndlovu
- Department of Biotechnology and Food Technology, Faculty of Science, University of Johannesburg, Johannesburg 2092, South Africa;
| | - Nompumelelo P. Mkhwanazi
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
3
|
Walsh RE, Nix A, Ackaert C, Mazy A, Schockaert J, Pattyn S, Malherbe L. Preclinical immunogenicity risk assessment of biotherapeutics using CD4 T cell assays. Front Immunol 2024; 15:1406040. [PMID: 38863708 PMCID: PMC11165089 DOI: 10.3389/fimmu.2024.1406040] [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: 03/24/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
T-cell dependent antibody responses to biotherapeutics remain a challenge to the optimal clinical application of biotherapeutics because of their capacity to impair drug efficacy and their potential to cause safety issues. To minimize this clinical immunogenicity risk, preclinical assays measuring the capacity of biotherapeutics to elicit CD4 T cell response in vitro are commonly used. However, there is considerable variability in assay formats and a general poor understanding of their respective predictive value. In this study, we evaluated the performance of three different CD4 T cell proliferation assays in their capacity to predict clinical immunogenicity: a CD8 T cell depleted peripheral blood mononuclear cells (PBMC) assay and two co-culture-based assays between dendritic cells (DCs) and autologous CD4 T cells with or without restimulation with monocytes. A panel of 10 antibodies with a wide range of clinical immunogenicity was selected. The CD8 T cell depleted PBMC assay predicted the clinical immunogenicity in four of the eight highly immunogenic antibodies included in the panel. Similarly, five antibodies with high clinical immunogenicity triggered a response in the DC: CD4 T cell assay but the responses were of lower magnitude than the ones observed in the PBMC assay. Remarkably, three antibodies with high clinical immunogenicity did not trigger any response in either platform. The addition of a monocyte restimulation step to the DC: CD4 T cell assay did not further improve its predictive value. Overall, these results indicate that there are no CD4 T cell assay formats that can predict the clinical immunogenicity of all biotherapeutics and reinforce the need to combine results from various preclinical assays assessing antigen uptake and presentation to fully mitigate the immunogenicity risk of biotherapeutics.
Collapse
Affiliation(s)
- Robin E. Walsh
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - Angela Nix
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - Chloé Ackaert
- ImmunXperts SA| Rue August Piccard 48, Gosselies, Belgium
| | - Aurélie Mazy
- ImmunXperts SA| Rue August Piccard 48, Gosselies, Belgium
| | | | - Sofie Pattyn
- ImmunXperts SA| Rue August Piccard 48, Gosselies, Belgium
| | - Laurent Malherbe
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| |
Collapse
|
4
|
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
|
5
|
Nabhan M, Meunier S, Le-Minh V, Robin B, de Bourayne M, Smadja C, Maillère B, Pallardy M, Turbica I. Infliximab aggregates produced in severe and mild elevated temperature stress conditions induce an extended specific CD4 T-cell response. Eur J Pharm Sci 2024; 192:106670. [PMID: 38070782 DOI: 10.1016/j.ejps.2023.106670] [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: 06/27/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/22/2023]
Abstract
Aggregation has been widely described as a factor contributing to therapeutic antibody immunogenicity. Although production of high-affinity anti-drug antibodies depends on the activation of CD4 T lymphocytes, little is known about the T-cell response induced by antibody aggregates, especially for aggregates produced in mild conditions resulting from minor handling errors of vials. Large insoluble infliximab (IFX) aggregates produced in severe elevated temperature stress conditions have been previously shown to induce human monocyte-derived dendritic cell (moDC) maturation. We here showed that large IFX aggregates recruit in vitro a significantly higher number of CD4 T-cells compared to native IFX. Moreover, a larger array of T-cell epitopes encompassing the entire variable regions was evidenced compared to the native antibody. We then compared the responses of moDCs to different types of aggregates generated by submitting IFX to mild conditions of various times of incubation at an elevated temperature. Decreasing stress duration reduced aggregate size and quantity, and subsequently altered moDC activation. Of importance, IFX aggregates generated in mild conditions and not altering moDC phenotype generated an in vitro T-cell response with a higher frequency of CD4 T cells compared to native IFX. Moreover, cross-reactivity studies of aggregate-specific T cells showed that some T cells could recognize both native and aggregated IFX, while others responded only to IFX aggregates. Taken together, our results suggest that aggregation of antibodies in mild elevated temperature stress conditions is sufficient to alter moDC phenotype in a dose-dependent manner and to increase T-cell response.
Collapse
Affiliation(s)
- Myriam Nabhan
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 91 400 Orsay, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Sylvain Meunier
- Université de Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Victor Le-Minh
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91 400 Orsay, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Baptiste Robin
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91 400 Orsay, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Marie de Bourayne
- Université de Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Claire Smadja
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91 400 Orsay, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Bernard Maillère
- Université de Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Marc Pallardy
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 91 400 Orsay, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France
| | - Isabelle Turbica
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 91 400 Orsay, France; Université Paris-Saclay, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LABEX LERMIT), 91400 Orsay, France.
| |
Collapse
|
6
|
Cho J, Yang B, Lee JH, Kim H, Kim H, Go EB, Bak DH, Park SJ, Kwon I, Choi JI, Lee K. In vivo study of newly developed albumin-conjugated urate oxidase for gout treatment. Arthritis Res Ther 2023; 25:247. [PMID: 38111075 PMCID: PMC10726570 DOI: 10.1186/s13075-023-03231-3] [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: 10/18/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Exogenously providing engineered Uox with enhanced half-life is one of the important urate-lowering treatments for gout. The potential of PAT101, a recombinant human albumin (rHA)-conjugated variant, was evaluated and compared as a novel gout treatment through various in vivo studies with PAT101 and competing drugs. METHODS PAT101 was produced by site-specific conjugation of rHA and Aspergillus flavus Uox (AfUox-rHA) through clickable non-natural amino acid (frTet) and Inverse electron demand Diels-Alder (IEDDA) reaction. In vivo pharmacokinetics, efficacy tests and in vitro immunogenetic assay were performed after single or multiple doses of PAT101 and its competitors in BALB/c mice, transgenic (TG) mice, Sprague-Dawley (SD) rats, and non-human primate (NHP). RESULTS The half-life of PAT101 in single-dose treated TG mice was more than doubled compared to pegloticase. In SD rats with 4 weeks of repeated administration of rasburicase, only 24% of Uox activity remained, whereas in PAT101, it was maintained by 86%. In the Uox KO model, the survival rate of PAT101 was comparable to that of pegloticase. In addition, human PBMC-based CD4+/CD8+ T-cell activation analysis demonstrated that PAT101 has a lower immune response compared to the original drug, rasburicase. CONCLUSION All results suggest that this rHA-conjugated AfUox, PAT101, can be provided as a reliable source of Uox for gout treatment.
Collapse
Affiliation(s)
- Jeonghaeng Cho
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Byungseop Yang
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyunwoo Kim
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Hyeongseok Kim
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Eun Byeol Go
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Dong-Ho Bak
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Su Jin Park
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
| | - Inchan Kwon
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jong-Il Choi
- Department of Biotechnology and Bioengineering, Interdisciplinary Program for Bioenergy and Biomaterials, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kyunghee Lee
- Research and Development, ProAbTech Co., Ltd, Seoul, 07807, Republic of Korea.
| |
Collapse
|
7
|
In vitro and in vivo immunogenicity assessment of protein aggregate characteristics. Int J Pharm 2023; 631:122490. [PMID: 36521637 DOI: 10.1016/j.ijpharm.2022.122490] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The immunogenicity risk of therapeutic protein aggregates has been extensively investigated over the past decades. While it is established that not all aggregates are equally immunogenic, the specific aggregate characteristics, which are most likely to induce an immune response, remain ambiguous. The aim of this study was to perform comprehensive in vitro and in vivo immunogenicity assessment of human insulin aggregates varying in size, structure and chemical modifications, while keeping other morphological characteristics constant. We found that flexible aggregates with highly altered secondary structure were most immunogenic in all setups, while compact aggregates with native-like structure were found to be immunogenic primarily in vivo. Moreover, sub-visible (1-100 µm) aggregates were found to be more immunogenic than sub-micron (0.1-1 µm) aggregates, while chemical modifications (deamidation, ethylation and covalent dimers) were not found to have any measurable impact on immunogenicity. The findings highlight the importance of utilizing aggregates varying in few characteristics for assessment of immunogenicity risk of specific morphological features and may provide a workflow for reliable particle analysis in biotherapeutics.
Collapse
|
8
|
Arata Y, Motoyama S, Yano M, Ikuno T, Ito S, Matsushita T, Takeiri A, Nishito Y, Yabuki N, Mizuno H, Sampei Z, Mishima M, Honda M, Kiyokawa J, Suzuki H, Chiba S, Tabo M, Kubo C. Rapid in vitro assessment of the immunogenicity potential of engineered antibody therapeutics through detection of CD4 + T cell interleukin-2 secretion. MAbs 2023; 15:2253570. [PMID: 37682072 PMCID: PMC10494738 DOI: 10.1080/19420862.2023.2253570] [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: 03/14/2023] [Revised: 08/15/2023] [Accepted: 08/27/2023] [Indexed: 09/09/2023] Open
Abstract
Therapeutic antibodies sometimes elicit anti-drug antibodies (ADAs) that can affect efficacy and safety. Engineered antibodies that contain artificial amino acid sequences are potentially highly immunogenic, but this is currently difficult to predict. Therefore, it is important to efficiently assess immunogenicity during the development of complex antibody-based formats. Here, we present an in vitro peripheral blood mononuclear cell-based assay that can be used to assess immunogenicity potential within 3 days. This method involves examining the frequency and function of interleukin (IL)-2-secreting CD4+ T cells induced by therapeutic antibodies. IL-2-secreting CD4+ T cells seem to be functionally relevant to the immunogenic potential due to their proliferative activity and the expression of several cytokines. The rates of the donors responding to low and high immunogenic proteins, mAb1, and keyhole limpet hemocyanin were 1.3% and 93.5%, respectively. Seven antibodies with known rates of immunogenicity (etanercept, emicizumab, abciximab, romosozumab, blosozumab, humanized anti-human A33 antibody, and bococizumab) induced responses in 1.9%, 3.8%, 6.4%, 10.0%, 29.2%, 43.8%, and 89.5% of donors, respectively. These data are comparable with ADA incidences in clinical settings. Our results show that this assay can contribute to the swift assessment and mechanistic understanding of the immunogenicity of therapeutic antibodies.
Collapse
Affiliation(s)
- Yoshiyuki Arata
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Shigeki Motoyama
- Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Mariko Yano
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Tatsuya Ikuno
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Shunsuke Ito
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Tomochika Matsushita
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Akira Takeiri
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Yukari Nishito
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
- Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Nami Yabuki
- Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Hideaki Mizuno
- Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Masayuki Mishima
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Masaki Honda
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Jumpei Kiyokawa
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Hiromi Suzuki
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Shuichi Chiba
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Mitsuyasu Tabo
- Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Chiyomi Kubo
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| |
Collapse
|
9
|
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: 2.0] [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
|
10
|
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: 0.5] [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
|
11
|
Ducret A, Ackaert C, Bessa J, Bunce C, Hickling T, Jawa V, Kroenke MA, Lamberth K, Manin A, Penny HL, Smith N, Terszowski G, Tourdot S, Spindeldreher S. Assay format diversity in pre-clinical immunogenicity risk assessment: Toward a possible harmonization of antigenicity assays. MAbs 2021; 14:1993522. [PMID: 34923896 PMCID: PMC8726688 DOI: 10.1080/19420862.2021.1993522] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
A major impediment to successful use of therapeutic protein drugs is their ability to induce anti-drug antibodies (ADA) that can alter treatment efficacy and safety in a significant number of patients. To this aim, in silico, in vitro, and in vivo tools have been developed to assess sequence and other liabilities contributing to ADA development at different stages of the immune response. However, variability exists between similar assays developed by different investigators due to the complexity of assays, a degree of uncertainty about the underlying science, and their intended use. The impact of protocol variations on the outcome of the assays, i.e., on the immunogenicity risk assigned to a given drug candidate, cannot always be precisely assessed. Here, the Non-Clinical Immunogenicity Risk Assessment working group of the European Immunogenicity Platform (EIP) reviews currently used assays and protocols and discusses feasibility and next steps toward harmonization and standardization.
Collapse
Affiliation(s)
- Axel Ducret
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Chloé Ackaert
- ImmunXperts SA (A Nexelis Group Company), Gosselies, Belgium
| | - Juliana Bessa
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Timothy Hickling
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Vibha Jawa
- Biotherapeutics and Bioanalysis Non-Clinical Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Mark A Kroenke
- Clinical Immunology-Translational Medicine, Amgen Inc, Thousand Oaks, CA, USA
| | - Kasper Lamberth
- Analysis & Characterisation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Anaïs Manin
- Abzena, Babraham Research Campus, Cambridge, UK
| | - Hweixian L Penny
- Clinical Immunology-Translational Medicine, Amgen Inc, Thousand Oaks, CA, USA
| | - Noel Smith
- Lonza Biologics, Chesterford Research Park, Saffron Walden, UK
| | - Grzegorz Terszowski
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | |
Collapse
|
12
|
In vitro immunogenicity prediction: bridging between innate and adaptive immunity. Bioanalysis 2021; 13:1071-1081. [PMID: 34124935 DOI: 10.4155/bio-2021-0077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Development of antidrug antibodies (ADAs) is an undesirable potential outcome of administration of biotherapeutics and involves the innate and adaptive immune systems. ADAs can have detrimental clinical consequences: they can reduce biotherapeutic efficacy or produce adverse events. Because animal models are considered poor predictors of immunogenicity in humans, in vitro assays with human innate and adaptive immune cells are commonly used alternatives that can reveal cell-mediated unwanted immune responses. Multiple methods have been developed to assess the immune cell response following exposure to biotherapeutics and estimate the potential immunogenicity of biotherapeutics. This review highlights the role of innate and adaptive immune cells as the drivers of immunogenicity and summarizes the use of these cells in assays to predict clinical ADA.
Collapse
|
13
|
Jambari NN, Liddell S, Martinez-Pomares L, Alcocer MJC. Effect of O-linked glycosylation on the antigenicity, cellular uptake and trafficking in dendritic cells of recombinant Ber e 1. PLoS One 2021; 16:e0249876. [PMID: 33914740 PMCID: PMC8084162 DOI: 10.1371/journal.pone.0249876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 03/29/2021] [Indexed: 11/18/2022] Open
Abstract
Ber e 1, a major Brazil nut allergen, has been successfully produced in the yeast Pichia pastoris expression system as homogenous recombinant Ber e 1 (rBer e 1) with similar physicochemical properties and identical immunoreactivity to its native counterpart, nBer e 1. However, O-linked glycans was detected on the P.pastoris-derived rBer e 1, which is not naturally present in nBer e 1, and may contribute to the allergic sensitisation. In this study, we addressed the glycosylation differences between P. pastoris-derived recombinant Ber e 1 and its native counterparts. We also determined whether this fungal glycosylation could affect the antigenicity and immunogenicity of the rBer e 1 by using dendritic cells (DC) as an immune cell model due to their role in modulating the immune response. We identified that the glycosylation occurs at Ser96, Ser101 and Ser110 on the large chain and Ser19 on the small polypeptide chain of rBer e 1 only. The glycosylation on rBer e 1 was shown to elicit varying degree of antigenicity by binding to different combination of human leukocyte antigens (HLA) at different frequencies compared to nBer e 1 when tested using human DC-T cell assay. However, both forms of Ber e 1 are weak immunogens based from their low response indexes (RI). Glycans present on rBer e 1 were shown to increase the efficiency of the protein recognition and internalization by murine bone marrow-derived dendritic cells (bmDC) via C-type lectin receptors, particularly the mannose receptor (MR), compared to the non-glycosylated nBer e 1 and SFA8, a weak allergenic 2S albumin protein from sunflower seed. Binding of glycosylated rBer e 1 to MR alone was found to not induce the production of IL-10 that modulates bmDC to polarise Th2 cell response by suppressing IL-12 production and DC maturation. Our findings suggest that the O-linked glycosylation by P. pastoris has a small but measurable effect on the in vitro antigenicity of the rBer e 1 compared to its non-glycosylated counterpart, nBer e 1, and thus may influence its applications in diagnostics and immunotherapy.
Collapse
Affiliation(s)
- Nuzul N. Jambari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Susan Liddell
- Division of Animal Science, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Luisa Martinez-Pomares
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| | - Marcos J. C. Alcocer
- Division of Food Sciences, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| |
Collapse
|
14
|
Cohen S, Myneni S, Batt A, Guerrero J, Brumm J, Chung S. Immunogenicity risk assessment for biotherapeutics through in vitro detection of CD134 and CD137 on T helper cells. MAbs 2021; 13:1898831. [PMID: 33729092 PMCID: PMC7993230 DOI: 10.1080/19420862.2021.1898831] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/11/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Biotherapeutics, which are biologic medications that are natural or bioengineered products of living cells, have revolutionized the treatment of many diseases. However, unwanted immune responses still present a major challenge to their widespread adoption. Many patients treated with biotherapeutics develop antigen-specific anti-drug antibodies (ADAs) that may reduce the efficacy of the therapy or cross-react with the endogenous counterpart of a protein therapeutic, or both. Here, we describe an in vitro method for assessing the immunogenic risk of a biotherapeutic. We found a correlation between clinical immunogenicity and the frequency with which a biotherapeutic stimulated an increase in CD134, CD137, or both cell surface markers on CD4+ T cells. Using high-throughput flow cytometry, we examined the effects of 14 biotherapeutics with diverse rates of clinical immunogenicity on peripheral blood mononuclear cells from 120 donors with diverse human leukocyte antigen class II-encoding alleles. Biotherapeutics with high rates of ADA development in the clinic had higher proportions of CD4+ T cells positive for CD134 or CD137 than biotherapeutics with low clinical immunogenicity. This method provides a rapid and simple preclinical test of the immunogenic potential of a new candidate biotherapeutic or biosimilar. Implementation of this approach during biotherapeutic research and development enables rapid elimination of candidates that are likely to cause ADA-related adverse events and detrimental consequences.
Collapse
Affiliation(s)
- Sivan Cohen
- Department of BioAnalytical Sciences, Genentech Inc, South San Francisco, CA, USA
| | - Srividya Myneni
- Department of BioAnalytical Sciences, Genentech Inc, South San Francisco, CA, USA
| | - Anna Batt
- Department of BioAnalytical Sciences, Genentech Inc, South San Francisco, CA, USA
| | - Joyce Guerrero
- Department of BioAnalytical Sciences, Genentech Inc, South San Francisco, CA, USA
| | - Jochen Brumm
- Department of Biostatistics, Genentech Inc, South San Francisco, CA, USA
| | - Shan Chung
- Department of BioAnalytical Sciences, Genentech Inc, South San Francisco, CA, USA
| |
Collapse
|
15
|
Di Blasi D, Claessen I, Turksma AW, van Beek J, Ten Brinke A. Guidelines for analysis of low-frequency antigen-specific T cell results: Dye-based proliferation assay vs 3H-thymidine incorporation. J Immunol Methods 2020; 487:112907. [PMID: 33152332 DOI: 10.1016/j.jim.2020.112907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
It is generally recognized that dysregulation of the immune system plays a critical role in many diseases, including autoimmune diseases and cancer. T cells play a crucial role in maintaining self-tolerance, while loss of immune tolerance and T cell activation can lead to severe inflammation and tissue damage. T cell responses have a key role in the effectiveness of vaccination strategies and immunomodulating therapies. Immunomonitoring methods have the ability to elucidate immunological processes, monitor the development of disease and assess therapeutic effects. In this respect, it is of particular interest to evaluate antigen (Ag)-specific T cells by determining their frequency, type and functionality in cellular assays. Nevertheless, Ag-specific T cells are detected infrequently in most diseases using current techniques. Many efforts have been made to develop more sensitive, reproducible, and reliable methods for Ag-specific T cell detection. It has been found that analysis of cellular proliferation can be a useful tool to determine the presence and frequency of Ag-specific T cell and to provides insight into modulation of the T cell response by a specific antigen or therapy. However, the selection of a cut-off value for a positive response and therefore a more accurate interpretation of the data, continues to be a major concern. Here, we provide guidelines to select a proper cut-off for monitoring of Ag-specific CD4+ T cell responses. In vitro Ag-stimulation has been assessed with two methods; a dye-based proliferation assay and 3H-thymidine-based assay. Two cut-off approaches are compared; mean and variance of control wells, and the stimulation index. By evaluating the proliferative response to the in vitro Ag-stimulation using these two methods, we demonstrate the importance of taking into consideration the variability of the control wells to distinguish a positive from a false positive response.
Collapse
Affiliation(s)
- Daniela Di Blasi
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands.
| | - Iris Claessen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands; Sanquin Diagnostics B.V., Amsterdam, the Netherlands
| | | | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, the Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Meunier S, de Bourayne M, Hamze M, Azam A, Correia E, Menier C, Maillère B. Specificity of the T Cell Response to Protein Biopharmaceuticals. Front Immunol 2020; 11:1550. [PMID: 32793213 PMCID: PMC7387651 DOI: 10.3389/fimmu.2020.01550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/12/2020] [Indexed: 12/17/2022] Open
Abstract
The anti-drug antibody (ADA) response is an undesired humoral response raised against protein biopharmaceuticals (BPs) which can dramatically disturb their therapeutic properties. One particularity of the ADA response resides in the nature of the immunogens, which are usually human(ized) proteins and are therefore expected to be tolerated. CD4 T cells initiate, maintain and regulate the ADA response and are therefore key players of this immune response. Over the last decade, advances have been made in characterizing the T cell responses developed by patients treated with BPs. Epitope specificity and phenotypes of BP-specific T cells have been reported and highlight the effector and regulatory roles of T cells in the ADA response. BP-specific T cell responses are assessed in healthy subjects to anticipate the immunogenicity of BP prior to their testing in clinical trials. Immunogenicity prediction, also called preclinical immunogenicity assessment, aims at identifying immunogenic BPs and immunogenic BP sequences before any BP injection in humans. All of the approaches that have been developed to date rely on the detection of BP-specific T cells in donors who have never been exposed to BPs. The number of BP-specific T cells circulating in the blood of these donors is therefore limited. T cell assays using cells collected from healthy donors might reveal the weak tolerance induced by BPs, whose endogenous form is expressed at a low level. These BPs have a complete human sequence, but the level of their endogenous form appears insufficient to promote the negative selection of autoreactive T cell clones. Multiple T cell epitopes have also been identified in therapeutic antibodies and some other BPs. The pattern of identified T cell epitopes differs across the antibodies, notwithstanding their humanized, human or chimeric nature. However, in all antibodies, the non-germline amino acid sequences mainly found in the CDRs appear to be the main driver of immunogenicity, provided they can be presented by HLA class II molecules. Considering the fact that the BP field is expanding to include new formats and gene and cell therapies, we face new challenges in understanding and mastering the immunogenicity of new biological products.
Collapse
Affiliation(s)
- Sylvain Meunier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Marie de Bourayne
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Moustafa Hamze
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Aurélien Azam
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Evelyne Correia
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Catherine Menier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Bernard Maillère
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| |
Collapse
|
17
|
Jawa V, Terry F, Gokemeijer J, Mitra-Kaushik S, Roberts BJ, Tourdot S, De Groot AS. T-Cell Dependent Immunogenicity of Protein Therapeutics Pre-clinical Assessment and Mitigation-Updated Consensus and Review 2020. Front Immunol 2020; 11:1301. [PMID: 32695107 PMCID: PMC7338774 DOI: 10.3389/fimmu.2020.01301] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023] Open
Abstract
Immune responses to protein and peptide drugs can alter or reduce their efficacy and may be associated with adverse effects. While anti-drug antibodies (ADA) are a standard clinical measure of protein therapeutic immunogenicity, T cell epitopes in the primary sequences of these drugs are the key drivers or modulators of ADA response, depending on the type of T cell response that is stimulated (e.g., T helper or Regulatory T cells, respectively). In a previous publication on T cell-dependent immunogenicity of biotherapeutics, we addressed mitigation efforts such as identifying and reducing the presence of T cell epitopes or T cell response to protein therapeutics prior to further development of the protein therapeutic for clinical use. Over the past 5 years, greater insight into the role of regulatory T cell epitopes and the conservation of T cell epitopes with self (beyond germline) has improved the preclinical assessment of immunogenic potential. In addition, impurities contained in therapeutic drug formulations such as host cell proteins have also attracted attention and become the focus of novel risk assessment methods. Target effects have come into focus, given the emergence of protein and peptide drugs that target immune receptors in immuno-oncology applications. Lastly, new modalities are entering the clinic, leading to the need to revise certain aspects of the preclinical immunogenicity assessment pathway. In addition to drugs that have multiple antibody-derived domains or non-antibody scaffolds, therapeutic drugs may now be introduced via viral vectors, cell-based constructs, or nucleic acid based therapeutics that may, in addition to delivering drug, also prime the immune system, driving immune response to the delivery vehicle as well as the encoded therapeutic, adding to the complexity of assessing immunogenicity risk. While it is challenging to keep pace with emerging methods for the preclinical assessment of protein therapeutics and new biologic therapeutic modalities, this collective compendium provides a guide to current best practices and new concepts in the field.
Collapse
Affiliation(s)
- Vibha Jawa
- Predictive and Clinical Immunogenicity, PPDM, Merck & Co., Kenilworth, NJ, United States
| | | | - Jochem Gokemeijer
- Discovery Biotherapeutics, Bristol-Myers Squibb, Cambridge, MA, United States
| | | | | | - Sophie Tourdot
- BioMedicine Design, Pfizer Inc., Andover, MA, United States
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|
18
|
Walsh RE, Lannan M, Wen Y, Wang X, Moreland CA, Willency J, Knierman MD, Spindler L, Liu L, Zeng W, Rocha GV, Obungu V, Lu J, Kaliyaperumal A, Ferrante A, Siegel R, Malherbe LP. Post-hoc assessment of the immunogenicity of three antibodies reveals distinct immune stimulatory mechanisms. MAbs 2020; 12:1764829. [PMID: 32370596 PMCID: PMC8648324 DOI: 10.1080/19420862.2020.1764829] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Robin E. Walsh
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Megan Lannan
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Yi Wen
- Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA, USA
| | - Xiaoli Wang
- Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA, USA
| | | | - Jill Willency
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Michael D. Knierman
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Laura Spindler
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Ling Liu
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Wei Zeng
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Guilherme V. Rocha
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Victor Obungu
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Jirong Lu
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Arunan Kaliyaperumal
- Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA, USA
| | - Andrea Ferrante
- Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA, USA
| | - Robert Siegel
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Laurent P. Malherbe
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
19
|
Nabhan M, Pallardy M, Turbica I. Immunogenicity of Bioproducts: Cellular Models to Evaluate the Impact of Therapeutic Antibody Aggregates. Front Immunol 2020; 11:725. [PMID: 32431697 PMCID: PMC7214678 DOI: 10.3389/fimmu.2020.00725] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022] Open
Abstract
Patients treated with bioproducts (BPs) frequently develop anti-drug antibodies (ADAs) with potential neutralizing capacities leading to loss of clinical response or potential hypersensitivity reactions. Many factors can influence BP immunogenicity and could be related to the patient, the treatment, as well as to the product itself. Among these latter factors, it is now well accepted that BP aggregation is associated with an increased potential for immunogenicity, as aggregates seem to be correlated with ADA development. Moreover, the presence of high-affinity ADAs suggests a CD4 T-cell dependent adaptive immune response and therefore a pivotal role for antigen-presenting cells (APCs), such as dendritic cells (DCs). In this review, we address the in vitro methods developed to evaluate how monoclonal antibodies could trigger the immunization process by focusing on the role of aggregated antibodies in the establishment of this response. In particular, we will present the different cell-based assays that have been used to assess the potential of antibodies and their aggregates to modulate cellular mechanisms leading to activation and the biological parameters (cellular activation markers, proliferation and secreted molecules) that can be measured to evaluate the different cell activation stages and their consequences in the propagation of the immune response. Indeed, the use of such strategies could help evaluate the risk of BP immunogenicity and their role in mitigating this risk.
Collapse
Affiliation(s)
- Myriam Nabhan
- Inserm, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Marc Pallardy
- Inserm, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| | - Isabelle Turbica
- Inserm, Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
20
|
Ito S, Ikuno T, Mishima M, Yano M, Hara T, Kuramochi T, Sampei Z, Wakabayashi T, Tabo M, Chiba S, Kubo C. In vitro human helper T-cell assay to screen antibody drug candidates for immunogenicity. J Immunotoxicol 2020; 16:125-132. [PMID: 31179789 DOI: 10.1080/1547691x.2019.1604586] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibody (mAb) drugs offer a number of valuable treatments. Many newly developed mAb drugs include artificial modification of amino acid sequences from human origin, which may cause higher immunogenicity to induce anti-drug antibodies (ADA). If the immunogenicity of a new candidate can be understood in the nonclinical phase, clinical studies will be safer and the success rate of development improved. Empirically, in vitro immunogenicity assays with human cells have proved to be sufficiently sensitive to nonhuman proteins, but not to human/humanized mAb. To detect the weaker immunogenicity of human-based mAb, a more sensitive biomarker for in vitro assays is needed. The in vitro study here developed a proliferation assay (TH cell assay) using flow cytometry analysis that can detect a slight increase in proliferating TH cells. Samples from 218 donors treated with a low-immunogenic drug (etanercept) were measured to determine a positive threshold level. With this threshold, positive donor percentages among PBMC after treatment with higher-immunogenicity mAb drugs were noted, that is, 39.5% with humanized anti-human A33 antibody (hA33), 27.3% with abciximab, 25.9% with adalimumab, and 14.8% with infliximab. Biotherapeutics with low immunogenicity yielded values of 0% for basiliximab and 3.7% for etanercept. These data showed a good comparability with previously reported incidences of clinical ADA with the evaluated drugs. Calculations based on the data here showed that a TH cell assay with 40 donors could provide statistically significant differences when comparing low- (etanercept) versus highly immunogenic mAb (except for infliximab). Based on the outcomes here, for screening purposes, a practical cutoff point of 3/20 positives with 20 donors was proposed to alert immunogenicity of mAb drug candidates.
Collapse
Affiliation(s)
- Shunsuke Ito
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | - Tatsuya Ikuno
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | - Masayuki Mishima
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | - Mariko Yano
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | - Toshiko Hara
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | | | - Zenjiro Sampei
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | | | - Mitsuyasu Tabo
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | - Shuichi Chiba
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| | - Chiyomi Kubo
- a Research Division , Chugai Pharmaceutical Co., Ltd. Shizuoka , Japan
| |
Collapse
|
21
|
Stickler M, Reddy A, Xiong JM, Wong MH, Akamatsu Y, Hinton PR, Harding FA. Design, creation and in vitro testing of a reduced immunogenicity humanized anti-CD25 monoclonal antibody that retains functional activity. Protein Eng Des Sel 2019; 32:543-554. [PMID: 32725169 DOI: 10.1093/protein/gzaa017] [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: 04/04/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 11/14/2022] Open
Abstract
Humanized and fully human sequence-derived therapeutic antibodies retain the capacity to induce anti-drug antibodies. Daclizumab (humanized version of the murine anti-Tac antibody; E.HAT) was selected for a proof of concept application of engineering approaches to reduce potential immunogenicity due to its demonstrated immunogenicity in the clinic. Reduced immunogenicity variants of E.HAT were created by identifying and modifying a CD4+ T cell epitope region in the VH region. Variant epitope region peptides were selected for their reduced capacity to induce CD4+ T cell proliferative responses in vitro. Variant antibody molecules were created, and CD25 affinity and potency were similar to the unmodified parent antibody. Fab fragments from the variant antibodies induced a lower frequency and magnitude of responses in human peripheral blood mononuclear cells proliferation tests. By the empirical selection of two amino acid mutations, fully functional humanized E.HAT antibodies with reduced potential to induce immune responses in vitro were created.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul R Hinton
- Formerly of AbbVie, Redwood City, CA, USA.,IGM Biosciences, Mountain View, CA, USA
| | | |
Collapse
|
22
|
Tapia-Calle G, Born PA, Koutsoumpli G, Gonzalez-Rodriguez MI, Hinrichs WLJ, Huckriede ALW. A PBMC-Based System to Assess Human T Cell Responses to Influenza Vaccine Candidates In Vitro. Vaccines (Basel) 2019; 7:vaccines7040181. [PMID: 31766202 PMCID: PMC6963913 DOI: 10.3390/vaccines7040181] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 01/08/2023] Open
Abstract
Vaccine development is an expensive and time-consuming process that heavily relies on animal models. Yet, vaccine candidates that have previously succeeded in animal experiments often fail in clinical trials questioning the predictive value of animal models. Alternative assay systems that can add to the screening and evaluation of functional characteristics of vaccines in a human context before embarking on costly clinical trials are therefore urgently needed. In this study, we have established an in vitro system consisting of long-term cultures of unfractionated peripheral blood mononuclear cells (PBMCs) from healthy volunteers to assess (recall) T cell responses to vaccine candidates. We observed that different types of influenza vaccines (whole inactivated virus (WIV), split, and peptide vaccines) were all able to stimulate CD4 and CD8 T cell responses but to different extents in line with their reported in vivo properties. In-depth analyses of different T cell subsets revealed that the tested vaccines evoked mainly recall responses as indicated by the fact that the vast majority of the responding T cells had a memory phenotype. Furthermore, we observed vaccine-induced activation of T follicular helper cells, which are associated with the induction of humoral immune responses. Our results demonstrate the suitability of the established PBMC-based system for the in vitro evaluation of memory T cell responses to vaccines and the comparison of vaccine candidates in a human immune cell context. As such, it can help to bridge the gap between animal experiments and clinical trials and assist in the selection of promising vaccine candidates, at least for recall antigens.
Collapse
Affiliation(s)
- Gabriela Tapia-Calle
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, 9713AV Groningen, The Netherlands
| | - Philip A Born
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| | - Georgia Koutsoumpli
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, 9713AV Groningen, The Netherlands
| | - Martin Ignacio Gonzalez-Rodriguez
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, 9713AV Groningen, The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| | - Anke L W Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, 9713AV Groningen, The Netherlands
| |
Collapse
|
23
|
Current In Vitro Assays for Prediction of T Cell Mediated Immunogenicity of Biotherapeutics and Manufacturing Impurities. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09412-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
24
|
Getter T, Margalit R, Kahremany S, Levy L, Blum E, Khazanov N, Keshet-Levy NY, Tamir TY, Ben Major M, Lahav R, Zilber S, Senderowitz H, Bradfield P, Imhof BA, Alpert E, Gruzman A. Novel inhibitors of leukocyte transendothelial migration. Bioorg Chem 2019; 92:103250. [PMID: 31580982 DOI: 10.1016/j.bioorg.2019.103250] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023]
Abstract
Leukocyte transendothelial migration is one of the most important step in launching an inflammatory immune response and chronic inflammation can lead to devastating diseases. Leukocyte migration inhibitors are considered as promising and potentially effective therapeutic agents to treat inflammatory and auto-immune disorders. In this study, based on previous trioxotetrahydropyrimidin based integrin inhibitors that suboptimally blocked leukocyte adhesion, twelve molecules with a modified scaffold were designed, synthesized, and tested in vitro for their capacity to block the transendothelial migration of immune cells. One of the molecules, namely, methyl 4-((2-(tert-butyl)-6-((2,4,6-trioxotetrahydropyrimidin-5(2H)-ylidene) methyl) phenoxy) methyl) benzoate, (compound 12), completely blocked leukocyte transendothelial migration, without any toxic effects on immune or endothelial cells (IC50 = 2.4 µM). In vivo, compound 12 exhibited significant therapeutic effects in inflammatory bowel disease (IBD)/Crohn's disease, multiple sclerosis, fatty liver disease, and rheumatoid arthritis models. A detailed acute and chronic toxicity profile of the lead compound in vivo did not reveal any toxic effects. Such a type of molecule might therefore provide a unique starting point for designing a novel class of leukocyte transmigration blocking agents with broad therapeutic applications in inflammatory and auto-immune pathologies.
Collapse
Affiliation(s)
- Tamar Getter
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Raanan Margalit
- "Science in Action", Ness-Ziona, Israel; "AltA-ZuZ Therapeutics", Ness-Ziona, Israel
| | - Shirin Kahremany
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Laura Levy
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Eliav Blum
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Netaly Khazanov
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Nimrod Y Keshet-Levy
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel; Department of Pathology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Tigist Y Tamir
- Department of Pharmacology and the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - M Ben Major
- Department of Pharmacology and the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ron Lahav
- "AltA-ZuZ Therapeutics", Ness-Ziona, Israel
| | - Sofia Zilber
- Department of Pathology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Hanoch Senderowitz
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Beat A Imhof
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Arie Gruzman
- Division of Medicinal Chemistry, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
25
|
Yogurtcu ON, Sauna ZE, McGill JR, Tegenge MA, Yang H. TCPro: an In Silico Risk Assessment Tool for Biotherapeutic Protein Immunogenicity. AAPS JOURNAL 2019; 21:96. [PMID: 31376048 DOI: 10.1208/s12248-019-0368-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022]
Abstract
Most immune responses to biotherapeutic proteins involve the development of anti-drug antibodies (ADAs). New drugs must undergo immunogenicity assessments to identify potential risks at early stages in the drug development process. This immune response is T cell-dependent. Ex vivo assays that monitor T cell proliferation often are used to assess immunogenicity risk. Such assays can be expensive and time-consuming to carry out. Furthermore, T cell proliferation requires presentation of the immunogenic epitope by major histocompatibility complex class II (MHCII) proteins on antigen-presenting cells. The MHC proteins are the most diverse in the human genome. Thus, obtaining cells from subjects that reflect the distribution of the different MHCII proteins in the human population can be challenging. The allelic frequencies of MHCII proteins differ among subpopulations, and understanding the potential immunogenicity risks would thus require generation of datasets for specific subpopulations involving complex subject recruitment. We developed TCPro, a computational tool that predicts the temporal dynamics of T cell counts in common ex vivo assays for drug immunogenicity. Using TCPro, we can test virtual pools of subjects based on MHCII frequencies and estimate immunogenicity risks for different populations. It also provides rapid and inexpensive initial screens for new biotherapeutics and can be used to determine the potential immunogenicity risk of new sequences introduced while bioengineering proteins. We validated TCPro using an experimental immunogenicity dataset, making predictions on the population-based immunogenicity risk of 15 protein-based biotherapeutics. Immunogenicity rankings generated using TCPro are consistent with the reported clinical experience with these therapeutics.
Collapse
Affiliation(s)
- Osman N Yogurtcu
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, 10903 New Hampshire Ave, Silver Spring, 20993, Maryland, USA
| | - Zuben E Sauna
- Office of Tissues and Advanced Therapy, Center for Biologics Evaluation and Research, US FDA, 10903 New Hampshire Ave, Silver Spring, 20993, Maryland, USA
| | - Joseph R McGill
- Office of Tissues and Advanced Therapy, Center for Biologics Evaluation and Research, US FDA, 10903 New Hampshire Ave, Silver Spring, 20993, Maryland, USA
| | - Million A Tegenge
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, 10903 New Hampshire Ave, Silver Spring, 20993, Maryland, USA
| | - Hong Yang
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, 10903 New Hampshire Ave, Silver Spring, 20993, Maryland, USA.
| |
Collapse
|
26
|
Abstract
Therapeutic protein drugs have significantly improved the management of many severe and chronic diseases. However, their development and optimal clinical application are complicated by the induction of unwanted immune responses. Therapeutic protein-induced antidrug antibodies can alter drug pharmacokinetics and pharmacodynamics leading to impaired efficacy and occasionally serious safety issues. There has been a growing interest over the past decade in developing methods to assess the risk of unwanted immunogenicity during preclinical drug development, with the aim to mitigate the risk during the molecular design phase, clinical development and when products reach the market. Here, we discuss approaches to therapeutic protein immunogenicity risk assessment, with attention to assays and in vivo models used to mitigate this risk.
Collapse
|
27
|
Sauna ZE, Lagassé D, Pedras-Vasconcelos J, Golding B, Rosenberg AS. Evaluating and Mitigating the Immunogenicity of Therapeutic Proteins. Trends Biotechnol 2018; 36:1068-1084. [DOI: 10.1016/j.tibtech.2018.05.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/19/2022]
|
28
|
Groell F, Jordan O, Borchard G. In vitro models for immunogenicity prediction of therapeutic proteins. Eur J Pharm Biopharm 2018; 130:128-142. [DOI: 10.1016/j.ejpb.2018.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/09/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
|
29
|
Gokemeijer J, Jawa V, Mitra-Kaushik S. How Close Are We to Profiling Immunogenicity Risk Using In Silico Algorithms and In Vitro Methods?: an Industry Perspective. AAPS JOURNAL 2017; 19:1587-1592. [PMID: 28971356 DOI: 10.1208/s12248-017-0143-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/30/2017] [Indexed: 12/22/2022]
Abstract
In silico HLA-binding algorithms and in vitro T cell-based assays as predictive tools for human immunogenicity risk have made inroads in the biotherapeutic drug discovery and development process. Currently, these tools are being used only for candidate selection or characterization and not for making a go/no-go decision for further development. A clear limitation for a broader implementation is the lack of correlation between the predicted T cell epitope content/immune reactivity potential of a biotherapeutic and the subsequent ADA-related clinical immunogenicity outcome. The current state of technologies and their pros and cons were discussed as a part of the 2016 AAPS National Biotechnology Conference in a themed session. A review of the advances in the area and the session talks along with the ensuing discussions are summarized in this commentary.
Collapse
Affiliation(s)
- Jochem Gokemeijer
- Discovery Chemistry and Molecular Technologies, Bristol-Myers Squibb, Waltham, Massachusetts, USA
| | - Vibha Jawa
- Biologics and Vaccine Formulations, Merck Sharp & Dohme Corp, 2000 Galloping Hill Road, K-15 H406, Kenilworth, New Jersey, 07033, USA
| | - Shibani Mitra-Kaushik
- Biomarker and Clinical Bioanalysis, Sanofi, 1 The Mountain Road, Framingham, Massachusetts, 01701, USA.
| |
Collapse
|
30
|
Rosenberg AS, Sauna ZE. Immunogenicity assessment during the development of protein therapeutics. J Pharm Pharmacol 2017; 70:584-594. [DOI: 10.1111/jphp.12810] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/26/2017] [Indexed: 12/29/2022]
Abstract
Abstract
Objective
Here we provide a critical review of the state of the art with respect to non-clinical assessments of immunogenicity for therapeutic proteins.
Key findings
The number of studies on immunogenicity published annually has more than doubled in the last 5 years. The science and technology, which have reached a critical mass, provide multiple of non-clinical approaches (computational, in vitro, ex vivo and animal models) to first predict and then to modify or eliminate T-cell or B-cell epitopes via de-immunization strategies. We discuss how these may be used in the context of drug development in assigning the immunogenicity risk of new and marketed therapeutic proteins.
Summary
Protein therapeutics represents a large share of the pharma market and provide medical interventions for some of the most complex and intractable diseases. Immunogenicity (the development of antibodies to therapeutic proteins) is an important concern for both the safety and efficacy of protein therapeutics as immune responses may neutralize the activity of life-saving and highly effective protein therapeutics and induce hypersensitivity responses including anaphylaxis. The non-clinical computational tools and experimental technologies that offer a comprehensive and increasingly accurate estimation of immunogenic potential are surveyed here. This critical review also discusses technologies which are promising but are not as yet ready for routine use.
Collapse
Affiliation(s)
- Amy S Rosenberg
- Laboratory of Immunology, Division of Biotechnology Product Review and Research 3, Office of Biotechnology Products, Center for Drugs Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Zuben E Sauna
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapeutics, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|