1
|
Jeong YH, Lennon G, Veldman G, Serna DM, Ibrahimov A. Establishing endotoxin limits to enhance the reliability of in vitro immunogenicity risk assessments. MAbs 2025; 17:2458627. [PMID: 39893505 PMCID: PMC11792839 DOI: 10.1080/19420862.2025.2458627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
Immunogenic responses to biotherapeutics often lead to termination of their development because the resulting anti-drug-antibodies (ADA) can negatively impact pharmacology, safety, and efficacy. To mitigate ADA risks, in vitro risk assessment assays in non-clinical settings are essential to enhance safety and efficacy of protein-based therapeutics. This study aimed to develop and validate a human in vitro immunogenicity T cell proliferation assay. However, there is a lack of comprehensive guidelines for managing product-related factors such as endotoxin contamination, which can significantly influence assay sensitivity and accuracy. Our investigation of the impact of endotoxins revealed that levels above 0.1 EU/mg significantly induce T cell proliferation and CD14+ myeloid cell expansion, leading to potential false-positive outcomes in immunogenicity assessments. These findings suggest the importance of developing standardized protocols to enhance the predictive capability of in vitro methods, ensuring the assessment of therapeutic proteins accurately reflects their immunogenic potential without interference from contaminants.
Collapse
Affiliation(s)
- Yun Hee Jeong
- In Vitro Immunosafety, Development Biological Sciences, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Gillian Lennon
- In Vitro Immunosafety, Development Biological Sciences, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Geertruida Veldman
- Biotherapeutics Discovery Research, AbbVie Bioresearch Center, AbbVie Bioresearch Center, Worcester, MA, USA
| | - Daniel M. Serna
- In Vitro Immunosafety, Development Biological Sciences, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Alexander Ibrahimov
- In Vitro Immunosafety, Development Biological Sciences, Abbvie Bioresearch Center, Worcester, MA, USA
| |
Collapse
|
2
|
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
|
3
|
Liu W, Wang X, Zhao S, Yang S, Zheng X, Gong F, Pei L, Xu D, Li R, Yang Z, Mao E, Chen E, Chen Y. Unraveling the immunological landscape in acute pancreatitis progression to sepsis: insights from a Mendelian randomization study on immune cell traits. Front Immunol 2024; 15:1374787. [PMID: 38601150 PMCID: PMC11004341 DOI: 10.3389/fimmu.2024.1374787] [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: 01/22/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is a severe digestive system disorder with a significant risk of progressing to sepsis, a major cause of mortality. Unraveling the immunological pathways in AP is essential for developing effective treatments, particularly understanding the role of specific immune cell traits in this progression. METHODS Employing a bidirectional two-sample Mendelian Randomization (MR) approach, this study first examined the causal relationship between AP and 731 immune cell traits to identify those significantly associated with AP. Subsequently, we explored the causal associations between 731 immune cell traits and sepsis. The analysis utilized extensive genome-wide association studies (GWAS) summary datasets, with a focus on identifying common immune cell traits with statistically significant causal associations between AP and sepsis. RESULTS Our investigation identified 44 immune cell traits unidirectionally associated with AP and 36 traits unidirectionally associated with sepsis. Among these, CD127 on CD28+ CD45RA- CD8+ T cells emerged as a common mediator, accounting for 5.296% of the increased risk of sepsis in AP patients. This finding highlights the significant role of specific memory CD8+ T cells in the pathophysiology of AP and its progression to sepsis. CONCLUSION This study elucidates the critical role of specific immune cell traits, particularly CD127hi memory CD8+ T cells, in the progression of AP to sepsis. Our findings provide a foundation for future research into targeted immune-modulatory therapies, potentially improving patient outcomes in AP-related sepsis and offering new insights into the complex immunological dynamics of this condition.
Collapse
Affiliation(s)
- Wenbin Liu
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofeng Wang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanzhi Zhao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Yang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangtao Zheng
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangchen Gong
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Pei
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Xu
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhitao Yang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Noe P, Wang JH, Chung K, Cheng Z, Field JJ, Shen X, Cortesio CL, Pastuskovas CV, Phee H, Tarbell KV, Egen JG, Casbon AJ. Therapeutically targeting type I interferon directly to XCR1+ dendritic cells reveals the role of cDC1s in anti-drug antibodies. Front Immunol 2023; 14:1272055. [PMID: 37942313 PMCID: PMC10628189 DOI: 10.3389/fimmu.2023.1272055] [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: 08/03/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Conventional type 1 dendritic cells (cDC1s) are superior in antigen cross-presentation and priming CD8+ T cell anti-tumor immunity and thus, are a target of high interest for cancer immunotherapy. Type I interferon (IFN) is a potent inducer of antigen cross-presentation, but, unfortunately, shows only modest results in the clinic given the short half-life and high toxicity of current type I IFN therapies, which limit IFN exposure in the tumor. CD8+ T cell immunity is dependent on IFN signaling in cDC1s and preclinical studies suggest targeting IFN directly to cDC1s may be sufficient to drive anti-tumor immunity. Here, we engineered an anti-XCR1 antibody (Ab) and IFN mutein (IFNmut) fusion protein (XCR1Ab-IFNmut) to determine whether systemic delivery could drive selective and sustained type I IFN signaling in cDC1s leading to anti-tumor activity and, in parallel, reduced systemic toxicity. We found that the XCR1Ab-IFNmut fusion specifically enhanced cDC1 activation in the tumor and spleen compared to an untargeted control IFN. However, multiple treatments with the XCR1Ab-IFNmut fusion resulted in robust anti-drug antibodies (ADA) and loss of drug exposure. Using other cDC1-targeting Ab-IFNmut fusions, we found that localizing IFN directly to cDC1s activates their ability to promote ADA responses, regardless of the cDC1 targeting antigen. The development of ADA remains a major hurdle in immunotherapy drug development and the cellular and molecular mechanisms governing the development of ADA responses in humans is not well understood. Our results reveal a role of cDC1s in ADA generation and highlight the potential ADA challenges with targeting immunostimulatory agents to this cellular compartment.
Collapse
Affiliation(s)
- Paul Noe
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Joy H. Wang
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Kyu Chung
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Zhiyong Cheng
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Jessica J. Field
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, United States
| | - Xiaomeng Shen
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, United States
| | - Christa L. Cortesio
- Therapeutics Discovery, Amgen Research, South San Francisco, CA, United States
| | - Cinthia V. Pastuskovas
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, United States
| | - Hyewon Phee
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Kristin V. Tarbell
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Jackson G. Egen
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| | - Amy-Jo Casbon
- Oncology Research, Amgen Research, South San Francisco, CA, United States
| |
Collapse
|
5
|
Tolerogenic nanoparticles mitigate the formation of anti-drug antibodies against pegylated uricase in patients with hyperuricemia. Nat Commun 2022; 13:272. [PMID: 35022448 PMCID: PMC8755849 DOI: 10.1038/s41467-021-27945-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022] Open
Abstract
Biologic drugs have transformed the standard of care for many diseases. However, many biologics induce the formation of anti-drug antibodies (ADAs), which can compromise their safety and efficacy. Preclinical studies demonstrate that biodegradable nanoparticles-encapsulating rapamycin (ImmTOR), but not free rapamycin, mitigate the immunogenicity of co-administered biologic drugs. Here we report the outcomes from two clinical trials for ImmTOR. In the first ascending dose, open-label study (NCT02464605), pegadricase, an immunogenic, pegylated uricase enzyme derived from Candida utilis, is assessed for safety and tolerability (primary endpoint) as well as activity and immunogenicity (secondary endpoint); in the second single ascending dose Phase 1b trial (NCT02648269) composed of both a double-blind and open-label parts, we evaluate the safety of ImmTOR (primary endpoint) and its ability to prevent the formation of anti-drug antibodies against pegadricase and enhance its pharmacodynamic activity (secondary endpoint) in patients with hyperuricemia. The combination of ImmTOR and pegadricase is well tolerated. ImmTOR inhibits the development of uricase-specific ADAs in a dose-dependent manner, thus enabling sustained enzyme activity and reduction in serum uric acid levels. ImmTOR may thus represent a feasible approach for preventing the formation of ADAs to a broad range of immunogenic biologic therapies.
Collapse
|
6
|
Wen Y, Wang X, Cahya S, Anderson P, Velasquez C, Torres C, Ferrante A, Kaliyaperumal A. Comparability study of monocyte derived dendritic cells, primary monocytes, and THP1 cells for innate immune responses. J Immunol Methods 2021; 498:113147. [PMID: 34508774 DOI: 10.1016/j.jim.2021.113147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/06/2021] [Indexed: 01/23/2023]
Abstract
Immunogenicity is one major challenge to the successful development of biotherapeutics because it could adversely affect PK/PD, safety, and efficacy. Preclinical immunogenicity risk assessment strategies and assays have been developed and implemented to screen and optimize discovery molecules. Internalization by antigen presenting cells (APC) and innate immune activation are initial prerequisite steps in eliciting immune responses to biotherapeutics. Dendritic cells (DC)- and monocyte-based assays are employed to interrogate such risks, and their value has been well documented in the literature. However, these assays have limited throughput, exhibit higher variability, and entail lengthy and complex procedures as they are based on primary cells such as peripheral blood mononuclear cells (PBMC) from individual donors. Herein, we investigated THP1 cells as surrogate cells to study APC internalization and innate immune activation. Comparability studies showed that THP1 cells could resemble innate immune responses of monocyte-derived DC and primary CD14+ monocytes using a panel of therapeutic antibodies. In addition, an automated high throughput THP1 internalization assay was qualified to enable risk assessment at pre‑lead stages. The results demonstrated that THP1 cells can be utilized to assess immunogenicity risk in a high throughput manner.
Collapse
Affiliation(s)
- Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Xiaoli Wang
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Suntara Cahya
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Paul Anderson
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Candyd Velasquez
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Carina Torres
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Andrea Ferrante
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | | |
Collapse
|