1
|
Zhou J, Lei B, Shi F, Luo X, Wu K, Xu Y, Zhang Y, Liu R, Wang H, Zhou J, He X. CAR T-cell therapy for systemic lupus erythematosus: current status and future perspectives. Front Immunol 2024; 15:1476859. [PMID: 39749335 PMCID: PMC11694027 DOI: 10.3389/fimmu.2024.1476859] [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: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Systemic lupus erythematosus (SLE) and lupus nephritis (LN) are debilitating autoimmune disorders characterized by pathological autoantibodies production and immune dysfunction, causing chronic inflammation and multi-organ damage. Despite current treatments with antimalarial drugs, glucocorticoids, immunosuppressants, and monoclonal antibodies, a definitive cure remains elusive, highlighting an urgent need for novel therapeutic strategies. Recent studies indicate that chimeric antigen receptor T-cell (CAR-T) therapy has shown promising results in treating B-cell malignancies and may offer a significant breakthrough for non-malignant conditions like SLE. In this paper, we aim to provide an in-depth analysis of the advancements in CAR-T therapy for SLE, focusing on its potential to revolutionize treatment for this complex disease. We explore the fundamental mechanisms of CAR-T cell action, the rationale for its application in SLE, and the immunological underpinnings of the disease. We also summarize clinical data on the safety and efficacy of anti-CD19 and anti-B cell maturation antigen (BCMA) CAR-T cells in targeting B-cells in SLE. We discuss the clinical implications of these findings and the potential for CAR-T therapy to improve outcomes in severe or refractory SLE cases. The integration of CAR-T therapy into the SLE treatment paradigm presents a new horizon in autoimmunity research and clinical practice. This review underscores the need for continued exploration and optimization of CAR-T strategies to address the unmet needs of SLE patients.
Collapse
Affiliation(s)
- Jincai Zhou
- Innovation & Research Department, OriCell Therapeutics Co. Ltd., Shanghai, China
| | | | | | | | | | | | | | | | | | - Joy Zhou
- Innovation & Research Department, OriCell Therapeutics Co. Ltd., Shanghai, China
| | - Xiaowen He
- Innovation & Research Department, OriCell Therapeutics Co. Ltd., Shanghai, China
| |
Collapse
|
2
|
Swain HN, Boyce PD, Bromet BA, Barozinksy K, Hance L, Shields D, Olbricht GR, Semon JA. Mesenchymal stem cells in autoimmune disease: A systematic review and meta-analysis of pre-clinical studies. Biochimie 2024; 223:54-73. [PMID: 38657832 DOI: 10.1016/j.biochi.2024.04.009] [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: 01/30/2024] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Mesenchymal Stem Cells (MSCs) are of interest in the clinic because of their immunomodulation capabilities, capacity to act upstream of inflammation, and ability to sense metabolic environments. In standard physiologic conditions, they play a role in maintaining the homeostasis of tissues and organs; however, there is evidence that they can contribute to some autoimmune diseases. Gaining a deeper understanding of the factors that transition MSCs from their physiological function to a pathological role in their native environment, and elucidating mechanisms that reduce their therapeutic relevance in regenerative medicine, is essential. We conducted a Systematic Review and Meta-Analysis of human MSCs in preclinical studies of autoimmune disease, evaluating 60 studies that included 845 patient samples and 571 control samples. MSCs from any tissue source were included, and the study was limited to four autoimmune diseases: multiple sclerosis, rheumatoid arthritis, systemic sclerosis, and lupus. We developed a novel Risk of Bias tool to determine study quality for in vitro studies. Using the International Society for Cell & Gene Therapy's criteria to define an MSC, most studies reported no difference in morphology, adhesion, cell surface markers, or differentiation into bone, fat, or cartilage when comparing control and autoimmune MSCs. However, there were reported differences in proliferation. Additionally, 308 biomolecules were differentially expressed, and the abilities to migrate, invade, and form capillaries were decreased. The findings from this study could help to explain the pathogenic mechanisms of autoimmune disease and potentially lead to improved MSC-based therapeutic applications.
Collapse
Affiliation(s)
- Hailey N Swain
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Parker D Boyce
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Bradley A Bromet
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Kaiden Barozinksy
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Lacy Hance
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Dakota Shields
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, USA.
| |
Collapse
|
3
|
Peng Y, Zhang X, Tang Y, He S, Rao G, Chen Q, Xue Y, Jin H, Liu S, Zhou Z, Xiang Y. Role of autoreactive Tc17 cells in the pathogenesis of experimental autoimmune encephalomyelitis. NEUROPROTECTION 2024; 2:49-59. [DOI: 10.1002/nep3.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/16/2024] [Indexed: 07/04/2024]
Abstract
AbstractBackgroundThe pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE—an animal model of MS) is primarily mediated by T cells. However, recent studies have only focused on interleukin (IL)‐17‐secreting CD4+ T‐helper cells, also known as Th17 cells. This study aimed to compare Th17 cells and IL‐17‐secreting CD8+ T‐cytotoxic cells (Tc17) in the context of MS/EAE.MethodsFemale C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein peptides 35–55 (MOG35–55), pertussis toxin, and complete Freund's adjuvant to establish the EAE animal model. T cells were isolated from the spleen (12–14 days postimmunization). CD4+ and CD8+ T cells were purified using isolation kit and then differentiated into Th17 and Tc17, respectively, using MOG35–55 and IL‐23. The secretion levels of interferon‐γ (IFN‐γ) and IL‐17 were measured via enzyme‐linked immunosorbent assay using cultured CD4+ and CD8+ T cell supernatants. The pathogenicity of Tc17 and Th17 cells was assessed through adoptive transfer (tEAE), with the clinical course assessed using an EAE score (0–5). Hematoxylin and eosin as well as Luxol fast blue staining were used to examine the spinal cord. Purified CD8+ CD3+ and CD4+ CD3+ cells differentiated into Tc17 and Th17 cells, respectively, were stimulated with MOG35–55 peptide for proliferation assays.ResultsThe results showed that Tc17 cells (15,951 ± 1985 vs. 55,709 ± 4196 cpm; p < 0.050) exhibited a weaker response to highest dose (20 μg/mL) MOG35–55 than Th17 cells. However, this response was not dependent on Th17 cells. After the 48 h stimulation, at the highest dose (20 μg/mL) of MOG35–55. Tc17 cells secreted lower levels of IFN‐γ (280.00 ± 15.00 vs. 556.67 ± 15.28 pg/mL, p < 0.050) and IL‐17 (102.67 ± 5.86 pg/mL vs. 288.33 ± 12.58 pg/mL; p < 0.050) than Th17 cells. Similar patterns were observed for IFN‐γ secretion at 96 and 144 h. Furthermore, Tc17 cell‐induced tEAE mice exhibited similar EAE scores to Th17 cell‐induced tEAE mice and also showed similar inflammation and demyelination.ConclusionThe degree of pathogenicity of Tc17 cells in EAE is lower than that of Th17 cells. Future investigation on different immune cells and EAE models is warranted to determine the mechanisms underlying MS.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Xiuli Zhang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yandan Tang
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shunqing He
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Guilan Rao
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Quan Chen
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Yahui Xue
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Hong Jin
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shu Liu
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Ziyang Zhou
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yun Xiang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| |
Collapse
|
4
|
Wemlinger SM, Cambier JC. Therapeutic tactics for targeting B lymphocytes in autoimmunity and cancer. Eur J Immunol 2024; 54:e2249947. [PMID: 37816494 DOI: 10.1002/eji.202249947] [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: 01/13/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
B lymphocytes have become a very popular therapeutic target in a number of autoimmune indications due to their newly appreciated roles, and approachability, in these diseases. Many of the therapies now applied in autoimmunity were initially developed to deplete malignant B cells. These strategies have also been found to benefit patients suffering from such autoimmune diseases as multiple sclerosis, type I diabetes, systemic lupus erythematosus, and rheumatoid arthritis, to name a few. These observations have supported the expansion of research addressing the mechanistic contributions of B cells in these diseases, as well as blossoming of therapeutics that target them. This review seeks to summarize cutting-edge modalities for targeting B cells, including monoclonal antibodies, bispecific antibodies, antibody-drug conjugates, chimeric antigen receptor-T cells, and small molecule inhibitors. Efforts to refine B-cell targeted therapy to eliminate only pathogenic autoreactive cells will be addressed as well as the potential for future B-cell-based cellular therapeutics. Finally, we also address approaches that seek to silence B-cell function without depletion.
Collapse
Affiliation(s)
- Scott M Wemlinger
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
5
|
Tkachenko A, Kupcova K, Havranek O. B-Cell Receptor Signaling and Beyond: The Role of Igα (CD79a)/Igβ (CD79b) in Normal and Malignant B Cells. Int J Mol Sci 2023; 25:10. [PMID: 38203179 PMCID: PMC10779339 DOI: 10.3390/ijms25010010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
B-cell receptor (BCR) is a B cell hallmark surface complex regulating multiple cellular processes in normal as well as malignant B cells. Igα (CD79a)/Igβ (CD79b) are essential components of BCR that are indispensable for its functionality, signal initiation, and signal transduction. CD79a/CD79b-mediated BCR signaling is required for the survival of normal as well as malignant B cells via a wide signaling network. Recent studies identified the great complexity of this signaling network and revealed the emerging role of CD79a/CD79b in signal integration. In this review, we have focused on functional features of CD79a/CD79b, summarized signaling consequences of CD79a/CD79b post-translational modifications, and highlighted specifics of CD79a/CD79b interactions within BCR and related signaling cascades. We have reviewed the complex role of CD79a/CD79b in multiple aspects of normal B cell biology and how is the normal BCR signaling affected by lymphoid neoplasms associated CD79A/CD79B mutations. We have also summarized important unresolved questions and highlighted issues that remain to be explored for better understanding of CD79a/CD79b-mediated signal transduction and the eventual identification of additional therapeutically targetable BCR signaling vulnerabilities.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Kristyna Kupcova
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
- First Department of Internal Medicine–Hematology, General University Hospital and First Faculty of Medicine, Charles University, 128 08 Prague, Czech Republic
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
- First Department of Internal Medicine–Hematology, General University Hospital and First Faculty of Medicine, Charles University, 128 08 Prague, Czech Republic
| |
Collapse
|
6
|
Liu Y, Zhang Z, Kang Z, Zhou XJ, Liu S, Guo S, Jin Q, Li T, Zhou L, Wu X, Wang YN, Lu L, He Y, Li F, Zhang H, Liu Y, Xu H. Interleukin 4-driven reversal of self-reactive B cell anergy contributes to the pathogenesis of systemic lupus erythematosus. Ann Rheum Dis 2023; 82:1444-1454. [PMID: 37567607 DOI: 10.1136/ard-2023-224453] [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: 05/19/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023]
Abstract
OBJECTIVES Reactivation of anergic autoreactive B cells (BND cells) is a key aetiological process in systemic lupus erythematosus (SLE), yet the underlying mechanism remains largely elusive. This study aimed to investigate how BND cells participate in the pathogenesis of SLE and the underlying mechanism. METHODS A combination of phenotypical, large-scale transcriptome and B cell receptor (BCR) repertoire profiling were employed at molecular and single cell level on samples from healthy donors and patients with SLE. Isolated naïve B cells from human periphery blood were treated with anti-CD79b mAb in vitro to induce anergy. IgM internalisation was tracked by confocal microscopy and was qualified by flow cytometer. RESULTS We characterised the decrease and disruption of BND cells in SLE patients and demonstrated IL-4 as an important cytokine to drive such pathological changes. We then elucidated that IL-4 reversed B cell anergy by promoting BCR recycling to the cell surface via STAT6 signalling. CONCLUSIONS We demonstrated the significance of IL-4 in reversing B cell anergy and established the scientific rationale to treat SLE via blocking IL-4 signalling, also providing diagnostic and prognostic biomarkers to identify patients who are most likely going to benefit from such treatments.
Collapse
Affiliation(s)
- Yaoyang Liu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhiguo Zhang
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zijian Kang
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xu-Jie Zhou
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) Ministry of Education, Beijing, China
| | - Shujun Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shicheng Guo
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Qianmei Jin
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ting Li
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ling Zhou
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Wu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yan-Na Wang
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) Ministry of Education, Beijing, China
| | - Liangjing Lu
- Department of Rheumatology and Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanran He
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Fubin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) Ministry of Education, Beijing, China
| | - Yuncai Liu
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Boyles JS, Sadowski D, Potter S, Vukojicic A, Parker J, Chang WY, Ma YL, Chambers MG, Nelson J, Barmettler B, Smith EM, Kersjes K, Himes ER, Lin C, Lucchesi J, Brahmbhatt J, Sina R, Martin JA, Maestri E, Wiethoff CM, Dyas GL, Linnik MD, Na S, Witcher DR, Budelsky A, Rubtsova K. A nondepleting anti-CD19 antibody impairs B cell function and inhibits autoimmune diseases. JCI Insight 2023; 8:e166137. [PMID: 37427592 PMCID: PMC10371335 DOI: 10.1172/jci.insight.166137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/19/2023] [Indexed: 07/11/2023] Open
Abstract
B cells contribute to multiple aspects of autoimmune disorders, and B cell-targeting therapies, including B cell depletion, have been proven to be efficacious in treatment of multiple autoimmune diseases. However, the development of novel therapies targeting B cells with higher efficacy and a nondepleting mechanism of action is highly desirable. Here we describe a nondepleting, high-affinity anti-human CD19 antibody LY3541860 that exhibits potent B cell inhibitory activities. LY3541860 inhibits B cell activation, proliferation, and differentiation of primary human B cells with high potency. LY3541860 also inhibits human B cell activities in vivo in humanized mice. Similarly, our potent anti-mCD19 antibody also demonstrates improved efficacy over CD20 B cell depletion therapy in multiple B cell-dependent autoimmune disease models. Our data indicate that anti-CD19 antibody is a highly potent B cell inhibitor that may have potential to demonstrate improved efficacy over currently available B cell-targeting therapies in treatment of autoimmune conditions without causing B cell depletion.
Collapse
Affiliation(s)
- Jeffrey S. Boyles
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Dorota Sadowski
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Scott Potter
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Aleksandra Vukojicic
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - James Parker
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - William Y. Chang
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Yanfei L. Ma
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Mark G. Chambers
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - James Nelson
- Biotechnology Discovery Research, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Barbra Barmettler
- Biotechnology Discovery Research, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Eric M. Smith
- Biotechnology Discovery Research, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Kara Kersjes
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Evan R. Himes
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Chaohua Lin
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jonathan Lucchesi
- Immunology Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jaladhi Brahmbhatt
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Ramtin Sina
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jennifer A. Martin
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Evan Maestri
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Christopher M. Wiethoff
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Gregory L. Dyas
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Matthew D. Linnik
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Songqing Na
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Derrick R. Witcher
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Alison Budelsky
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| | - Kira Rubtsova
- Immunology Discovery, Lilly Biotechnology Center, Lilly Research Laboratories, Eli Lilly and Company, San Diego, California, USA
| |
Collapse
|
8
|
Fong CC, Spencer J, Howlett-Prieto Q, Feng X, Reder AT. Adaptive and innate immune responses in multiple sclerosis with anti-CD20 therapy: Gene expression and protein profiles. Front Neurol 2023; 14:1158487. [PMID: 37168665 PMCID: PMC10166068 DOI: 10.3389/fneur.2023.1158487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Background Anti-CD20 is a highly effective therapy for multiple sclerosis (MS), a disease with multiple abnormalities in function of B and T cells and innate immune cells. Anti-CD20 therapy depletes B cells, which alters antibody production and has diverse effects on B cell immunity. These changes potentially affect immunity beyond B cells in MS. Objective Determine if anti-CD20 therapy effects non-B cell, as well as B cell, gene expression, and serum protein levels. Methods Samples were collected from 10 healthy controls and from clinically stable relapsing-remitting MS - 10 untreated, 9 interferon-β-treated, and 15 ocrelizumab-treated patients were studied before, and 2 weeks and 6 months after, the first anti-CD20 infusion. Peripheral blood mononuclear cells (PBMC) were analyzed with sensitive, 135,000-transcript RNA expression microarrays, using stringent criteria. Gene expression was compared to 43 MS-relevant serum immune and neurotrophic proteins, using multiplex protein assays. Results Anti-CD20 therapy reduced expression of 413 total genes and 185 B-cell-regulated genes at 2 weeks vs. pre-therapy. Expression of 19 (15%) of these B cell genes returned toward baseline by 6 months, including genes for the B cell activation protein, CD79A, and for immunoglobulin A, D, and G heavy chains. Expression pathways for Th17 and CD4 regulatory T-cell (Treg) development, differentiation, and proliferation also quieted. In contrast, expression increased in Th1 and myeloid cell antiviral, pro-inflammatory, and toll-like receptor (TLR) gene pathways. Conclusion These findings have clinical implications. B cell gene expression diminishes 2 weeks after anti-CD20 antibody infusion, but begins to rebound by 6 months. This suggests that the optimum time for vaccination is soon before reinfusion of anti-CD20 therapy. In addition, at 6 months, there is enhanced Th1 cell gene expression and induction of innate immune response genes and TLR expression, which can enhance anti-viral and anti-tumor immunity. This may compensate for diminished B cell gene expression after therapy. These data suggest that anti-CD20 therapy has dynamic effect on B cells and causes a compensatory rise in Th1 and myeloid immunity.
Collapse
Affiliation(s)
| | | | | | - Xuan Feng
- Department of Neurology, University of Chicago Medicine, Chicago, IL, United States
| | - Anthony T. Reder
- Department of Neurology, University of Chicago Medicine, Chicago, IL, United States
| |
Collapse
|
9
|
Courtwright A, Atkinson C, Pelaez A. The Highly Sensitized Recipient: Pretransplant and Posttransplant Considerations. Clin Chest Med 2023; 44:85-93. [PMID: 36774171 DOI: 10.1016/j.ccm.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Highly sensitized patients, who are often black and Hispanic women, are less likely to be listed for lung transplant and are at higher risk for prolonged waitlist time and waitlist death. In this review, the authors discuss strategies for improving access to transplant in this population, including risk stratification of crossing pretransplant donor-specific antibodies, based on antibody characteristics. The authors also review institutional protocols, such as perioperative desensitization, for tailoring transplant immunosuppression in the highly sensitized population. The authors conclude with suggestions for future research, including development of novel donor-specific antibody-directed therapeutics.
Collapse
Affiliation(s)
- Andrew Courtwright
- Hospital of University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Carl Atkinson
- University of Florida, 1600 Southwest Archer Road, Gainesville, FL 32608, USA
| | - Andres Pelaez
- Jackson Health System, University of Miami, Miller School of Medicine, Miami Transplant Institute, 1801 Northwest 9th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
10
|
Harley ITW, Allison K, Scofield RH. Polygenic autoimmune disease risk alleles impacting B cell tolerance act in concert across shared molecular networks in mouse and in humans. Front Immunol 2022; 13:953439. [PMID: 36090990 PMCID: PMC9450536 DOI: 10.3389/fimmu.2022.953439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Most B cells produced in the bone marrow have some level of autoreactivity. Despite efforts of central tolerance to eliminate these cells, many escape to periphery, where in healthy individuals, they are rendered functionally non-responsive to restimulation through their antigen receptor via a process termed anergy. Broad repertoire autoreactivity may reflect the chances of generating autoreactivity by stochastic use of germline immunoglobulin gene segments or active mechanisms may select autoreactive cells during egress to the naïve peripheral B cell pool. Likewise, it is unclear why in some individuals autoreactive B cell clones become activated and drive pathophysiologic changes in autoimmune diseases. Both of these remain central questions in the study of the immune system(s). In most individuals, autoimmune diseases arise from complex interplay of genetic risk factors and environmental influences. Advances in genome sequencing and increased statistical power from large autoimmune disease cohorts has led to identification of more than 200 autoimmune disease risk loci. It has been observed that autoantibodies are detectable in the serum years to decades prior to the diagnosis of autoimmune disease. Thus, current models hold that genetic defects in the pathways that control autoreactive B cell tolerance set genetic liability thresholds across multiple autoimmune diseases. Despite the fact these seminal concepts were developed in animal (especially murine) models of autoimmune disease, some perceive a disconnect between human risk alleles and those identified in murine models of autoimmune disease. Here, we synthesize the current state of the art in our understanding of human risk alleles in two prototypical autoimmune diseases - systemic lupus erythematosus (SLE) and type 1 diabetes (T1D) along with spontaneous murine disease models. We compare these risk networks to those reported in murine models of these diseases, focusing on pathways relevant to anergy and central tolerance. We highlight some differences between murine and human environmental and genetic factors that may impact autoimmune disease development and expression and may, in turn, explain some of this discrepancy. Finally, we show that there is substantial overlap between the molecular networks that define these disease states across species. Our synthesis and analysis of the current state of the field are consistent with the idea that the same molecular networks are perturbed in murine and human autoimmune disease. Based on these analyses, we anticipate that murine autoimmune disease models will continue to yield novel insights into how best to diagnose, prognose, prevent and treat human autoimmune diseases.
Collapse
Affiliation(s)
- Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
- Rheumatology Section, Medicine Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - R. Hal Scofield
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Medical/Research Service, US Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|