1
|
Sevdali E, Block V, Lataretu M, Li H, Smulski CR, Briem JS, Heitz Y, Fischer B, Ramirez NJ, Grimbacher B, Jäck HM, Voll RE, Hölzer M, Schneider P, Eibel H. BAFFR activates PI3K/AKT signaling in human naive but not in switched memory B cells through direct interactions with B cell antigen receptors. Cell Rep 2022; 39:111019. [PMID: 35767961 DOI: 10.1016/j.celrep.2022.111019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/27/2022] [Accepted: 06/08/2022] [Indexed: 12/28/2022] Open
Abstract
Binding of BAFF to BAFFR activates in mature B cells PI3K/AKT signaling regulating protein synthesis, metabolic fitness, and survival. In humans, naive and memory B cells express the same levels of BAFFR, but only memory B cells seem to survive without BAFF. Here, we show that BAFF activates PI3K/AKT only in naive B cells and changes the expression of genes regulating migration, proliferation, growth, and survival. BAFF-induced PI3K/AKT activation requires direct interactions between BAFFR and the B cell antigen receptor (BCR) components CD79A and CD79B and is enhanced by the AKT coactivator TCL1A. Compared to memory B cells, naive B cells express more surface BCRs, which interact better with BAFFR than IgG or IgA, thus allowing stronger responses to BAFF. As ablation of BAFFR in naive and memory B cells causes cell death independent of BAFF-induced signaling, BAFFR seems to act also as an intrinsic factor for B cell survival.
Collapse
Affiliation(s)
- Eirini Sevdali
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Violeta Block
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Marie Lataretu
- RNA Bioinformatics and High-Throughput Analysis, Faculty of Mathematics and Computer Science, University of Jena, Leutragraben 1, 07743 Jena, Germany
| | - Huiying Li
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Cristian R Smulski
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida E-Bustillo 9500, R8402AGP Río Negro, San Carlos de Bariloche, Argentina
| | - Jana-Susann Briem
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Yannic Heitz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Beate Fischer
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Neftali-Jose Ramirez
- Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany; Institute for Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Bodo Grimbacher
- Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany; Institute for Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Hans-Martin Jäck
- Department of Medicine, Division of Immunology, University of Erlangen, Glückstraße 6, 91054 Erlangen, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Martin Hölzer
- Methodology and Research Infrastructure, MF1 Bioinformatics, Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Ch. des Boveresses 155, 1066 Epalinges, Switzerland
| | - Hermann Eibel
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79110 Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany.
| |
Collapse
|
2
|
Borhis G, Trovato M, Ibrahim HM, Isnard S, Le Grand R, Bosquet N, Richard Y. Impact of BAFF Blockade on Inflammation, Germinal Center Reaction and Effector B-Cells During Acute SIV Infection. Front Immunol 2020; 11:252. [PMID: 32194549 PMCID: PMC7061218 DOI: 10.3389/fimmu.2020.00252] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
Memory B-cell dysfunctions and inefficient antibody response suggest germinal center (GC) impairments during HIV/SIV infection with possible contribution of overproduced B-cell activating factor (BAFF). To address this question, we compared proportions and functions of various B-cell subsets and follicular helper T-cells (TFH) in untreated (Placebo) and BR3-Fc treated (Treated) SIV-infected macaques. From day 2 post-infection (dpi), Treated macaques received one weekly injection of BR3-Fc molecule, a soluble BAFF antagonist, for 4 weeks. Whereas, the kinetics of CD4+ T-cell loss and plasma viral loads were comparable in both groups, BAFF blockade delayed the peak of inflammatory cytokines (CXCL10, IFNα), impaired the renewal of plasmacytoid dendritic cells and fostered the decline of plasma CXCL13 titers after 14 dpi. In Treated macaques, proportions of total and naïve B-cells were reduced in blood and spleen whereas SIV-induced loss of marginal zone (MZ) B-cells was only accentuated in blood and terminal ileum. Proportions of spleen GC B-cells and TFH were similar in both groups, with CD8+ T-cells and rare Foxp3+ being present in spleen GC. Regardless of treatment, sorted TFH produced similar levels of IL21, CXCL13, and IFNγ but no IL2, IL4, or BAFF and exhibited similar capacities to support IgG production by autologous or heterologous B-cells. Consistently, most TFH were negative for BAFF-R and TACI. Higher proportions of resting and atypical (CD21lo) memory B-cells were present in Treated macaques compared to Placebo. In both groups, we found higher levels of BAFF-R expression on MZ and resting memory B-cells but low levels on atypical memory B-cells. TACI was present on 20-30% of MZ, resting and atypical memory B-cells in Placebo macaques. BAFF blockade decreased TACI expression on these B-cell subsets as well as titers of SIV-specific and vaccine-specific antibodies arguing for BAFF being mandatory for plasma cell survival. Irrespective of treatment, GC B-cells expressed BAFF-R at low level and were negative for TACI. In addition to key information on spleen BAFF-R and TACI expression, our data argue for BAFF contributing to the GC reaction in terminal ileum but being dispensable for the generation of atypical memory B-cells and GC reaction in spleen during T-dependent response against SIV.
Collapse
Affiliation(s)
- Gwenoline Borhis
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Maria Trovato
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Hany M. Ibrahim
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Stephane Isnard
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Roger Le Grand
- CEA, Université Paris Sud, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department/IBFJ, Fontenay-aux-Roses, France
| | - Nathalie Bosquet
- CEA, Université Paris Sud, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department/IBFJ, Fontenay-aux-Roses, France
| | - Yolande Richard
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| |
Collapse
|
3
|
Hale M, Rawlings DJ, Jackson SW. The long and the short of it: insights into the cellular source of autoantibodies as revealed by B cell depletion therapy. Curr Opin Immunol 2018; 55:81-88. [PMID: 30390507 DOI: 10.1016/j.coi.2018.10.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 10/17/2018] [Indexed: 10/28/2022]
Abstract
High titers of pathogenic autoantibodies are a hallmark of many autoimmune diseases. However, much remains unknown about the self-reactive plasma cells that are key mediators of disease. We propose a model in which the varying efficacy of precursor B cell depletion for the treatment of humoral autoimmunity can be explained by differences in the relative contributions of pathogenic antibodies by short-lived versus long-lived plasma cells. Beyond therapeutic considerations, this model suggests that we can infer the cellular source of disease-associated autoantibodies by the durability of serum titers following B cell depletion. Data from clinical trials and animal models across different autoimmune diseases may provide useful insights into the lifespan, lifestyle and fate of autoreactive plasma cells.
Collapse
Affiliation(s)
- Malika Hale
- Seattle Children's Research Institute, Seattle, WA, United States
| | - David J Rawlings
- Seattle Children's Research Institute, Seattle, WA, United States; Department of Immunology, University of Washington, School of Medicine, United States; Department of Pediatrics, University of Washington, School of Medicine, United States
| | - Shaun W Jackson
- Seattle Children's Research Institute, Seattle, WA, United States; Department of Pediatrics, University of Washington, School of Medicine, United States.
| |
Collapse
|
4
|
B-1a lymphocytes promote oligodendrogenesis during brain development. Nat Neurosci 2018; 21:506-516. [PMID: 29507409 DOI: 10.1038/s41593-018-0106-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 01/08/2018] [Indexed: 01/22/2023]
Abstract
During brain development, the immune system mediates neurogenesis, gliogenesis and synapse formation. However, it remains unclear whether peripheral lymphocytes contribute to brain development. Here we identified the subtypes of lymphocytes that are present in neonatal mouse brains and investigated their functions. We found that B-1a cells, a subtype of B cells, were abundant in the neonatal mouse brain and infiltrated into the brain in a CXCL13-CXCR5-dependent manner. B-1a cells promoted the proliferation of oligodendrocyte-precursor cells (OPCs) in vitro, and depletion of B-1a cells from developing brains resulted in a reduction of numbers of OPCs and mature oligodendrocytes. Furthermore, neutralizing Fcα/μR, the receptor for the Fc region of IgM secreted by B-1a cells, inhibited OPC proliferation and reduced the proportion of myelinated axons in neonatal mouse brains. Our results demonstrate that B-1a cells infiltrate into the brain and contribute to oligodendrogenesis and myelination by promoting OPC proliferation via IgM-Fcα/μR signaling.
Collapse
|
5
|
Liu Y, Zhan F, Zhang X, Lin S. Toll-like receptor-9 is involved in the development of B cell stimulating factor-induced systemic lupus erythematosus. Exp Ther Med 2018; 15:585-591. [PMID: 29387207 DOI: 10.3892/etm.2017.5411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/25/2017] [Indexed: 11/06/2022] Open
Abstract
The objective of the present study was to investigate the role of Toll-like receptor (TLR)-9 in B lymphocyte stimulating factor (BLyS)-induced systemic lupus erythematosus (SLE) in mice. The anti-double stranded (ds)DNA antibody titer, levels of complement proteins (C3 and C4), interleukin (IL)-10 and the disease activity [assessed by the erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) level] were measured. A total of 21 transgenic female mice (aged 8-10 weeks and weighing 30-40 g) expressing the Epstein-Barr virus membrane antigen, BLLF1, were studied. Mice were randomly divided into the control, the BLyS inhibition and the TLR-9 inhibition groups, with 7 mice in each group. Mice in the blank control group received intraperitoneal injections of normal saline, mice in the BLyS inhibition group received intraperitoneal injections of anti-BR3 monoclonal antibody (5,000 ng/day) and mice in the TLR-9 inhibition group received intraperitoneal injections of anti-human TLR-9 antibody (250 ng/day). The treatment regimens continued for 10 days, followed by the collection of peripheral venous blood. The relative levels of TLR-9 mRNA were measured by reverse transcription-quantitative polymerase chain reaction. Furthermore, the BLyS protein concentration and IL-10 levels were measured by ELISA. TLR-9 mRNA, BLyS, IL-10, anti-dsDNA antibody titer, C3, C4, ESR and CRP levels of the blank control group were significantly higher than those of the other two groups (P<0.05). The differences in comparison of these indexes between the BLyS inhibition and TLR-9 inhibition groups were not statistically significant (P>0.05), with the exception of TLR-9 mRNA and BLyS. In conclusion, the TLR-9 signaling pathway may be important for BLyS-induced SLE, and regulation of the inflammatory immune level.
Collapse
Affiliation(s)
- Ying Liu
- Graduate School of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Rheumatology and Clinical Immunology, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China.,Department of Rheumatology and Clinical Immunology, Hainan General Hospital, Haikou, Hainan 570000, P.R. China
| | - Feng Zhan
- Department of Rheumatology and Clinical Immunology, Hainan General Hospital, Haikou, Hainan 570000, P.R. China
| | - Xiao Zhang
- Graduate School of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Rheumatology and Clinical Immunology, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Shudian Lin
- Department of Rheumatology and Clinical Immunology, Hainan General Hospital, Haikou, Hainan 570000, P.R. China
| |
Collapse
|
6
|
Qin H, Wei G, Sakamaki I, Dong Z, Cheng WA, Smith DL, Wen F, Sun H, Kim K, Cha S, Bover L, Neelapu SS, Kwak LW. Novel BAFF-Receptor Antibody to Natively Folded Recombinant Protein Eliminates Drug-Resistant Human B-cell Malignancies In Vivo. Clin Cancer Res 2017; 24:1114-1123. [PMID: 29180606 DOI: 10.1158/1078-0432.ccr-17-1193] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/20/2017] [Accepted: 11/21/2017] [Indexed: 02/05/2023]
Abstract
Purpose: mAbs such as anti-CD20 rituximab are proven therapies in B-cell malignancies, yet many patients develop resistance. Novel therapies against alternative targets are needed to circumvent resistance mechanisms. We sought to generate mAbs against human B-cell-activating factor receptor (BAFF-R/TNFRSF13C), which has not yet been targeted successfully for cancer therapy.Experimental Design: Novel mAbs were generated against BAFF-R, expressed as a natively folded cell surface immunogen on mouse fibroblast cells. Chimeric BAFF-R mAbs were developed and assessed for in vitro and in vivo monotherapy cytotoxicity. The chimeric mAbs were tested against human B-cell tumor lines, primary patient samples, and drug-resistant tumors.Results: Chimeric antibodies bound with high affinity to multiple human malignant B-cell lines and induced potent antibody-dependent cellular cytotoxicity (ADCC) against multiple subtypes of human lymphoma and leukemia, including primary tumors from patients who had relapsed after anti-CD20 therapy. Chimeric antibodies also induced ADCC against ibrutinib-resistant and rituximab-insensitive CD20-deficient variant lymphomas, respectively. Importantly, they demonstrated remarkable in vivo growth inhibition of drug-resistant tumor models in immunodeficient mice.Conclusions: Our method generated novel anti-BAFF-R antibody therapeutics with remarkable single-agent antitumor effects. We propose that these antibodies represent an effective new strategy for targeting and treating drug-resistant B-cell malignancies and warrant further development. Clin Cancer Res; 24(5); 1114-23. ©2017 AACR.
Collapse
Affiliation(s)
- Hong Qin
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California.
| | - Guowei Wei
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Ippei Sakamaki
- Department of Clinical Infectious diseases, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Zhenyuan Dong
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Wesley A Cheng
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - D Lynne Smith
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Feng Wen
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California.,Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| | - Han Sun
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California
| | - Kunhwa Kim
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Soungchul Cha
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Bover
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Larry W Kwak
- Beckman Research Institute City of Hope National Medical Center, Toni Stephenson Lymphoma Center, and Department of Hematology and Hematopoietic Stem Cell Transplantation, Duarte, California.
| |
Collapse
|
7
|
B-Cell Depletion Reduces the Maturation of Cerebral Cavernous Malformations in Murine Models. J Neuroimmune Pharmacol 2016; 11:369-77. [PMID: 27086141 DOI: 10.1007/s11481-016-9670-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/30/2016] [Indexed: 02/05/2023]
Abstract
Cerebral cavernous malformations (CCMs) are relatively common vascular malformations, characterized by increased Rho kinase (ROCK) activity, vascular hyper-permeability and the presence of blood degradation products including non-heme iron. Previous studies revealed robust inflammatory cell infiltration, selective synthesis of IgG, in situ antigen driven B-cell clonal expansion, and deposition of immune complexes and complement proteins within CCM lesions. We aimed to evaluate the impact of suppressing the immune response on the formation and maturation of CCM lesions, as well as lesional iron deposition and ROCK activity. Two murine models of heterozygous Ccm3 (Pdcd10), which spontaneously develop CCM lesions with severe and milder phenotypes, were either untreated or received anti-mouse BR3 to deplete B cells. Brains from anti-mouse BR3-treated mice exhibited significantly fewer mature CCM lesions and smaller lesions compared to untreated mice. B cell depletion halted the progression of lesions into mature stage 2 lesions but did not prevent their genesis. Non-heme iron deposition and ROCK activity was decreased in lesions of B cell depleted mice. This represents the first report of the therapeutic benefit of B-cell depletion in the development and progression of CCMs, and provides a proof of principle that B cells play a critical role in CCM lesion genesis and maturation. These findings add biologics to the list of potential therapeutic agents for CCM disease. Future studies would characterize the putative antigenic trigger and further define the mechanism of immune response in the lesions.
Collapse
|
8
|
Kaileh M, Vazquez E, MacFarlane AW, Campbell K, Kurosaki T, Siebenlist U, Sen R. mTOR-Dependent and Independent Survival Signaling by PI3K in B Lymphocytes. PLoS One 2016; 11:e0146955. [PMID: 26785352 PMCID: PMC4718598 DOI: 10.1371/journal.pone.0146955] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/22/2015] [Indexed: 01/16/2023] Open
Abstract
Peripheral B lymphocyte survival requires the B cell receptor (BCR) and B cell activating factor (BAFF) binding to its receptor (BAFF-R). Deletion of the BCR, or its signal transducing chaperone Igβ, leads to rapid loss of mature B cells, indicating that signals initiated at the BCR are crucial for B cell survival. BAFF or BAFF-R deficiency also significantly reduces the numbers of mature B cells despite normal BCR expression. Together, these observations indicate that continued BCR and BAFF-R signaling are essential for the survival of mature resting B cells in the periphery. Here we demonstrate that tonic BCR signals up-regulate p100 (Nfkb2) as well as Mcl-1 protein expression at a post-transcriptional level via a PI3K-dependent pathway. p100 expression is mTOR-independent, whereas Mcl-1 expression is mTOR-dependent. BAFF treatment further elevated Mcl-1 levels by an mTOR-independent pathway, while consuming p100. Accordingly, Mcl-1 induction by BAFF is abrogated in Nfkb2-/- B cells. We propose that the cumulative effects of the BCR and BAFF-R signaling pathways increase Mcl-1 levels beyond the threshold required for B cell survival.
Collapse
Affiliation(s)
- Mary Kaileh
- Gene Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Estefania Vazquez
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexander W. MacFarlane
- Fox Chase Cancer Center, Division of Basic Science, Institute for Cancer Research, Philadelphia, Pennsylvania, United States of America
| | - Kerry Campbell
- Fox Chase Cancer Center, Division of Basic Science, Institute for Cancer Research, Philadelphia, Pennsylvania, United States of America
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Ulrich Siebenlist
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ranjan Sen
- Gene Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
9
|
Brightbill HD, Jackman JK, Suto E, Kennedy H, Jones C, Chalasani S, Lin Z, Tam L, Roose-Girma M, Balazs M, Austin CD, Lee WP, Wu LC. Conditional Deletion of NF-κB-Inducing Kinase (NIK) in Adult Mice Disrupts Mature B Cell Survival and Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:953-64. [PMID: 26116508 DOI: 10.4049/jimmunol.1401514] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 05/30/2015] [Indexed: 01/19/2023]
Abstract
NF-κB-inducing kinase (NIK) is a primary regulator of the noncanonical NF-κB signaling pathway, which plays a vital role downstream of BAFF, CD40L, lymphotoxin, and other inflammatory mediators. Germline deletion or inactivation of NIK in mice results in the defective development of B cells and secondary lymphoid organs, but the role of NIK in adult animals has not been studied. To address this, we generated mice containing a conditional allele of NIK. Deletion of NIK in adult mice results in decreases in B cell populations in lymph nodes and spleen, similar to what is observed upon blockade of BAFF. Consistent with this, B cells from mice in which NIK is acutely deleted fail to respond to BAFF stimulation in vitro and in vivo. In addition, mice with induced NIK deletion exhibit a significant decrease in germinal center B cells and serum IgA, which is indicative of roles for NIK in additional pathways beyond BAFF signaling. Our conditional NIK-knockout mice may be broadly useful for assessing the postdevelopmental and cell-specific roles of NIK and the noncanonical NF-κB pathway in mice.
Collapse
Affiliation(s)
- Hans D Brightbill
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080;
| | - Janet K Jackman
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Eric Suto
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Heather Kennedy
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080; and
| | - Charles Jones
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080; and
| | - Sreedevi Chalasani
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080; and
| | - Zhonghua Lin
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Lucinda Tam
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080
| | - Meron Roose-Girma
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080
| | - Mercedesz Balazs
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Cary D Austin
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080; and
| | - Wyne P Lee
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Lawren C Wu
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080;
| |
Collapse
|
10
|
Lin W, Seshasayee D, Lee WP, Caplazi P, McVay S, Suto E, Nguyen A, Lin Z, Sun Y, DeForge L, Balazs M, Martin F, Zarrin AA. Dual B cell immunotherapy is superior to individual anti-CD20 depletion or BAFF blockade in murine models of spontaneous or accelerated lupus. Arthritis Rheumatol 2015; 67:215-24. [PMID: 25303150 PMCID: PMC4312898 DOI: 10.1002/art.38907] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 10/02/2014] [Indexed: 12/18/2022]
Abstract
Objective To determine whether a combination of B cell depletion and BAFF blockade is more effective than monotherapy in treating models of spontaneous or accelerated systemic lupus erythematosus (SLE) in (NZB × NZW)F1 mice. Methods Clinical parameters such as disease progression–free survival, proteinuria, and renal injury were assessed in models of spontaneous, interferon-α (IFNα)–accelerated, or pristane-accelerated lupus in (NZB × NZW)F1 mice. Treatment arms included anti-CD20 (B cell depletion), B lymphocyte stimulator receptor 3 fusion protein (BR-3-Fc) (BAFF blockade), the combination of anti-CD20 and BR-3-Fc, isotype control, or cyclophosphamide. In models of spontaneous, IFNα-accelerated, or pristane-accelerated lupus, mice were treated for 24 weeks, 8 weeks, or 12 weeks, respectively. Peripheral and resident B cell subsets and various autoantibodies were examined. Results Compared to B cell depletion or BAFF blockade alone, combined therapy significantly improved disease manifestations in all 3 lupus models. In addition, marginal zone B cells, plasmablasts, and circulating and tissue plasma cells were decreased more effectively. Dual B cell immunotherapy also reduced multiple classes of pathogenic autoantibodies, consistent with its observed effectiveness in reducing immune complex–mediated renal injury. Conclusion Dual immunotherapy via B cell depletion and BAFF blockade is more efficacious than single agent immunotherapy in murine SLE models, and this combination treatment is predicted to be an effective strategy for immunotherapy in human SLE.
Collapse
Affiliation(s)
- WeiYu Lin
- Genentech, Inc., South San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Stohl W. Editorial: The BAFFling Immunology of Systemic Lupus Erythematosus: Beyond B Cells. Arthritis Rheumatol 2015; 67:612-5. [DOI: 10.1002/art.38951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 11/04/2014] [Indexed: 01/07/2023]
|
12
|
Khan AR, Hams E, Floudas A, Sparwasser T, Weaver CT, Fallon PG. PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun 2015; 6:5997. [DOI: 10.1038/ncomms6997] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/01/2014] [Indexed: 01/05/2023] Open
|
13
|
Yang S, Li JY, Xu W. Role of BAFF/BAFF-R axis in B-cell non-Hodgkin lymphoma. Crit Rev Oncol Hematol 2014; 91:113-22. [PMID: 24629840 DOI: 10.1016/j.critrevonc.2014.02.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/20/2014] [Accepted: 02/14/2014] [Indexed: 12/23/2022] Open
Abstract
B-cell activating factor (BAFF), as a member of the tumor necrosis factor (TNF) ligand family, plays important roles in B-cell homeostasis, tolerance, and malignancy. BAFF binds to three receptors of TNF, TACI, BCMA and BAFF-receptor (BAFF-R). In particular, the BAFF/BAFF-R pathway is crucial to the survival and growth of mature normal and malignant B-cells. BAFF is displayed on the cell surface or is released in a soluble form after cleavage from the plasma membrane. BAFF-R as the main BAFF receptor is expressed mainly on B-cells. Aberrant BAFF expression was found in malignant B-cells from B-cell non-Hodgkin lymphoma (B-NHL) patients, which protects these cells from spontaneous or drug-induced apoptosis and stimulated NF-κB activation via autocrine and/or paracrine pathways. However, the mechanisms involved in the gene expression and regulation of BAFF or BAFF-R has not been elucidated. More importantly, the design of reagents able to counteract BAFF/BAFF-R pathways may be of therapeutic value for B-NHL. Results of ongoing clinical trials with BAFF or BAFF-R antagonists are eagerly awaited.
Collapse
Affiliation(s)
- Shu Yang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Jian-Yong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China.
| |
Collapse
|
14
|
Bonnan M. Intrathecal immune reset in multiple sclerosis: exploring a new concept. Med Hypotheses 2013; 82:300-9. [PMID: 24417802 DOI: 10.1016/j.mehy.2013.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/10/2013] [Accepted: 12/19/2013] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis impairment is mainly driven by the progressive phase, whose pathology remains elusive. No drug has yet been able to halt this phase so therapeutic management remains challenging. It was recently demonstrated that late disability correlates with the spreading of cortical subpial lesions, and tertiary lymphoid organs (TLO) were identified in close apposition with these lesions. TLO are of crucial importance since they are able to mount a complete local immune response, as observed in the intrathecal compartment from the moment MS is diagnosed (i.e. oligoclonal bands). This article examines the consequences of this intrathecal response: giving a worst clinical prognostic value and bearing arguments for possible direct brain toxicity, intrathecal secretion should be targeted by drugs abating both B-lymphocytes and plasma cells. Another consequence is that intrathecal secretion has value as a surrogate marker of the persistence of an ongoing intrathecal immune reaction after treatment. Although it is still unsure which mechanism or byproduct secreted by TLO triggers cortical lesions, we propose to target TLO components as a new therapeutic avenue in progressive MS. Whereas it was long considered that the inability of therapies to penetrate the blood-brain-barrier was a crucial obstacle, our proposed strategy will take advantage of the properties of the BBB to safely reset the intrathecal immune system in order to halt the slow axonal burning underlying secondary MS. We review the literature in support of the rationale for treating MS with intrathecal drugs dedicated to clearing the local immune response. Since many targets are involved, achieving this goal may require a combination of monoclonal antibodies targeting each cell sub-type. Hope might be rekindled with a one-shot intrathecal multi-drug treatment in progressive MS.
Collapse
Affiliation(s)
- Mickael Bonnan
- Service de Neurologie, Hôpital F. Mitterrand, 4 bd Hauterive, 64046 Pau, France.
| |
Collapse
|
15
|
Abstract
With the approval by the FDA in 2011 of a biologic agent (namely belimumab) for the treatment of systemic lupus erythematosus (SLE), optimism abounds that additional biologic (and nonbiologic) agents will be similarly endorsed. Given the numerous immune-based abnormalities associated with SLE, the potential therapeutic targets for biologic agents and the candidate biologic approaches are also numerous. These approaches include: biologic agents that promote B-cell depletion, B-cell inactivation, or the generation of regulatory B cells; biologic agents that induce T-cell tolerance, block T-cell activation and differentiation, or alter T-cell trafficking; biologic agents that target the B-cell activating factor (BAFF) axis, type I interferons, IL-6 and its receptor, or TNF; and the adoptive transfer of ex vivo-generated regulatory T cells. Owing to the great heterogeneity inherent to SLE, no single approach should be expected to be effective in all patients. As our understanding of the pathogenic mechanisms of SLE continues to expand, additional therapeutic targets and approaches will undoubtedly be identified and should be fully exploited.
Collapse
|
16
|
Page E, Kwun J, Oh B, Knechtle S. Lymphodepletional strategies in transplantation. Cold Spring Harb Perspect Med 2013; 3:3/7/a015511. [PMID: 23818516 DOI: 10.1101/cshperspect.a015511] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Because lymphocytes were shown to mediate transplant rejection, their depletion has been studied as a mechanism of preventing rejection and perhaps inducing immunologic tolerance. Agents that profoundly deplete lymphocytes have included monoclonal antibodies, cytotoxic drugs, and radiation. We have studied several such agents but focused on antibodies that deplete not only peripheral blood lymphocytes, but also lymph node lymphocytes. Depletion of lymph node T lymphocytes appears to permit peripheral tolerance at least for T cells in animal models. Nevertheless, B-cell responses may be resistant to such approaches, and T memory cells are likewise relatively resistant to depleting antibodies. We review the experimental and clinical approaches to depletion strategies and outline some of the pitfalls of depletion, such as limitations of currently available agents, duration of tolerance, infection, and malignancy. It is notable that most tolerogenic strategies that have been attempted experimentally and clinically include depleting agents even when they are not named as the underlying strategy. Thus, there is an implicitly acknowledged role for reducing the precursor frequency of donor antigen-specific lymphocytes when approaching the daunting goal of transplant tolerance.
Collapse
Affiliation(s)
- Eugenia Page
- Department of Surgery, Division of Transplantation, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | |
Collapse
|
17
|
Berrih-Aknin S, Ragheb S, Le Panse R, Lisak RP. Ectopic germinal centers, BAFF and anti-B-cell therapy in myasthenia gravis. Autoimmun Rev 2013; 12:885-93. [DOI: 10.1016/j.autrev.2013.03.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 12/19/2022]
|
18
|
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a systemic disease and the most prevalent of all autoimmune disorders. Here we review recent advances in the development and availability of biologic agents with a focus on monoclonal antibody or smaller formats of targeted engineered therapeutics including novel, non-antibody-based therapeutics. AREAS COVERED Today an array of biologics blocking either proinflammatory cytokines or lymphocyte activation/survival are available that enable a substantial improvement over conventional disease-modifying antirheumatic drugs (DMARDs). We review the engineering process of antibody-based biologics, their preclinical and clinical application, and current efforts to treat RA by interfering with B-cell function (notable targets covered are CD20, CD38, B-cell activating factor, transmembrane activator and calcium-modulating and cyclophilin interactor), with T-cell function (CD3, CD4, CD28), with bone erosion (RANKL), and with cytokines or growth factors (tumor necrosis factor, interleukin-1 [IL-1], IL-6, IL-17, VEGF). Future treatment choices might encompass the blockade or modulation of danger-associated molecular patterns such as HMGB1, pattern recognition receptors, messenger RNAs or noncoding RNAs, histone acetylation, and inflammasome components. EXPERT OPINION Although current therapies can reduce the signs and symptoms of RA for many patients, the quest for a cure (or a more complete blockade of the structural damage) in RA is still ongoing and will need treatment approaches, which are not exclusively confined to blocking a particular cytokine, receptor, or autoreactive B or T cell involved in disease progression. To this end exciting treatment alternatives and drug targets are on the horizon that may become available to patients in the future.
Collapse
Affiliation(s)
- Lukas Bossaller
- University of Massachusetts Medical School, Division of Infectious Diseases and Immunology, Worcester, MA, USA.
| | | |
Collapse
|
19
|
Abstract
Inhibitors of tumour necrosis factor (TNF) are among the most successful protein-based drugs (biologics) and have proven to be clinically efficacious at reducing inflammation associated with several autoimmune diseases. As a result, attention is focusing on the therapeutic potential of additional members of the TNF superfamily of structurally related cytokines. Many of these TNF-related cytokines or their cognate receptors are now in preclinical or clinical development as possible targets for modulating inflammatory diseases and cancer as well as other indications. This Review focuses on the biologics that are currently in clinical trials for immune-related diseases and other syndromes, discusses the successes and failures to date as well as the expanding therapeutic potential of modulating the activity of this superfamily of molecules.
Collapse
Affiliation(s)
- Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
20
|
Lisak RP, Ragheb S. The role of B cell-activating factor in autoimmune myasthenia gravis. Ann N Y Acad Sci 2012; 1274:60-7. [DOI: 10.1111/j.1749-6632.2012.06842.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Marathe A, Iyer S, Qiu ZJ, Visich J, Mager DE. Pharmacokinetics and pharmacodynamics of anti-BR3 monoclonal antibody in mice. Pharm Res 2012; 29:3180-7. [PMID: 22806404 DOI: 10.1007/s11095-012-0813-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 06/20/2012] [Indexed: 11/24/2022]
Abstract
PURPOSE To characterize the pharmacokinetic (PK) and pharmacodynamic (PD) properties of a monoclonal antibody directed against the B-cell activating factor (BAFF) receptor 3 (BR3), following intravenous (IV) and subcutaneous (SC) administration in mice. METHODS Single IV doses of 0.2, 2.0 and 20 mg/kg and a single SC injection of 20 mg/kg of anti-BR3 antibody was administered to mice. Serum drug and BAFF concentrations and splenic B-cell concentrations were measured at various time points. Pooled PK profiles were described by a two-compartmental model with time-dependent nonlinear elimination, and BAFF profiles were defined by an indirect response model. Fractional receptor occupancy served as the driving function for a competitive reversible antagonism model to characterize B-cell dynamics. RESULTS Noncompartmental analysis revealed a decrease in drug clearance (31.3 to 7.93 mL/day/kg) with increasing IV doses. The SC dose exhibited slow absorption (T(max) = 2 days) and complete bioavailability. All doses resulted in a dose-dependent increase in BAFF concentrations and decrease in B-cell counts. The proposed model reasonably captured complex PK/PD profiles of anti-BR3 antibody after IV and SC administration. CONCLUSIONS A mechanistic model was developed that describes the reversible competition between anti-BR3 antibody and BAFF for BR3 receptors and its influence on B-cell pharmacodynamics.
Collapse
Affiliation(s)
- Anshu Marathe
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, 431 Kapoor Hall, Buffalo, New York 14214, USA
| | | | | | | | | |
Collapse
|
22
|
Amezcua Vesely MC, Schwartz M, Bermejo DA, Montes CL, Cautivo KM, Kalergis AM, Rawlings DJ, Acosta-Rodríguez EV, Gruppi A. FcγRIIb and BAFF differentially regulate peritoneal B1 cell survival. THE JOURNAL OF IMMUNOLOGY 2012; 188:4792-800. [PMID: 22516957 DOI: 10.4049/jimmunol.1102070] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
B1 cells produce most natural Abs in unimmunized mice and play a key role in the response to thymus-independent Ags and microbial infection. Enlargement of B1 cell number in mice is often associated with autoimmunity. However, the factors that control peripheral B1 cell survival remain poorly characterized. Mice lacking the inhibitory receptor FcγRIIb exhibit a massive expansion in peritoneal B1 cells, implicating this receptor in B1 cell homeostasis. In this study, we show that peritoneal B1 cells express the highest levels of FcγRIIb among B cell subsets and are highly susceptible to FcγRIIb-mediated apoptosis. B1 cells upregulate FcγRIIb in response to innate signals, including CpG, and the B cell homeostatic cytokine BAFF efficiently protects activated B1 cells from FcγRIIb-mediated apoptosis via receptor downregulation. BAFF-transgenic mice manifest an expansion of peritoneal B1 cells that express lower levels of FcγRIIb and exhibit reduced susceptibility to apoptosis. Whereas both peritoneal B1 cells from wild-type and BAFF-transgenic mice immunized with CpG exhibit an increase in FcγRIIb levels, this change is blunted in BAFF-transgenic animals. Our combined results demonstrate that FcγRIIb controls peritoneal B1 cell survival and this program can be modulated by the BAFF signaling axis.
Collapse
Affiliation(s)
- María C Amezcua Vesely
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba CP 5000, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Danilenko DM, Wang H. The yin and yang of immunomodulatory biologics: assessing the delicate balance between benefit and risk. Toxicol Pathol 2012; 40:272-87. [PMID: 22222884 DOI: 10.1177/0192623311430237] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A number of therapeutic immunomodulatory biologics, including antibodies, fusion proteins, and recombinant proteins, have been causally linked with serious adverse effects in humans. In nearly all cases, these serious adverse effects have been directly associated with the immunomodulatory biologic's intended pharmacologic activity or exaggerated pharmacology. Examples of immunomodulatory biologics known to cause serious adverse effects in the clinic ranging from immunostimulation and cytokine release syndrome (e.g., TGN1412) to immunosuppression with increased risk of opportunistic infections (e.g., TNF-α antagonists, anti-integrins) are presented. Specific examples of the nonclinical testing strategy used for the clinical risk assessment of these immunomodulatory biologics are discussed, with an emphasis on the clinical relevance and predictivity of the models. Infectious challenge animal models, in particular, were critically evaluated for their utility in evaluating clinical risk assessment versus understanding mechanism of action. The nonclinical safety testing strategy for an immunomodulatory biologic should be custom tailored to interrogate the biology of the immunologic target in order to best assess potential clinical risk. This nonclinical strategy should include mechanistic and efficacy models of pharmacologic activity and immunologic signaling pathways, in vitro immunologic assays such as cytokine release, and immunophenotypic assessment by flow cytometry, immunohistochemistry, and/or immunofluorescence, as appropriate.
Collapse
|
24
|
Ragheb S, Lisak RP. B-cell-activating factor and autoimmune myasthenia gravis. Autoimmune Dis 2011; 2011:939520. [PMID: 22235365 PMCID: PMC3251912 DOI: 10.4061/2011/939520] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 10/08/2011] [Indexed: 11/26/2022] Open
Abstract
BAFF is a potent B-cell survival factor, and it plays an essential role in B-cell homeostasis and B-cell function in the periphery. Both normal and autoreactive B cells are BAFF dependent; however, excess BAFF promotes the survival, growth, and maturation of autoreactive B cells. When overexpressed, BAFF protects B cells from apoptosis, thereby contributing to autoimmunity. Three independent studies have shown higher BAFF levels in the circulation of MG patients. BAFF may play an important role in the pathogenesis of MG. BAFF antagonists may well provide new treatment options for MG patients, particularly those patients with thymic lymphoid follicular hyperplasia.
Collapse
Affiliation(s)
- Samia Ragheb
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | |
Collapse
|
25
|
Abstract
New targeted anti-inflammatory drugs have revolutionized the therapeutic strategies in rheumatology. Recombinant DNA selection technologies have enabled the isolation and humanization of specific antibody fragments of any specificity that can be 'armed' to deliver effective anti-inflammatory 'payloads'. Antibodies and other targeted provide the opportunity to block key soluble mediators of inflammation in their milieu, or alternatively to block intracellular inflammation-triggering pathways by binding to an upstream cell-surface receptor. Designed proteins can be improved with respect to desired pharmacokinetic and pharmacodynamic effects. They facilitate the delivery of the required immunosuppressive effect. However, the individual extent of desired and undesired effects of a particular biologic therapy can be broader than initially predicted and requires careful evaluation during clinical trials. This review highlights advances in the application of recombinant antibody technology for novel biologic therapies in rheumatology.
Collapse
Affiliation(s)
- Achim Rothe
- Department of Internal Medicine I, University Hospital Cologne, Joseph Stelzmann Street 9, 50931 Cologne, Germany.
| | | |
Collapse
|
26
|
|
27
|
Mihalcik SA, Tschumper RC, Jelinek DF. Transcriptional and post-transcriptional mechanisms of BAFF-receptor dysregulation in human B lineage malignancies. Cell Cycle 2010; 9:4884-92. [PMID: 21099364 DOI: 10.4161/cc.9.24.14156] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Together, circulating BAFF and dominant receptor BAFF-R homeostatically regulate the humoral immune system. Consistently aberrant BAFF-R expression in leukemic cells reveals an intimate connection of these cells' malignant physiology to the BAFF/BAFF-R axis and also provides an additional survival mechanism to the expressing cells. In this study, we used primary cells and cell lines to interrogate the mechanisms underlying aberrant BAFF-R expression in precursor B acute lymphoblastic leukemia (precursor B-ALL) and mature B chronic lymphocytic leukemia (CLL). Here we demonstrate the aberrant expression of BAFF-R in precursor B-ALL cell lines and reveal that these cells acquire BAFF-R expression through premature transcriptional activation of the BAFF-R promoter in coordination with regulatory transcription factor c-Rel. Investigations using primary CLL cells provide a crucial counterpoint through their paucity of BAFF-R relative to their benign mature B cell counterparts, which we establish as functionally significant in its depletion of the CLL cells' BAFF-binding capacity. Furthermore, BAFF-R downregulation in CLL patients is revealed here to be restricted to the malignant compartment and mediated post-transcriptionally in order to compensate for the consistently unchanged levels of transcription factor c-Rel and BAFF-R mRNA. Finally, we present evidence that CLL cells retain endogenous mechanisms of BAFF-R regulatory control despite active receptor dysregulation.
Collapse
Affiliation(s)
- Stephen A Mihalcik
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | |
Collapse
|
28
|
Abstract
The dysfunctional immune response that characterizes systemic lupus erythematosus (SLE) associates with an unbalanced production of soluble mediators that are crucial in promoting and sustaining chronic inflammation. The successful use of biologics in several autoimmune diseases has led to studies in SLE aimed at contrasting the proinflammatory responses that contribute to tissue and organ damage in the disease. Several approaches have been developed and tested as potential therapeutic agents in SLE in preclinical studies and in clinical trials. This article provides an overview on antibody-based approaches in SLE that, although preliminary, have the potential to expand the current therapeutic possibilities in the disease.
Collapse
Affiliation(s)
- Antonio La Cava
- Division of Rheumatology, Department of Medicine, University of California Los Angeles, 1000 Veteran Avenue 32-59, Los Angeles, CA 90095-1670, USA.
| |
Collapse
|
29
|
Davidson A. Targeting BAFF in autoimmunity. Curr Opin Immunol 2010; 22:732-9. [PMID: 20970975 DOI: 10.1016/j.coi.2010.09.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/27/2010] [Accepted: 09/28/2010] [Indexed: 11/18/2022]
Abstract
BAFF and APRIL are TNF-like cytokines that support survival and differentiation of B cells. The early appreciation that overexpression of BAFF leads to B cell expansion and a lupus-like syndrome in mice, and the demonstration that BAFF inhibition delays lupus onset in spontaneous mouse models of SLE and other autoimmune diseases has rapidly led to the development of strategies for inhibiting both BAFF and APRIL. The commercialization of this new class of drugs has proceeded in parallel with the continuing elucidation of the biology of the cytokines and their receptors. Recent studies have uncovered a role for BAFF in enhancing both innate and adaptive immune responses and in amplifying aberrant pathways that arise during inflammation. Two phase III studies of an anti-BAFF antibody have yielded positive, although modest, results in SLE and alternate inhibitors are being tested in a variety of autoimmune diseases in which BAFF may play a pathogenic role.
Collapse
Affiliation(s)
- Anne Davidson
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, United States.
| |
Collapse
|
30
|
Parameswaran R, Müschen M, Kim YM, Groffen J, Heisterkamp N. A functional receptor for B-cell-activating factor is expressed on human acute lymphoblastic leukemias. Cancer Res 2010; 70:4346-56. [PMID: 20460528 DOI: 10.1158/0008-5472.can-10-0300] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
B-lineage acute lymphoblastic leukemia (ALL) arises by transformation of a progenitor (pre-B) cell. Cure rates in adults remain low and treatment is complicated by support provided by the microenvironment to the leukemic cells, indicating an urgent need to better understand the factors that promote their survival. B-cell-activating factor (BAFF) and its receptor BAFF-R are important for survival and growth of mature normal and malignant B cells but are not expressed on pre-B cells. Unexpectedly, all cells in the primary Philadelphia chromosome (Ph)-positive and Ph-negative ALL samples tested were positive for high BAFF-R cell surface expression. BAFF-R was fully competent to bind BAFF, and stimulation of the receptor activated both the classic and the noncanonical NF-kappaB pathways. Recombinant BAFF supported survival of the ALL cells in the absence of stroma, and it significantly attenuated the rate of apoptosis caused by exposure to nilotinib, a drug used therapeutically to treat Ph-positive ALLs. Surprisingly, BAFF mRNA and protein were also expressed in the same cells but BAFF was not shed into the medium. Our report is the first showing universal expression of BAFF-R by pre-B ALL cells and opens the possibility of blocking its function as an adjuvant therapeutic strategy.
Collapse
Affiliation(s)
- Reshmi Parameswaran
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, Childrens Hospital Los Angeles, CA 90027, USA
| | | | | | | | | |
Collapse
|
31
|
Bermejo DA, Amezcua-Vesely MC, Montes CL, Merino MC, Gehrau RC, Cejas H, Acosta-Rodríguez EV, Gruppi A. BAFF mediates splenic B cell response and antibody production in experimental Chagas disease. PLoS Negl Trop Dis 2010; 4:e679. [PMID: 20454564 PMCID: PMC2864296 DOI: 10.1371/journal.pntd.0000679] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/24/2010] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND B cells and antibodies are involved not only in controlling the spread of blood circulating Trypanosoma cruzi, but also in the autoreactive manifestations observed in Chagas disease. Acute infection results in polyclonal B cell activation associated with hypergammaglobulinemia, delayed specific humoral immunity and high levels of non-parasite specific antibodies. Since TNF superfamily B lymphocyte Stimulator (BAFF) mediates polyclonal B cell response in vitro triggered by T. cruzi antigens, and BAFF-Tg mice show similar signs to T. cruzi infected mice, we hypothesized that BAFF can mediate polyclonal B cell response in experimental Chagas disease. METHODOLOGY/PRINCIPAL FINDINGS BAFF is produced early and persists throughout the infection. To analyze BAFF role in experimental Chagas disease, Balb/c infected mice were injected with BR3:Fc, a soluble receptor of BAFF, to block BAFF activity. By BAFF blockade we observed that this cytokine mediates the mature B cell response and the production of non-parasite specific IgM and IgG. BAFF also influences the development of antinuclear IgG and parasite-specific IgM response, not affecting T. cruzi-specific IgG and parasitemia. Interestingly, BAFF inhibition favors the parasitism in heart. CONCLUSIONS/SIGNIFICANCE Our results demonstrate, for the first time, an active role for BAFF in shaping the mature B cell repertoire in a parasite infection.
Collapse
Affiliation(s)
- Daniela A. Bermejo
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - María C. Amezcua-Vesely
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Carolina L. Montes
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - María C. Merino
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Ricardo C. Gehrau
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Hugo Cejas
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Eva V. Acosta-Rodríguez
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Adriana Gruppi
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
32
|
Swee LK, Tardivel A, Schneider P, Rolink A. Rescue of the mature B cell compartment in BAFF-deficient mice by treatment with recombinant Fc-BAFF. Immunol Lett 2010; 131:40-8. [PMID: 20350570 DOI: 10.1016/j.imlet.2010.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/21/2010] [Indexed: 11/18/2022]
Abstract
BAFF deficiency in mice impairs B cell development beyond the transitional stage 1 in the spleen and thus severely reduces the size of follicular and marginal zone B cell compartments. Moreover, humoral immune responses in these mice are dramatically impaired. We now addressed the question whether the decrease in mature B cell numbers and the reduced humoral immune responses in BAFF-deficient mice could be overcome by the injection of recombinant BAFF. We therefore engineered a recombinant protein containing the human IgG1 Fc moiety fused to receptor-binding domain of human BAFF (Fc-BAFF). At 1 week after the second injection of this fusion protein a complete rescue of the marginal zone B cell compartment and a 50% rescue of the follicular B cell compartment was observed. Moreover these mice mounted a T cell-dependent humoral immune response indistinguishable from wild-type mice. By day 14 upon arrest of Fc-BAFF treatment mature B cell numbers in the blood dropped by 50%, indicating that the life span of mature B cells in the absence of BAFF is 14 days or less. Collectively these findings demonstrate that injection of Fc-BAFF in BAFF-deficient mice results in a temporary rescue of a functional mature B cell compartment.
Collapse
Affiliation(s)
- Lee Kim Swee
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Mattenstrasse 28, Basel, Switzerland
| | | | | | | |
Collapse
|
33
|
Abstract
B cells represent an important link between the adaptive and innate immune systems as they express both antigen-specific B-cell receptors (BCRs) as well as various Toll-like receptors (TLRs). Several checkpoints in B-cell development ensure that self-specific cells are eliminated from the mature B-cell repertoire to avoid harmful autoreactive responses. These checkpoints are controlled by BCR-mediated events but are also influenced by TLR-dependent signals from the innate immune system. Additionally, B-cell-intrinsic and extrinsic TLR signaling are critical for inflammatory events required for the clearance of microbial infections. Factors secreted by TLR-activated macrophages or dendritic cells directly influence the fate of protective and autoreactive B cells. Additionally, naive and memory B cells respond differentially to TLR ligands, as do different B-cell subsets. We review here recent literature describing intrinsic and extrinsic effects of TLR stimulation on the fate of B cells, with particular attention to autoimmune diseases.
Collapse
Affiliation(s)
- Steve P Crampton
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| | | | | |
Collapse
|
34
|
Chronic lymphocytic leukemia of Emu-TCL1 transgenic mice undergoes rapid cell turnover that can be offset by extrinsic CD257 to accelerate disease progression. Blood 2009; 114:4469-76. [PMID: 19755673 DOI: 10.1182/blood-2009-06-230169] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Results of heavy-water labeling studies have challenged the notion that chronic lymphocytic leukemia (CLL) represents an accumulation of noncycling B cells. We examined leukemia cell turnover in Emu-TCL1 transgenic (TCL1-Tg) mice, which develop a CLL-like disease at 8 to 12 months of age. We found that leukemia cells in these mice not only had higher proportions of proliferating cells but also apoptotic cells than did nonleukemic lymphocytes. We crossed TCL1-Tg with BAFF-Tg mice, which express high levels of CD257. TCL1 x BAFF-Tg mice developed CLL-like disease at a significantly younger age and had more rapid disease progression and shorter survival than TCL1-Tg mice. Leukemia cells of TCL1 x BAFF-Tg mice had similar proportions of proliferating cells, but fewer proportions of dying cells, than did the CLL cells of TCL1-Tg mice. Moreover, leukemia cells from either TCL1 x BAFF-Tg or TCL1-Tg mice produced more aggressive disease when transferred into BAFF-Tg mice than into wild-type (WT) mice. Neutralization of CD257 resulted in rapid reduction in circulating leukemia cells. These results indicate that the leukemia cells of TCL1-Tg mice undergo high levels of spontaneous apoptosis that is offset by relatively high rates of leukemia cell proliferation, which might allow for acquisition of mutations that contribute to disease evolution.
Collapse
|
35
|
Rauch M, Tussiwand R, Bosco N, Rolink AG. Crucial role for BAFF-BAFF-R signaling in the survival and maintenance of mature B cells. PLoS One 2009; 4:e5456. [PMID: 19421318 PMCID: PMC2673681 DOI: 10.1371/journal.pone.0005456] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 04/07/2009] [Indexed: 12/17/2022] Open
Abstract
Defects in the expression of either BAFF (B cell activating factor) or BAFF-R impairs B cell development beyond the immature, transitional type-1 stage and thus, prevents the formation of follicular and marginal zone B cells, whereas B-1 B cells remain unaffected. The expression of BAFF-R on all mature B cells might suggest a role for BAFF-R signaling also for their in vivo maintenance. Here, we show that, 14 days following a single injection of an anti-BAFF-R mAb that prevents BAFF binding, both follicular and marginal zone B cell numbers are drastically reduced, whereas B-1 cells are not affected. Injection of control, isotype-matched but non-blocking anti-BAFF-R mAbs does not result in B cell depletion. We also show that this depletion is neither due to antibody-dependent cellular cytotoxicity nor to complement-mediated lysis. Moreover, prevention of BAFF binding leads to a decrease in the size of the B cell follicles, an impairment of a T cell dependent humoral immune response and a reduction in the formation of memory B cells. Collectively, these results establish a central role for BAFF-BAFF-R signaling in the in vivo survival and maintenance of both follicular and marginal zone B cell pools.
Collapse
Affiliation(s)
- Melanie Rauch
- Developmental and Molecular Immunology, Department of Biomedicine (DBM), University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
36
|
Cancro MP, D'Cruz DP, Khamashta MA. The role of B lymphocyte stimulator (BLyS) in systemic lupus erythematosus. J Clin Invest 2009; 119:1066-73. [PMID: 19411764 PMCID: PMC2673851 DOI: 10.1172/jci38010] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
SLE, a chronic, multisystem autoimmune disorder with a broad range of symptoms, involves defective B cell selection and elimination of self-reactive B cells. B lymphocyte stimulator (BLyS), a soluble ligand of the TNF cytokine family, is a prominent factor in B cell differentiation, homeostasis, and selection. BLyS levels affect survival signals and selective apoptosis of autoantibody-producing B cells. High levels of BLyS may relax B cell selection and contribute to autoantibody production, exacerbating the SLE disease state. This review discusses the mechanism of BLyS action on B cells, its role in SLE, and specific targeting of BLyS in the treatment of SLE.
Collapse
Affiliation(s)
- Michael P Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, 19104-6082, USA.
| | | | | |
Collapse
|
37
|
Pulsipher MA, Bader P, Klingebiel T, Cooper LJN. Allogeneic transplantation for pediatric acute lymphoblastic leukemia: the emerging role of peritransplantation minimal residual disease/chimerism monitoring and novel chemotherapeutic, molecular, and immune approaches aimed at preventing relapse. Biol Blood Marrow Transplant 2009; 15:62-71. [PMID: 19147081 DOI: 10.1016/j.bbmt.2008.11.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although improved donor sources and supportive care have decreased transplantation-related mortality over the past decade, relapse remains the principal cause of failure after allogeneic transplantation for high-risk pediatric acute lymphoblastic leukemia (ALL). Emerging tools of minimal residual disease (MRD) and chimerism monitoring before and after transplantation have defined those children at highest risk for relapse and provide the opportunity for intervention to prevent relapse. Specific methods aimed at decreasing relapse include the use of intensive treatment before transplantation to increase the percentage of patients undergoing the procedure with negative MRD, optimal transplantation preparative regimens, and posttransplantation interventions with targeted or immunologic therapy. Early data demonstrate decreased relapse with the use of sirolimus for all types of ALL and imatinib for ALL with the Philadelphia chromosome (Ph(+) ALL) after transplantation. Patients with increasing chimerism or MRD have been shown to benefit from early withdrawal of immune suppression or donor lymphocyte infusion. Finally, various targeted immunologic therapies, including monoclonal antibodies, killer cell immunoglobulin-like receptor mismatching, natural killer cell therapy, and targeted T cell therapies, are emerging that also could have an affect on relapse and improve survival after transplantation for pediatric ALL.
Collapse
Affiliation(s)
- Michael A Pulsipher
- Division of Hematology/BMT, Primary Children's Medical Center, University of Utah School of Medicine, Salt Lake City, Utah
| | | | | | | |
Collapse
|
38
|
Waldburger JM, Firestein GS. Garden of therapeutic delights: new targets in rheumatic diseases. Arthritis Res Ther 2009; 11:206. [PMID: 19232066 PMCID: PMC2688217 DOI: 10.1186/ar2556] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advances in our understanding of the cellular and molecular mechanisms in rheumatic disease fostered the advent of the targeted therapeutics era. Intense research activity continues to increase the number of potential targets at an accelerated pace. In this review, examples of promising targets and agents that are at various stages of clinical development are described. Cytokine inhibition remains at the forefront with the success of tumor necrosis factor blockers, and biologics that block interleukin-6 (IL-6), IL-17, IL-12, and IL-23 and other cytokines are on the horizon. After the success of rituximab and abatacept, other cell-targeted approaches that inhibit or deplete lymphocytes have moved forward, such as blocking BAFF/BLyS (B-cell activation factor of the tumor necrosis factor family/B-lymphocyte stimulator) and APRIL (a proliferation-inducing ligand) or suppressing T-cell activation with costimulation molecule blockers. Small-molecule inhibitors might eventually challenge the dominance of biologics in the future. In addition to plasma membrane G protein-coupled chemokine receptors, small molecules can be designed to block intracellular enzymes that control signaling pathways. Inhibitors of tyrosine kinases expressed in lymphocytes, such as spleen tyrosine kinase and Janus kinase, are being tested in autoimmune diseases. Inactivation of the more broadly expressed mitogen-activated protein kinases could suppress inflammation driven by macrophages and mesenchymal cells. Targeting tyrosine kinases downstream of growth factor receptors might also reduce fibrosis in conditions like systemic sclerosis. The abundance of potential targets suggests that new and creative ways of evaluating safety and efficacy are needed.
Collapse
Affiliation(s)
- Jean M Waldburger
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego School of Medicine, Mail Code 0656, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | |
Collapse
|
39
|
Targeting of BAFF and APRIL for Autoimmunity and Oncology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:52-63. [DOI: 10.1007/978-0-387-89520-8_4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
The impact of Fc engineering on an anti-CD19 antibody: increased Fcgamma receptor affinity enhances B-cell clearing in nonhuman primates. Blood 2008; 113:3735-43. [PMID: 19109559 DOI: 10.1182/blood-2008-10-182048] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
CD19, a B cell-restricted receptor critical for B-cell development, is expressed in most B-cell malignancies. The Fc-engineered anti-CD19 antibody, XmAb5574, has enhanced Fcgamma receptor (FcgammaR) binding affinity, leading to improved FcgammaR-dependent effector cell functions and antitumor activity in murine xenografts compared with the non-Fc-engineered anti-CD19 IgG1 analog. Here, we use XmAb5574 and anti-CD19 IgG1 to further dissect effector cell functions in an immune system closely homologous to that of humans, the cynomolgus monkey. XmAb5574 infusion caused an immediate and dose-related B-cell depletion in the blood (to <10% of baseline levels) concomitant with a sustained reduction of natural killer (NK) cells. NK cells had fully recovered by day 15, whereas B-cell recovery was underway by day 57. B cells in secondary lymphoid tissues were depleted (to 34%-61% of vehicle), with involuted germinal centers apparent in the spleen. Anti-CD19 IgG1 had comparable serum exposure to XmAb5574 but demonstrated no B-cell depletion and no sustained NK-cell reduction. Thus, increasing FcgammaR binding affinity dramatically increased B-cell clearing. We propose that effector cell functions, possibly those involving NK cells, mediate XmAb5574 potency in cynomolgus monkeys, and that enhancing these mechanisms should advance the treatment of B-cell malignancies in humans.
Collapse
|
41
|
Therapeutic advances in rheumatology with the use of recombinant proteins. ACTA ACUST UNITED AC 2008; 4:605-14. [PMID: 18813220 DOI: 10.1038/ncprheum0909] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 08/11/2008] [Indexed: 12/27/2022]
Abstract
Antibody engineering and protein design have led to the creation of a new era of targeted anti-inflammatory therapies in rheumatology. Recombinant DNA technologies have enabled the selection and humanization of specific antibody fragments in order to develop therapeutic reagents of any specificity that can be 'armed' to deliver effective anti-inflammatory 'payloads'. Antibodies and antibody-like proteins provide the opportunity to block key soluble mediators of inflammation in their milieu, or alternatively to block intracellular inflammation-triggering pathways by binding to an upstream cell-surface receptor. These designer proteins can be tuned for desired pharmacokinetic and pharmacodynamic effects, and represent tools for specific therapeutic intervention by delivering precisely the required immunosuppressive effect. The extent of desired and undesired effects of a particular biologic therapy, however, can be broader than initially predicted and require careful evaluation during clinical trials. This Review highlights advances in recombinant technologies for the development of novel biologic therapies in rheumatology.
Collapse
|
42
|
Stefanich EG, Ren S, Danilenko DM, Lim A, Song A, Iyer S, Fielder PJ. Evidence for an asialoglycoprotein receptor on nonparenchymal cells for O-linked glycoproteins. J Pharmacol Exp Ther 2008; 327:308-15. [PMID: 18728239 DOI: 10.1124/jpet.108.142232] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
B cell-activating factor receptor 3 (BR3)-Fc is an IgG1-receptor dimeric fusion protein that has multiple O-linked glycosylation sites and sialylation levels that can vary in the manufacturing process. Increased sialic acid levels resulted from increased site occupancy with the O-linked N-acetylgalactosamine (GalNAc-Gal), but because the ratio of sialic acid per mole of oligosaccharide remained approximately 1, this led to increased asialo terminal GalNAc. Previous studies have demonstrated an effect of terminal asialo Gal or GalNAc on the clearance of glycoproteins due to uptake and degradation by lectin receptors in the liver. However, the previous studies examined N-linked oligosaccharides, and there are less data regarding O-linked oligosaccharides. The objective of these studies was to determine the effects on the pharmacokinetics and distribution of the asialo terminal GalNAc and varying amounts of sialic acid residues on BR3-Fc. The results of the data presented here suggest that exposed Gal on the desialylated BR3-Fc led to rapid clearance due to uptake and degradation in the liver that was associated with nonparenchymal cells. It is interesting to note that the data indicated a decreased clearance and increased exposure of BR3-Fc as the sialic acid levels increased, even though increased sialic acid was associated with increased asialo GalNAc. Therefore, the exposed GalNAc did not seem to play a role in the clearance of BR3-Fc; although the Gal linked to the hydroxyl group at position 3 may have prevented an interaction. Because we did not see uptake of desialylated BR3-Fc in hepatocytes where the asialoglycoprotein receptor is localized, this nonparenchymal cell lectin may have preference for O-linked glycoproteins.
Collapse
Affiliation(s)
- Eric G Stefanich
- Department of Pharmacokinetic and Pharmacodynamic Sciences, Genentech, Inc., South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Systemic lupus erythematosus (SLE) is a complex immune disorder in which loss of tolerance to nucleic acid antigens and other crossreactive antigens is associated with the development of pathogenic autoantibodies that damage target organs, including the skin, joints, brain and kidney. New drugs based on modulation of the immune system are currently being developed for the treatment of SLE. Many of these new therapies do not globally suppress the immune system but target specific activation pathways relevant to SLE pathogenesis. Immune modulation in SLE is complicated by differences in the immune defects between patients and at different disease stages. Since both deficiency and hyperactivity of the immune system can give rise to SLE, the ultimate goal for SLE therapy is to restore homeostasis without affecting protective immune responses to pathogens. Here we review recent immunological advances that have enhanced our understanding of SLE pathogenesis and discuss how they may lead to the development of new treatment regimens.
Collapse
|