1
|
Kellermann G, Leulliot N, Cherfils-Vicini J, Blaud M, Brest P. Activated B-Cells enhance epitope spreading to support successful cancer immunotherapy. Front Immunol 2024; 15:1382236. [PMID: 38571942 PMCID: PMC10989059 DOI: 10.3389/fimmu.2024.1382236] [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: 02/05/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Immune checkpoint therapies (ICT) have transformed the treatment of cancer over the past decade. However, many patients do not respond or suffer relapses. Successful immunotherapy requires epitope spreading, but the slow or inefficient induction of functional antitumoral immunity delays the benefit to patients or causes resistances. Therefore, understanding the key mechanisms that support epitope spreading is essential to improve immunotherapy. In this review, we highlight the major role played by B-cells in breaking immune tolerance by epitope spreading. Activated B-cells are key Antigen-Presenting Cells (APC) that diversify the T-cell response against self-antigens, such as ribonucleoproteins, in autoimmunity but also during successful cancer immunotherapy. This has important implications for the design of future cancer vaccines.
Collapse
Affiliation(s)
| | - Nicolas Leulliot
- Université Paris Cité, Centre national de la recherche scientifique (CNRS), Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), Paris, France
| | - Julien Cherfils-Vicini
- Université Côte d’Azur, Institute for Research on Cancer and Aging, Nice (IRCAN), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), Centre Antoine Lacassagne, Institut Hospitalo-Universitaire (IHU), RESPIRera, Fédérations Hospitalo-Universitaires (FHU)OncoAge, Nice, France
| | - Magali Blaud
- Université Paris Cité, Centre national de la recherche scientifique (CNRS), Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), Paris, France
| | - Patrick Brest
- Université Côte d’Azur, Institute for Research on Cancer and Aging, Nice (IRCAN), Centre national de la recherche scientifique (CNRS), Institut national de la santé et de la recherche médicale (INSERM), Centre Antoine Lacassagne, Institut Hospitalo-Universitaire (IHU), RESPIRera, Fédérations Hospitalo-Universitaires (FHU)OncoAge, Nice, France
| |
Collapse
|
2
|
Zhang J, Tsukui T, Wu X, Brito A, Trumble JM, Caraballo JC, Allen GM, Zavala-Solorio J, Zhang C, Paw J, Lim WA, Geng J, Kutskova Y, Freund A, Kolumam G, Sheppard D, Cohen RL. An immune-based tool platform for in vivo cell clearance. Life Sci Alliance 2023; 6:e202201869. [PMID: 37311583 PMCID: PMC10264967 DOI: 10.26508/lsa.202201869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
Immunological targeting of pathological cells has been successful in oncology and is expanding to other pathobiological contexts. Here, we present a flexible platform that allows labeling cells of interest with the surface-expressed model antigen ovalbumin (OVA), which can be eliminated via either antigen-specific T cells or newly developed OVA antibodies. We demonstrate that hepatocytes can be effectively targeted by either modality. In contrast, pro-fibrotic fibroblasts associated with pulmonary fibrosis are only eliminated by T cells in initial experiments, which reduced collagen deposition in a fibrosis model. This new experimental platform will facilitate development of immune-based approaches to clear potential pathological cell types in vivo.
Collapse
Affiliation(s)
| | - Tatsuya Tsukui
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Xiumin Wu
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | | | - Juan C Caraballo
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Greg M Allen
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Jonathan Paw
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Wendell A Lim
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Cell Design Institute, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Adam Freund
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | - Dean Sheppard
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
3
|
Fu T, Zhang L, Zuo M, Li F, Shi C, Chen H. FCGR2A as one novel potential target for poor survival prognosis of clear cell renal cell carcinoma. Medicine (Baltimore) 2023; 102:e33324. [PMID: 36930102 PMCID: PMC10019103 DOI: 10.1097/md.0000000000033324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cell carcinoma. Immunoglobulin FcγRIIa receptor (FCGR2A) has been implicated in various cancers, however, its role on ccRCC is not well studied. A total of 151 patients with ccRCC were recruited for the study. Cox proportional hazards regression analysis was performed to calculate the hazard radios of FCGR2A expression and tumor characteristics. Pathological changes associated with ccRCC in tumor tissue sections were analyzed by hematoxylin-eosin staining. Immunohistochemical and immunofluorescence staining were used to detect the protein expression of FCGR2A in the tissue sections. Correlation between the expression of FCGR2A and the overall survival (OS) of ccRCC patients was analyzed by biological process neural network and support vector machine. The expression of FCGR2A was significantly correlated with the TNM of tumor, family history of ccRCC and Fuhrman stage of ccRCC. Patients with high FCGR2A expression in the tumor tissue, had poorer OS than the patients with low and moderate FCGR2A expression. The Receiver operating characteristic curve showed that FCGR2A can be used as a sensitive and specific biomarker for the diagnosis of ccRCC. Western blotting revealed that the FCGR2A was expressed at higher levels in the ccRCC tissues. Biological process neural network and support vector machine fitting showed that the R2 between FCGR2A and survival time of ccRCC patients was 0.8429 and 0.7669, respectively. FCGR2A is highly expressed in ccRCC, higher expression of FCGR2A is associated with poorer OS of ccRCC.
Collapse
Affiliation(s)
- Taozhu Fu
- Department of Urology, China Aerospace Science & Industry Corporation 731 Hospital, Feng Tai District, Beijing, China
| | - Lianfeng Zhang
- Department of Urology, China Aerospace Science & Industry Corporation 731 Hospital, Feng Tai District, Beijing, China
| | - Meini Zuo
- Department of Urology, China Aerospace Science & Industry Corporation 731 Hospital, Feng Tai District, Beijing, China
| | - Feng Li
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Changjin Shi
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Hongrun Chen
- Department of Urology, China Aerospace Science & Industry Corporation 731 Hospital, Feng Tai District, Beijing, China
| |
Collapse
|
4
|
Drouin M, Saenz J, Gauttier V, Evrard B, Teppaz G, Pengam S, Mary C, Desselle A, Thepenier V, Wilhelm E, Merieau E, Ligeron C, Girault I, Lopez MD, Fourgeux C, Sinha D, Baccelli I, Moreau A, Louvet C, Josien R, Poschmann J, Poirier N, Chiffoleau E. CLEC-1 is a death sensor that limits antigen cross-presentation by dendritic cells and represents a target for cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eabo7621. [PMID: 36399563 PMCID: PMC9674301 DOI: 10.1126/sciadv.abo7621] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Tumors exploit numerous immune checkpoints, including those deployed by myeloid cells to curtail antitumor immunity. Here, we show that the C-type lectin receptor CLEC-1 expressed by myeloid cells senses dead cells killed by programmed necrosis. Moreover, we identified Tripartite Motif Containing 21 (TRIM21) as an endogenous ligand overexpressed in various cancers. We observed that the combination of CLEC-1 blockade with chemotherapy prolonged mouse survival in tumor models. Loss of CLEC-1 reduced the accumulation of immunosuppressive myeloid cells in tumors and invigorated the activation state of dendritic cells (DCs), thereby increasing T cell responses. Mechanistically, we found that the absence of CLEC-1 increased the cross-presentation of dead cell-associated antigens by conventional type-1 DCs. We identified antihuman CLEC-1 antagonist antibodies able to enhance antitumor immunity in CLEC-1 humanized mice. Together, our results demonstrate that CLEC-1 acts as an immune checkpoint in myeloid cells and support CLEC-1 as a novel target for cancer immunotherapy.
Collapse
Affiliation(s)
- Marion Drouin
- OSE Immunotherapeutics, Nantes, France
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Javier Saenz
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Berangere Evrard
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | | | | | | | | | | | - Emmanuel Merieau
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Camille Ligeron
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Maria-Dolores Lopez
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Cynthia Fourgeux
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Debajyoti Sinha
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Aurelie Moreau
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Cedric Louvet
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | - Regis Josien
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
- CHU Nantes, Nantes Université, Laboratoire d’Immunologie, CIMNA, Nantes, France
| | - Jeremie Poschmann
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
| | | | - Elise Chiffoleau
- Nantes Université, INSERM, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France
- Corresponding author.
| |
Collapse
|
5
|
Abstract
Follicular helper T (TFH) cells provide help to B cells, supporting the formation of germinal centres that allow affinity maturation of antibody responses. Although usually located in secondary lymphoid organs, T cells bearing features of TFH cells can also be identified in human blood, and their frequency and phenotype are often altered in people with autoimmune diseases. In this Perspective article, I discuss the increase in circulating TFH cells seen in autoimmune settings and explore potential explanations for this phenomenon. I consider the multistep regulation of TFH cell differentiation by the CTLA4 and IL-2 pathways as well as by regulatory T cells and highlight that these same pathways are crucial for regulating autoimmune diseases. The propensity of infection to serve as a cue for TFH cell differentiation and a potential trigger for autoimmune disease development is also discussed. Overall, I postulate that alterations in pathways that regulate autoimmunity are coupled to alterations in TFH cell homeostasis, suggesting that this population may serve as a core sentinel of dysregulated immunity.
Collapse
|
6
|
Differences of Circulating CD25hi Bregs and Their Correlations with CD4 Effector and Regulatory T Cells in Autoantibody-Positive T1D Compared with Age-Matched Healthy Individuals. J Immunol Res 2022; 2022:2269237. [PMID: 35083339 PMCID: PMC8786465 DOI: 10.1155/2022/2269237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/23/2021] [Accepted: 12/11/2021] [Indexed: 11/17/2022] Open
Abstract
Circulating CD25hi B cells, a subset of regulatory B cells in humans, are closely related to inflammation and autoimmune diseases. This study is aimed at investigating the alternation of CD25hi Bregs and their correlation with CD4 effector and regulatory T cells in T1D individuals. We included 68 autoantibody-positive T1D and 68 age-matched healthy individuals with peripheral blood mononuclear cells (PBMCs) and assessed them with CD25hi Bregs and CD4 effector or regulatory T cells by flow cytometry. Here, we demonstrate that the frequency of CD25hi Bregs was significantly decreased in T1D subjects (P = 0.0016), but they were not affected by disease status (age at T1D diagnosis or duration) or T1D risk loci (rs2104286 or rs12251307) in IL2RA (all P > 0.05). Moreover, higher IgD (P = 0.043) and lower CD27 (P = 0.0003) expression was observed in CD25hi Bregs of T1D individuals, but not the expression of IgM, CD24, or CD38 (all P > 0.05). Although there was no correlation between CD25hi Bregs and CD4 effector T cell subsets in either T1D or healthy individuals (all P > 0.05), we found a positive correlation between CD25hi Bregs and CD4 Tregs in healthy controls (Sp. r = 0.3544, P = 0.0249), which disappeared in T1D subjects (Sp. r = 0.137, P = 0.401). In conclusion, our results suggest that decreased CD25hi Bregs and alternation of their phenotypes are features of T1D regardless of disease duration and T1D genetic risk loci, and an impaired balance between CD25hi Bregs and CD4 Tregs might contribute to the pathogenesis of T1D.
Collapse
|
7
|
Wang J, Yang J, Kopeček J. Nanomedicines in B cell-targeting therapies. Acta Biomater 2022; 137:1-19. [PMID: 34687954 PMCID: PMC8678319 DOI: 10.1016/j.actbio.2021.10.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
B cells play multiple roles in immune responses related to autoimmune diseases as well as different types of cancers. As such, strategies focused on B cell targeting attracted wide interest and developed intensively. There are several common mechanisms various B cell targeting therapies have relied on, including direct B cell depletion, modulation of B cell antigen receptor (BCR) signaling, targeting B cell survival factors, targeting the B cell and T cell costimulation, and immune checkpoint blockade. Nanocarriers, used as drug delivery vehicles, possess numerous advantages to low molecular weight drugs, reducing drug toxicity, enhancing blood circulation time, as well as augmenting targeting efficacy and improving therapeutic effect. Herein, we review the commonly used targets involved in B cell targeting approaches and the utilization of various nanocarriers as B cell-targeted delivery vehicles. STATEMENT OF SIGNIFICANCE: As B cells are engaged significantly in the development of many kinds of diseases, utilization of nanomedicines in B cell depletion therapies have been rapidly developed. Although numerous studies focused on B cell targeting have already been done, there are still various potential receptors awaiting further investigation. This review summarizes the most relevant studies that utilized nanotechnologies associated with different B cell depletion approaches, providing a useful tool for selection of receptors, agents and/or nanocarriers matching specific diseases. Along with uncovering new targets in the function map of B cells, there will be a growing number of candidates that can benefit from nanoscale drug delivery.
Collapse
Affiliation(s)
- Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
8
|
Singh A, Behl T, Sehgal A, Singh S, Sharma N, Naved T, Bhatia S, Al-Harrasi A, Chakrabarti P, Aleya L, Vargas-De-La-Cruz C, Bungau S. Mechanistic insights into the role of B cells in rheumatoid arthritis. Int Immunopharmacol 2021; 99:108078. [PMID: 34426116 DOI: 10.1016/j.intimp.2021.108078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease epitomized by severe inflammation that induces tendon, cartilage, and bone damage over time. Although different types of cells undertake pathogenic functions in RA, the B cell's significant involvement has increasingly been known following the development of rheumatoid factor and it has been re-emphasized in recent years. Therefore, the rheumatoid factors and anti-cyclic citrullinated peptide antibodies are well-known indications of infection and clinical manifestations, and that they can precede the development of illness by several years. The emergence of rituximab a B cell reducing chimeric antidote in 1997 and 1998 transformed B-cell-targeted therapy for inflammatory disorder from a research hypothesis to a functional fact. Ever since, several autoantibody-related conditions were addressed, including the more intriguing indications of effectiveness seen in rheumatoid arthritis patients. Numerous types of B-cell-targeted compounds are currently being researched. From the beginning, one of the primary goals of B-cell therapy was to reinstate some kind of immune tolerance. While B cells have long been recognized as essential autoantibody producers, certain antibody-independent functions and usefulness as a key targeted therapy were not recognized until recently. The knowledge of B cells' diverse physical and pathogenic roles in autoimmune diseases is growing. As a result, the number of successful agents targeting the B cell complex is becoming more ubiquitous. Therefore, in this article, we explore fresh perspectives upon the roles of B cells in arthritis treatment, as well as new evidence regarding the effectiveness of B lymphocytes reduction and the therapeutic outcome of biological markers.
Collapse
Affiliation(s)
- Anuja Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tanveer Naved
- Amity Institute of Pharmacy, Amity University, Noida, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | | | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, France
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Ensenanza e Investigacion en Bacteriologia Alimentaria, Universidad Nacinol Mayor de San Marcos, Lima, Peru; E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, Peru
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
9
|
Thomas R, Oh J, Wang W, Su DM. Thymic atrophy creates holes in Treg-mediated immuno-regulation via impairment of an antigen-specific clone. Immunology 2021; 163:478-492. [PMID: 33786850 DOI: 10.1111/imm.13333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/14/2022] Open
Abstract
Age-related thymic atrophy results in reduced output of naïve conventional T (Tcon) cells. However, its impact on regulatory T (Treg) cells is insufficiently understood. Given evidence that thymic Treg (tTreg) cell generation is enhanced in the aged, atrophy thymus and that the aged periphery accumulates peripheral Treg (pTreg) cells, we asked why these Treg cells are unable to effectively attenuate increased autoreactivity-induced chronic inflammation in the elderly. We designed a mock-self-antigen chimera mouse model, in which membrane-bound ovalbumin (mOVA) transgenic mice, bearing a FoxN1-floxed gene for induction of conditional thymic atrophy, received OVA-specific (OT-II) T-cell receptor (TCR) transgenic progenitor cells. The chimeric mice with thymic atrophy exhibited a significant decrease in OVA-specific tTreg and pTreg cells but not polyclonal (pan)-Treg cells. These OVA-specific pTreg cells were significantly less able to suppress OVA-specific stimulation-induced proliferation in vitro and exhibited lower FoxP3 expression. Additionally, we conducted preliminary TCR repertoire diversity sequencing for Treg cells among recent thymic emigrants (RTEs) from RagGFP -FoxP3RFP dual-reporter mice and observed a trend for decreased diversity in mice with thymic atrophy compared to littermates with normal thymus. These data indicate that although the effects of age-related thymic atrophy do not affect pan-Treg generation, certain tissue-specific Treg clones may experience abnormal agonist selection. This, combined with enhanced pan-pTreg cells, may greatly contribute to age-related chronic inflammation, even in the absence of acute autoimmune disease in the elderly.
Collapse
Affiliation(s)
- Rachel Thomas
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Jiyoung Oh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weikan Wang
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Dong-Ming Su
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
10
|
Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 1:28-37. [PMID: 22720209 DOI: 10.4161/onci.1.1.17938] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Since the advent of hybridoma technology, dating back to 1975, monoclonal antibodies have become an irreplaceable diagnostic and therapeutic tool for a wide array of human diseases. During the last 15 years, several monoclonal antibodies (mAbs) have been approved by FDA for cancer therapy. These mAbs are designed to (1) activate the immune system against tumor cells, (2) inhibit cancer cell-intrinsic signaling pathways, (3) bring toxins in the close proximity of cancer cells, or (4) interfere with the tumor-stroma interaction. More recently, major efforts have been made for the development of immunostimulatory mAbs that either enhance cancer-directed immune responses or limit tumor- (or therapy-) driven immunosuppression. Some of these antibodies, which are thought to facilitate tumor eradication by initiating or sustaining a tumor-specific immune response, have already entered clinical trials. In this Trial Watch, we will review and discuss the clinical progress of the most important mAbs that are have entered clinical trials after January 2008.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- INSERM, U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Xiao Y, Deng C, Zhou Z. The Multiple Roles of B Lymphocytes in the Onset and Treatment of Type 1 Diabetes: Interactions between B Lymphocytes and T Cells. J Diabetes Res 2021; 2021:6581213. [PMID: 34778464 PMCID: PMC8580688 DOI: 10.1155/2021/6581213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/21/2021] [Indexed: 01/10/2023] Open
Abstract
Although type 1 diabetes is thought to be an organ-specific autoimmune disease, mediated by effective CD4+ and CD8+ T cells, it has recently become clear that B cells participate in the initiation and progress of this disease. Indeed, B cell deletion can prevent or reverse autoimmune diabetes in nonobese diabetic mice and even result in partially remaining β cell function in patients with new-onset type 1 diabetes. This review summarizes the dual role of B cells in this process not only of pathogenic effect but also of immunoregulatory function in type 1 diabetes. We focus on the impact that B cells have on regulating the activation, proliferation, and cytokine production of self-reactive T cells along with regulatory T cells, with the aim of providing a better understanding of the interactions between T and B cells in immunopathogenesis and improving the efficacy of interventions for clinical practice.
Collapse
Affiliation(s)
- Yangfan Xiao
- Clinical Nursing Teaching and Research Section, Department of Anesthesiology, and Anesthesia Medical Research Center, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Chao Deng
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, and Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, and Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
12
|
Sasaki T, Yajima T, Shimaoka T, Ogawa S, Saito T, Yamaoka K, Takeuchi T, Kubo M. Synergistic effect of IgG4 antibody and CTLs causes tissue inflammation in IgG4-related disease. Int Immunol 2020; 32:163-174. [PMID: 31713611 DOI: 10.1093/intimm/dxz073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/09/2019] [Indexed: 12/12/2022] Open
Abstract
IgG4-related disease (IgG4-RD) is characterized by multi-organ irreversible damage resulting from tissue-specific infiltration of IgG4+ plasma cells and cytotoxic T lymphocytes (CTLs). However, whether IgG4 antibody contributes to the inflammation remains unclear. In this study, we established a mouse model that enabled us to evaluate the pathogenic function of IgG4 antibodies in response to a tissue-specific autoantigen using recombinant ovalbumin (OVA)-specific human IgG4 monoclonal antibody (rOVA-hIgG4 mAb) and the mice expressing OVA of the pancreatic islets (RIP-mOVA mice). We found no inflammatory effect of rOVA-hIgG4 mAb transfer alone; however, co-transfer with OVA-specific CD8 CTLs (OT-I T cells) induced tissue damage with dense lymphocytic inflammation in the pancreas of RIP-mOVA mice. rOVA-hIgG4 mAb caused accumulation of conventional DC1 cells (cDC1s) in the lymphoid tissues, and the dendritic cells (DCs) activated the OT-I T cells via cross-presentation. We also revealed that the synergistic effects of CTLs and antibodies were observed in the other subclasses including endogenous antibodies if they recognized the same antigen. The transfer of OVA-specific CD4 helper T cells (OT-II T cells) into RIP-mOVA mice induced the production of anti-OVA antibody, which had a synergistic effect, through acquisition of a T follicular helper (TFH) phenotype. Moreover, using OT-II T cells deficient in Bcl6 caused lower anti-OVA antibody production and inflammation with OT-I T cells. Our results indicated that autoreactive IgG4 antibodies play an important role of the tissue-specific CTL response in IgG4-RD.
Collapse
Affiliation(s)
- Takanori Sasaki
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Chiba, Japan.,Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Kanagawa, Japan
| | - Taiki Yajima
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Chiba, Japan
| | - Tatsuro Shimaoka
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Chiba, Japan
| | - Shuhei Ogawa
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Chiba, Japan
| | - Takashi Saito
- Laboratory for Cell Signaling, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Kanagawa, Japan
| | - Kunihiro Yamaoka
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Chiba, Japan.,Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Kanagawa, Japan
| |
Collapse
|
13
|
Boccia R, Cooper N, Ghanima W, Boxer MA, Hill QA, Sholzberg M, Tarantino MD, Todd LK, Tong S, Bussel JB. Fostamatinib is an effective second-line therapy in patients with immune thrombocytopenia. Br J Haematol 2020; 190:933-938. [PMID: 33439486 PMCID: PMC7540289 DOI: 10.1111/bjh.16959] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022]
Abstract
Fostamatinib demonstrated efficacy in phase 3 trials of adults with immune thrombocytopenia (ITP). Post hoc analysis compared patients who received fostamatinib as second‐line therapy (after steroids ± immunoglobulins) versus third‐or‐later‐line therapy (after ≥2 prior lines of therapy including a second‐line agent). Platelet responses ≥50 000/µl were observed in 25/32 (78%) second‐line and 54/113 (48%) third‐or‐later‐line patients. Bleeding events were less frequent in second‐line (28%) versus third‐or‐later‐line (45%) patients. Responses once achieved tended to be durable in both groups. The safety profile was similar in both groups. In this post hoc analysis, fostamatinib was more effective as second‐line than third‐or‐later‐line therapy for ITP.
Collapse
Affiliation(s)
- Ralph Boccia
- Centre for Cancer and Blood Disorders, Bethesda, MD, USA
| | - Nichola Cooper
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Waleed Ghanima
- Department of Research, Østfold Hospital Trust, Sarpsborg, Norway.,Department of Hematology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Quentin A Hill
- Department of Haematology, St. James University Hospital, Leeds, UK
| | | | - Michael D Tarantino
- The Bleeding and Clotting Disorders Institute, University of Illinois College of Medicine-Peoria, Peoria, IL, USA
| | - Leslie K Todd
- Rigel Pharmaceuticals, Inc, South San Francisco, CA, USA
| | - Sandra Tong
- Rigel Pharmaceuticals, Inc, South San Francisco, CA, USA
| | - James B Bussel
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
14
|
Shao F, Zheng P, Yu D, Zhou Z, Jia L. Follicular helper T cells in type 1 diabetes. FASEB J 2019; 34:30-40. [PMID: 31914661 DOI: 10.1096/fj.201901637r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/09/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Feng Shao
- Department of Metabolism & Endocrinology The Second Xiangya HospitalCentral South University Changsha China
- Key Laboratory of Diabetes Immunology Central South University, Ministry of Education, National Clinical Research Center for Metabolic Diseases Changsha China
| | - Peilin Zheng
- Department of Endocrinology, Shenzhen People’s Hospital The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology Shenzhen China
| | - Di Yu
- The University of Queensland Diamantina Institute, Translational Research Institute Brisbane Queensland Australia
- Shandong Analysis and Test Center Shandong Academy of Sciences Jinan China
- China‐Australia Centre for Personalised Immunology Shanghai Renji Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| | - Zhiguang Zhou
- Department of Metabolism & Endocrinology The Second Xiangya HospitalCentral South University Changsha China
- Key Laboratory of Diabetes Immunology Central South University, Ministry of Education, National Clinical Research Center for Metabolic Diseases Changsha China
| | - Lijing Jia
- Department of Endocrinology, Shenzhen People’s Hospital The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology Shenzhen China
| |
Collapse
|
15
|
Synergistic cancer immunotherapy combines MVA-CD40L induced innate and adaptive immunity with tumor targeting antibodies. Nat Commun 2019; 10:5041. [PMID: 31695037 PMCID: PMC6834557 DOI: 10.1038/s41467-019-12998-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
Virus-based vaccines and appropriate costimulation potently enhance antigen-specific T cell immunity against cancer. Here we report the use of recombinant modified vaccinia virus Ankara (rMVA) encoding costimulatory CD40L against solid tumors. Therapeutic treatment with rMVA-CD40L-expressing tumor-associated antigens results in the control of established tumors. The expansion of tumor-specific cytotoxic CD8+ T cells is essential for the therapeutic antitumor effects. Strikingly, rMVA-CD40L also induces strong natural killer (NK) cell activation and expansion. Moreover, the combination of rMVA-CD40L and tumor-targeting antibodies results in increased therapeutic antitumor efficacy relying on the presence of Fc receptor and NK cells. We describe a translationally relevant therapeutic synergy between systemic viral vaccination and CD40L costimulation. We show strengthened antitumor immune responses when both rMVA-CD40L-induced innate and adaptive immune mechanisms are exploited by combination with tumor-targeting antibodies. This immunotherapeutic approach could translate into clinical cancer therapies where tumor-targeting antibodies are employed. CD40 agonists have been investigated as a strategy to awaken the immune system against cancers. Here, the authors use a virus encoding CD40L and tumour-associated antigens to enhance innate and adaptive immunity that together with tumour targeting antibodies controls the growth of tumours in mice.
Collapse
|
16
|
Pinto IB, dos Santos Machado L, Meneguetti BT, Nogueira ML, Espínola Carvalho CM, Roel AR, Franco OL. Utilization of antimicrobial peptides, analogues and mimics in creating antimicrobial surfaces and bio-materials. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.107237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
17
|
Liu C, Whitener RL, Lin A, Xu Y, Chen J, Savinov A, Leiding JW, Wallet MA, Mathews CE. Neutrophil Cytosolic Factor 1 in Dendritic Cells Promotes Autoreactive CD8 + T Cell Activation via Cross-Presentation in Type 1 Diabetes. Front Immunol 2019; 10:952. [PMID: 31118934 PMCID: PMC6504685 DOI: 10.3389/fimmu.2019.00952] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 04/12/2019] [Indexed: 12/30/2022] Open
Abstract
Aims: Reactive oxygen species (ROS) are critical in driving the onset of type 1 diabetes (T1D). Ablation of ROS derived from phagocytic NADPH oxidase 2 is protective against autoimmune diabetes in non-obese diabetic (NOD) mice. However, the mechanisms of NADPH oxidase 2-derived ROS in T1D pathogenesis need to be elucidated. Here, we have examined the role of Ncf1 (the regulatory subunit of NADPH oxidase 2) in dendritic cells (DC). Results:Ncf1-mutant DCs exhibit reduced ability to activate autoreactive CD8+ T cells despite no difference in co-stimulatory molecule expression or pro-inflammatory cytokine production. When provided with exogenous whole-protein antigen, Ncf1-mutant NOD DCs showed strong phagosome acidification and rapid antigen degradation, which lead to an absence of protein translocation into the cytoplasm and deficient antigenic peptide loading on MHC Class I molecules. Innovation: This study demonstrates that Ncf1 (p47phox) is required for activation and effector function of CD8+ T cells by acting both intrinsically within the T cell as well as within professional antigen presenting cells. Conclusion: ROS promote CD8+ T cell activation by facilitating autoantigen cross-presentation by DCs. ROS scavengers could potentially represent an important component of therapies aiming to disrupt autoantigen presentation and activation of CD8+ T cells in individuals at-risk for developing T1D.
Collapse
Affiliation(s)
- Chao Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Robert L Whitener
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Andrea Lin
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Yuan Xu
- Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Jing Chen
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Alexei Savinov
- Children's Health Research Center, Sanford Research, Sioux Falls, SD, United States
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins-All Children's Hospital, University of South Florida, St. Petersburg, FL, United States
| | - Mark A Wallet
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| |
Collapse
|
18
|
Abstract
Antibody/antigen binding results in immune complexes (IC) that have a variety of regulatory functions. One important feature is the enhanced host immune activation against antigen contained in the complex. ICs play important roles at several critical steps that lead to B and T cell activation, including antigen targeting/retention, facilitated antigen uptake, antigen presenting cell activation and proper balancing of positive and negative stimulatory signals. In both poultry industry and clinical health care, ICs have been used as preventive and therapeutic vaccines. With our deepening understanding of antibody biology, particularly in light of new revelations of regulatory functions of Fc receptors, mechanistically more precise engineering has spearheaded tailored use of this tool for infection control and cancer therapy. IC-based treatment and prophylaxis have been tested to different extents in HBV, HIV and influenza viral infection control and are actively examined as an alternative treatment for several forms of tumor. As a part of this book series, this chapter aims to discuss the mechanistic aspects of IC signaling and their impact on immune cells. We give samples how this old technology has been used by practitioners over the last several decades and suggest potential paths for future development of IC-based immune therapy.
Collapse
Affiliation(s)
- Yu-Mei Wen
- Key Laboratory of Molecular Virology, Shanghai Medical College, School of Basic Medical Sciences, Fudan University, Shanghai, China. .,Shanghai Medical College, Fudan University, Rm 401, Fuxing Bldg, 131 Yi Xue Yuan Rd, Shanghai, 200032, China.
| | - Yan Shi
- Department of Basic Medical Sciences, Center for Life Sciences, Institute of Immunology, Tsinghua University, Beijing, China.,Department of Microbiology, Immunology & Infectious Diseases and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.,D301 Medical Sciences Bldg, Tsinghua University, Beijing, 00084, China
| |
Collapse
|
19
|
Manfredi AA, Ramirez GA, Rovere-Querini P, Maugeri N. The Neutrophil's Choice: Phagocytose vs Make Neutrophil Extracellular Traps. Front Immunol 2018. [PMID: 29515586 PMCID: PMC5826238 DOI: 10.3389/fimmu.2018.00288] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophils recognize particulate substrates of microbial or endogenous origin and react by sequestering the cargo via phagocytosis or by releasing neutrophil extracellular traps (NETs) outside the cell, thus modifying and alerting the environment and bystander leukocytes. The signals that determine the choice between phagocytosis and the generation of NETs are still poorly characterized. Neutrophils that had phagocytosed bulky particulate substrates, such as apoptotic cells and activated platelets, appear to be “poised” in an unresponsive state. Environmental conditions, the metabolic, adhesive and activation state of the phagocyte, and the size of and signals associated with the tethered phagocytic cargo influence the choice of the neutrophils, prompting either phagocytic clearance or the generation of NETs. The choice is dichotomic and apparently irreversible. Defects in phagocytosis may foster the intravascular generation of NETs, thus promoting vascular inflammation and morbidities associated with diseases characterized by defective phagocytic clearance, such as systemic lupus erythematosus. There is a strong potential for novel treatments based on new knowledge of the events determining the inflammatory and pro-thrombotic function of inflammatory leukocytes.
Collapse
Affiliation(s)
- Angelo A Manfredi
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Giuseppe A Ramirez
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Norma Maugeri
- Università Vita-Salute San Raffaele, Milano, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
20
|
Roghanian A, Stopforth RJ, Dahal LN, Cragg MS. New revelations from an old receptor: Immunoregulatory functions of the inhibitory Fc gamma receptor, FcγRIIB (CD32B). J Leukoc Biol 2018; 103:1077-1088. [PMID: 29406570 DOI: 10.1002/jlb.2mir0917-354r] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/03/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
The Fc gamma receptor IIB (FcγRIIB/CD32B) was generated million years ago during evolution. It is the sole inhibitory receptor for IgG, and has long been associated with the regulation of humoral immunity and innate immune homeostasis. However, new and surprising functions of FcγRIIB are emerging. In particular, FcγRIIB has been shown to perform unexpected activatory roles in both immune-signaling and monoclonal antibody (mAb) immunotherapy. Furthermore, although ITIM signaling is an integral part of FcγRIIB regulatory activity, it is now clear that inhibition/activation of immune responses can occur independently of the ITIM. In light of these new findings, we present an overview of the established and noncanonical functions of FcγRIIB and discuss how this knowledge might be exploited therapeutically.
Collapse
Affiliation(s)
- Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard J Stopforth
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Lekh N Dahal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
21
|
Thorsen SU, Pipper CB, Johannesen J, Mortensen HB, Pociot F, Svensson J. '25-Hydroxyvitamin D, Autoantigenic and Total Antibody Concentrations: Results from a Danish Case-control Study of Newly Diagnosed Patients with Childhood Type 1 Diabetes and their Healthy Siblings'. Scand J Immunol 2017; 87:46-53. [PMID: 29125655 DOI: 10.1111/sji.12632] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 11/05/2017] [Indexed: 01/03/2023]
Abstract
B cells have recently entered the stage as an important accessory player in type 1 diabetes (T1D) etiopathogenesis. Experimental studies suggest regulatory functions of vitamin D on B cells. However, only a few human studies, with considerable methodological limitations, have been conducted within this field. Our objective was to investigate whether higher 25-hydroxyvitamin D (25(OH)D) concentrations were inversely associated with β-cell autoantigens glutamic acid decarboxylase (isoform 65) (GADA) and insulinoma-associated antigen-2A (IA-2A). Further, we also wanted to examine the relationship between 25(OH)D and total antibody concentrations. We randomly selected 500 patients with newly diagnosed T1D and 500 siblings for 25(OH)D, antibody and genetic analysis from the population-based Danish Registry of Childhood and Adolescent Diabetes. The relative change (RC) in the mean concentration of GADA, IA-2A and antibody isotypes by a 10 nmol/l increase in 25(OH)D concentration was modelled by a robust log-normal regression model. We found no association between 25(OH)D and GADA [adjusted RC per 10 nmol/l increase: 1.00; 95% confidence interval (CI): 0.98-1.02] and IA-2A [adjusted RC per 10 nmol/l increase: 0.92; CI: 0.76-1.12]. Further, 25(OH)D was not associated with the total concentration of antibody isotypes [immunoglobulin (Ig)A, IgE, IgG and IgM]. All null findings were unaltered after adjustment for genetic variation in the vitamin D pathway. Physiological concentrations of 25(OH)D are unlikely to have a clinically important effect on antibody concentrations in a paediatric population of newly diagnosed patients with T1D and their healthy siblings.
Collapse
Affiliation(s)
- S U Thorsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - C B Pipper
- Department of Public Health, Section of Biostatistics, University of Copenhagen, Copenhagen K, Denmark
| | - J Johannesen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - H B Mortensen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - F Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - J Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
22
|
Concha-Benavente F, Ferris RL. Reversing EGFR Mediated Immunoescape by Targeted Monoclonal Antibody Therapy. Front Pharmacol 2017; 8:332. [PMID: 28611673 PMCID: PMC5447743 DOI: 10.3389/fphar.2017.00332] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/16/2017] [Indexed: 12/29/2022] Open
Abstract
Uncontrolled growth is a signature of carcinogenesis, in part mediated by overexpression or overstimulation of growth factor receptors. The epidermal growth factor receptor (EGFR) mediates activation of multiple oncogenic signaling pathways and escape from recognition by the host immune system. We discuss how EGFR signaling downregulates tumor antigen presentation, upregulates suppressive checkpoint receptor ligand programmed death ligand (PD-L1), induces secretion of inhibitory molecules such as transforming growth factor beta (TGFβ) and reprograms the metabolic pathways in cancer cells to upregulate aerobic glycolysis and lactate secretion that ultimately lead to impaired cellular immunity mediated by natural killer (NK) cell and cytotoxic T lymphocytes (CTL). Ultimately, our understanding of EGFR-mediated escape mechanisms has led us to design EGFR-specific monoclonal antibody therapies that not only inhibit tumor cell metabolic changes and intrinsic oncogenic signaling but also activates immune cells that mediate tumor clearance. Importantly, targeted immunotherapy may also benefit from combination with antibodies that target other immunosuppressive pathways such PD-L1 or TGFβ and ultimately enhance clinical efficacy.
Collapse
Affiliation(s)
- Fernando Concha-Benavente
- Department of Otolaryngology, University of PittsburghPittsburgh, PA, United States.,University of Pittsburgh Cancer InstitutePittsburgh, PA, United States
| | - Robert L Ferris
- Department of Otolaryngology, University of PittsburghPittsburgh, PA, United States.,University of Pittsburgh Cancer InstitutePittsburgh, PA, United States
| |
Collapse
|
23
|
Rolim I, Duarte N, Barata G, Costa J, Gardete-Correia L, Boavida J, Duarte R, Raposo J, Peerally Z, Catarino M, Penha-Gonçalves C. Immunoglobulin M gene association with autoantibody reactivity and type 1 diabetes. Immunogenetics 2017; 69:429-437. [PMID: 28534223 PMCID: PMC5486809 DOI: 10.1007/s00251-017-0999-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/04/2017] [Indexed: 01/07/2023]
Abstract
Several lines of evidence show that autoimmune responses evolving in type 1 diabetes (T1D) patients include the generation of multi-reactive autoantibody (AutoAb) repertoires, but their role in T1D pathogenesis remains elusive. We tested the hypothesis that variants at the immunoglobulin heavy chain (IGH) locus are genetic determinants of AutoAbs against pancreatic antigens and contribute to T1D susceptibility. With this aim, two independent study designs were used: a case-control study and a family-based cohort comprising a total of 240 T1D patients, 172 first-degree relatives (mother and/or father), and 130 unrelated healthy controls living in Portugal. We found that three SNPs in the IGH locus show suggestive association with T1D with the highest nominal association at rs1950942 (in the IGHM-IGHJ gene region) in both the case-control study (P = 9.35E-03) and the family-based cohort (P = 3.08E-03). These SNPs were also associated with IgG AutoAbs against pancreatic antigens and with AutoAb multi-reactivity in T1D patients. Notably, we found that the SNP with the highest association with T1D susceptibility and IgG autoantibody reactivity (rs1950942) was also associated with anti-GAD IgM reactivity in T1D patients (P = 5.98E-03) and in non-affected parents (P = 4.17E-03). This finding implies that IGH association with autoreactive IgM is detectable irrespective of disease status.These results suggest that genetic variants at the IgM gene region of the IGH locus contribute to antibody autoreactivity and are associated with T1D. We propose that the control of autoantibody generation by IGH polymorphisms is a component of the complex architecture of T1D genetic susceptibility.
Collapse
Affiliation(s)
- Inês Rolim
- Instituto Gulbenkian de Ciência, Apartado 14, P-2781-901, Oeiras, Portugal.,Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Duarte
- Instituto Gulbenkian de Ciência, Apartado 14, P-2781-901, Oeiras, Portugal
| | - Gabriela Barata
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.,Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal
| | - João Costa
- Instituto Gulbenkian de Ciência, Apartado 14, P-2781-901, Oeiras, Portugal
| | - Luís Gardete-Correia
- Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal
| | - José Boavida
- Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal
| | - Rui Duarte
- Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal
| | - João Raposo
- Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal
| | - Zulmira Peerally
- Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal
| | | | - Carlos Penha-Gonçalves
- Instituto Gulbenkian de Ciência, Apartado 14, P-2781-901, Oeiras, Portugal. .,Portuguese Diabetes Association, Education and Research Center, Lisbon, Portugal.
| |
Collapse
|
24
|
Lehmann CHK, Baranska A, Heidkamp GF, Heger L, Neubert K, Lühr JJ, Hoffmann A, Reimer KC, Brückner C, Beck S, Seeling M, Kießling M, Soulat D, Krug AB, Ravetch JV, Leusen JHW, Nimmerjahn F, Dudziak D. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J Exp Med 2017; 214:1509-1528. [PMID: 28389502 PMCID: PMC5413326 DOI: 10.1084/jem.20160951] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/19/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Lehmann et al. targeted antigens to Fcγ receptors expressed on various antigen-presenting cells. Induced CD4+ and CD8+ T cell responses were solely dependent on CD11b+ and CD8+ DC subsets, respectively, but independent of receptor intrinsic ITAM or ITIM signaling domains. Dendritic cells (DCs) are efficient antigen-presenting cells equipped with various cell surface receptors for the direct or indirect recognition of pathogenic microorganisms. Interestingly, not much is known about the specific expression pattern and function of the individual activating and inhibitory Fcγ receptors (FcγRs) on splenic DC subsets in vivo and how they contribute to the initiation of T cell responses. By targeting antigens to select activating and the inhibitory FcγR in vivo, we show that antigen uptake under steady-state conditions results in a short-term expansion of antigen-specific T cells, whereas under inflammatory conditions especially, the activating FcγRIV is able to induce superior CD4+ and CD8+ T cell responses. Of note, this effect was independent of FcγR intrinsic activating signaling pathways. Moreover, despite the expression of FcγRIV on both conventional splenic DC subsets, the induction of CD8+ T cell responses was largely dependent on CD11c+CD8+ DCs, whereas CD11c+CD8− DCs were critical for priming CD4+ T cell responses.
Collapse
Affiliation(s)
- Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany.,Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale-Centre National de la Recherche Scientifique, 13288 Marseille-Luminy, France
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kirsten Neubert
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jennifer J Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Alana Hoffmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Katharina C Reimer
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christin Brückner
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Simone Beck
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michaela Seeling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Melissa Kießling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anne B Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany
| | - Jeffrey V Ravetch
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065
| | - Jeanette H W Leusen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, 3584 Utrecht, Netherlands
| | - Falk Nimmerjahn
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
25
|
Immune complexes and complexity: investigating mechanisms of renal disease. Int Urol Nephrol 2016; 49:735-739. [PMID: 27864659 DOI: 10.1007/s11255-016-1450-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
Abstract
The deposition of immune complexes is the causal factor in distinct renal pathologies, e.g., lupus nephritis and membranous nephritis. The location of these deposits within a tissue biopsy is often the key to establishing a diagnosis. However, how immune complexes come to be deposited below the vascular endothelium was, until now, a mystery, as was their contribution to inducing inflammation. A recent paper in Cell by Stamatiades et al. (Cell 164(4):991-1003, 2016) demonstrates the active transport of immune complexes by the vascular endothelial cells and an Fc receptor-dependent uptake by tissue-resident macrophages. This leads to the activation of these macrophages and the release of pro-inflammatory cytokines, which in turn recruits immune cells from the blood into the kidney. The identification of these mechanisms should lead to a better stratification of kidney diseases and hopefully to the development of specific therapies.
Collapse
|
26
|
Erbe AK, Wang W, Goldberg J, Gallenberger M, Kim K, Carmichael L, Hess D, Mendonca EA, Song Y, Hank JA, Cheng SC, Signoretti S, Atkins M, Carlson A, Mier JW, Panka DJ, McDermott DF, Sondel PM. FCGR Polymorphisms Influence Response to IL2 in Metastatic Renal Cell Carcinoma. Clin Cancer Res 2016; 23:2159-2168. [PMID: 27742794 DOI: 10.1158/1078-0432.ccr-16-1874] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/19/2016] [Accepted: 10/03/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Fc-gamma receptors (FCGRs) are expressed on immune cells, bind to antibodies, and trigger antibody-induced cell-mediated antitumor responses when tumor-reactive antibodies are present. The affinity of the FCGR/antibody interaction is variable and dependent upon FCGR polymorphisms. Prior studies of patients with cancer treated with immunotherapy indicate that FCGR polymorphisms can influence antitumor response for certain immunotherapies that act via therapeutically administered mAbs or via endogenous tumor-reactive antibodies induced from tumor antigen vaccines. The previously published "SELECT" trial of high-dose aldesleukin (HD-IL2) for metastatic renal cell carcinoma resulted in an objective response rate of 25%. We evaluated the patients in this SELECT trial to determine whether higher-affinity FCGR polymorphisms are associated with outcome.Experimental Design: SNPs in FCGR2A, FCGR3A, and FCGR2C were analyzed, individually and in combination, for associations between genotype and clinical outcome.Results: When higher-affinity genotypes for FCGR2A, FCGR3A, and FCGR2C were considered together, they were associated with significantly increased tumor shrinkage and prolonged survival in response to HD-IL2.Conclusions: Although associations of higher-affinity FCGR genotype with clinical outcome have been demonstrated with mAb therapy and with idiotype vaccines, to our knowledge, this is the first study to show associations of FCGR genotypes with outcome following HD-IL2 treatment. We hypothesize that endogenous antitumor antibodies may engage immune cells through their FCGRs, and HD-IL2 may enhance antibody-induced tumor destruction, or antibody-enhanced tumor antigen presentation, via augmented activation of innate or adaptive immune responses; this FCGR-mediated immune activity would be augmented through immunologically favorable FCGRs. Clin Cancer Res; 23(9); 2159-68. ©2016 AACR.
Collapse
Affiliation(s)
- Amy K Erbe
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Wei Wang
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Jacob Goldberg
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | | | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin
| | - Dustin Hess
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Eneida A Mendonca
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin.,Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Yiqiang Song
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin
| | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Su-Chun Cheng
- Department of Biostatistics, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael Atkins
- The Cytokine Working Group.,Department of Medicine, Georgetown University, Washington DC
| | - Alexander Carlson
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - James W Mier
- The Cytokine Working Group.,Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - David J Panka
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - David F McDermott
- The Cytokine Working Group.,Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin. .,Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
27
|
Wen YM, Mu L, Shi Y. Immunoregulatory functions of immune complexes in vaccine and therapy. EMBO Mol Med 2016; 8:1120-1133. [PMID: 27572622 PMCID: PMC5048363 DOI: 10.15252/emmm.201606593] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/01/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022] Open
Abstract
Clinical and experimental preparations of IgG/soluble antigen complexes, as well as those formed following antibody therapy in vivo, are multifaceted immune regulators. These immune complexes (ICs) have been tested in humans and animal models, mostly in forms of experimental or clinical vaccination, for at least a century. With intensified research on Fcγ receptor-mediated immune modulation, as well as with immune complex-directed antigen processing, presentation, and inflammatory responses, there are renewed interests of using ICs in vaccines and immunotherapies. Currently, IC-based immune therapy has been broadly experimented in HBV and HIV viral infection control and antitumor treatments. However, mechanistic insights of IC-based treatments are relatively recent subjects of study; strong efforts are needed to establish links to connect laboratory findings with clinical practices. This review covers the history, mechanisms, and in vivo outcomes of this safe and effective therapeutic tool, with a clear aim to bridge laboratory findings with evolving clinical applications.
Collapse
Affiliation(s)
- Yu-Mei Wen
- Key Laboratory of Molecular Medical Virology, MOE/MOH, School of Basic Medical Sciences Shanghai Medical College Fudan University, Shanghai, China
| | - Libing Mu
- Center for Life Sciences, Department of Basic Medical Sciences, Institute of Immunology Tsinghua University, Beijing, China
| | - Yan Shi
- Center for Life Sciences, Department of Basic Medical Sciences, Institute of Immunology Tsinghua University, Beijing, China Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
28
|
Selvaraj UM, Poinsatte K, Torres V, Ortega SB, Stowe AM. Heterogeneity of B Cell Functions in Stroke-Related Risk, Prevention, Injury, and Repair. Neurotherapeutics 2016; 13:729-747. [PMID: 27492770 PMCID: PMC5081124 DOI: 10.1007/s13311-016-0460-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is well established that post-stroke inflammation contributes to neurovascular injury, blood-brain barrier disruption, and poor functional recovery in both animal and clinical studies. However, recent studies also suggest that several leukocyte subsets, activated during the post-stroke immune response, can exhibit both pro-injury and pro-recovery phenotypes. In accordance with these findings, B lymphocytes, or B cells, play a heterogeneous role in the adaptive immune response to stroke. This review highlights what is currently understood about the various roles of B cells, with an emphasis on stroke risk factors, as well as post-stroke injury and repair. This includes an overview of B cell functions, such as antibody production, cytokine secretion, and contribution to the immune response as antigen presenting cells. Next, evidence for B cell-mediated mechanisms in stroke-related risk factors, including hypertension, diabetes, and atherosclerosis, is outlined, followed by studies that focus on B cells during endogenous protection from stroke. Subsequently, animal studies that investigate the role of B cells in post-stroke injury and repair are summarized, and the final section describes current B cell-related clinical trials for stroke, as well as other central nervous system diseases. This review reveals the complex role of B cells in stroke, with a focus on areas for potential clinical intervention for a disease that affects millions of people globally each year.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Katherine Poinsatte
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Vanessa Torres
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Sterling B Ortega
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA.
| |
Collapse
|
29
|
Wan X, Thomas JW, Unanue ER. Class-switched anti-insulin antibodies originate from unconventional antigen presentation in multiple lymphoid sites. J Exp Med 2016; 213:967-78. [PMID: 27139492 PMCID: PMC4886365 DOI: 10.1084/jem.20151869] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 03/09/2016] [Indexed: 01/03/2023] Open
Abstract
Unanue and colleagues show that activation of anti-insulin lymphocytes can occur at diverse anatomical sites in response to circulating insulin and may be driven by unconventional antigen presentation by germinal center B cells. Autoantibodies to insulin are a harbinger of autoimmunity in type 1 diabetes in humans and in non-obese diabetic mice. To understand the genesis of these autoantibodies, we investigated the interactions of insulin-specific T and B lymphocytes using T cell and B cell receptor transgenic mice. We found spontaneous anti-insulin germinal center (GC) formation throughout lymphoid tissues with GC B cells binding insulin. Moreover, because of the nature of the insulin epitope recognized by the T cells, it was evident that GC B cells presented a broader repertoire of insulin epitopes. Such broader recognition was reproduced by activating naive B cells ex vivo with a combination of CD40 ligand and interleukin 4. Thus, insulin immunoreactivity extends beyond the pancreatic lymph node–islets of Langerhans axis and indicates that circulating insulin, despite its very low levels, can have an influence on diabetogenesis.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - James W Thomas
- Department of Medicine, Vanderbilt University Medical School, Nashville, TN 37232
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
30
|
Targeting lymphoma with precision using semisynthetic anti-idiotype peptibodies. Proc Natl Acad Sci U S A 2016; 113:5376-81. [PMID: 27114517 DOI: 10.1073/pnas.1603335113] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
B-cell lymphomas express a functionally active and truly tumor-specific cell-surface product, the variable region of the B-cell receptor (BCR), otherwise known as idiotype. The tumor idiotype differs, however, from patient to patient, making it a technical challenge to exploit for therapy. We have developed a method of targeting idiotype by using a semisynthetic personalized therapeutic that is more practical to produce on a patient-by-patient basis than monoclonal antibodies. In this method, a small peptide with affinity for a tumor idiotype is identified by screening a library, chemically synthesized, and then affixed to the amino terminus of a premade IgG Fc protein. We demonstrate that the resultant semisynthetic anti-idiotype peptibodies kill tumor cells in vitro with specificity, trigger tumor cell phagocytosis by macrophages, and efficiently clear human lymphoma in a murine xenograft model. This method could be used to target tumor with true precision on a personalized basis.
Collapse
|
31
|
Roesner LM, Heratizadeh A, Wieschowski S, Mittermann I, Valenta R, Eiz-Vesper B, Hennig C, Hansen G, Falk CS, Werfel T. α-NAC-Specific Autoreactive CD8+ T Cells in Atopic Dermatitis Are of an Effector Memory Type and Secrete IL-4 and IFN-γ. THE JOURNAL OF IMMUNOLOGY 2016; 196:3245-52. [PMID: 26962231 DOI: 10.4049/jimmunol.1500351] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 02/16/2016] [Indexed: 12/20/2022]
Abstract
Autoreactivity may play a critical role in the chronification of atopic dermatitis (AD). Several studies showed that AD patients produce IgE Abs specific for autoantigens, and we described Th as well as CD8(+) T cells specific for the autoallergen Hom s 2, the α-chain of the nascent polypeptide-associated complex (α-NAC). This study aimed to investigate the frequency and inflammatory phenotype of autoallergen-specific CD8(+) T cells. CD8(+) T cell immunodominant epitopes of α-NAC were mapped by applying prediction softwares, and binding affinity was confirmed by stabilization of empty MHC complexes. MHC class I tetramers were assembled and binding cells were analyzed directly ex vivo by flow cytometry and in terms of single-cell assessment by ChipCytometry. We report significantly elevated numbers of α-NAC-specific peripheral T cells in sensitized patients compared with nonatopic controls. These cells secrete IL-4 and IFN-γ, and surface markers revealed significantly elevated frequencies of circulating terminally differentiated α-NAC-specific CD8(+) T cells in patients with AD compared with nonatopic donors. The observed phenotype of α-NAC-specific CD8(+) T cells indicates a role in the pathogenesis of AD.
Collapse
Affiliation(s)
- Lennart M Roesner
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, 30625 Hannover, Germany;
| | - Annice Heratizadeh
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, 30625 Hannover, Germany
| | - Susanne Wieschowski
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, 30625 Hannover, Germany
| | - Irene Mittermann
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Hennig
- Department of Pediatric Immunology, Allergology and Pneumology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Gesine Hansen
- Department of Pediatric Immunology, Allergology and Pneumology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Christine S Falk
- Institute for Transplant Immunology, Integrated Research and Treatment Center Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
32
|
Gutiérrez-Martínez E, Planès R, Anselmi G, Reynolds M, Menezes S, Adiko AC, Saveanu L, Guermonprez P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol 2015; 6:363. [PMID: 26236315 PMCID: PMC4505393 DOI: 10.3389/fimmu.2015.00363] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/05/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have the unique ability to pick up dead cells carrying antigens in tissue and migrate to the lymph nodes where they can cross-present cell-associated antigens by MHC class I to CD8+ T cells. There is strong in vivo evidence that the mouse XCR1+ DCs subset acts as a key player in this process. The intracellular processes underlying cross-presentation remain controversial and several pathways have been proposed. Indeed, a wide number of studies have addressed the cellular process of cross-presentation in vitro using a variety of sources of antigen and antigen-presenting cells. Here, we review the in vivo and in vitro evidence supporting the current mechanistic models and disscuss their physiological relevance to the cross-presentation of cell-associated antigens by DCs subsets.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Remi Planès
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Matthew Reynolds
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Aimé Cézaire Adiko
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Loredana Saveanu
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| |
Collapse
|
33
|
Abstract
For 20 years, monoclonal antibodies (mAbs) have been a standard component of cancer therapy, but there is still much room for improvement. Efforts continue to build better cancer therapeutics based on mAbs. Anticancer mAbs function through various mechanisms, including directly targeting the malignant cells, modifying the host response, delivering cytotoxic moieties and retargeting cellular immunity towards the malignant cells. Characteristics of mAbs that affect their efficacy include antigen specificity, overall structure, affinity for the target antigen and how a mAb component is incorporated into a construct that can trigger target cell death. This Review discusses the various approaches to using mAb-based therapeutics to treat cancer and the strategies used to take advantage of the unique potential of each approach, and provides examples of current mAb-based treatments.
Collapse
Affiliation(s)
- George J. Weiner
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, 5970Z‐JPP, Iowa City, Iowa 52242, USA.
| |
Collapse
|
34
|
Rodríguez-Rodríguez N, Apostolidis SA, Penaloza-MacMaster P, Martín Villa JM, Barouch DH, Tsokos GC, Crispín JC. Programmed cell death 1 and Helios distinguish TCR-αβ+ double-negative (CD4-CD8-) T cells that derive from self-reactive CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:4207-4214. [PMID: 25825451 PMCID: PMC4503929 DOI: 10.4049/jimmunol.1402775] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/01/2015] [Indexed: 01/20/2023]
Abstract
TCR-αβ(+) double-negative (DN; CD4(-)CD8(-)) T cells represent a poorly understood cellular subset suggested to contribute to the pathogenesis of the autoimmune disease systemic lupus erythematosus. DN T cells have been proposed to derive from CD8(+) cells. However, the conditions that govern the loss of CD8 expression after Ag encounter are unknown. In this study, we tracked the fate of CD8 T cells from transgenic TCR mice exposed to their cognate Ags as self or in the context of infection. We demonstrate that CD8 T cells lose CD8 expression and become DN only when cognate Ag is sensed as self. This process is restricted to tissues where the Ag is present. We also show that DN T cells derived from self-reactive CD8 cells express the inhibitory molecules PD-1 and Helios. These molecules identify a subset of DN T cells in normal mice. A similar population expands when CD8 T cells from repertoires enriched in self-reactive cells (Aire-deficient) are transferred into cognate hosts. Collectively, our data suggest that a subset of DN T cells, identified by the expression of PD-1 and Helios, represent self-reactive cells. Our results provide an explanation for the origin of DN T cells and introduce CD8 loss as a process associated with self-Ag encounter.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215; Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Sokratis A Apostolidis
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Pablo Penaloza-MacMaster
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
| | - José Manuel Martín Villa
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215;
| | - José C Crispín
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215;
| |
Collapse
|
35
|
Pathophysiology of T follicular helper cells in humans and mice. Nat Immunol 2015; 16:142-52. [PMID: 25594465 DOI: 10.1038/ni.3054] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/12/2014] [Indexed: 02/08/2023]
Abstract
Follicular helper T cells (TFH cells) compose a heterogeneous subset of CD4(+) T cells that induce the differentiation of B cells into plasma cells and memory cells. They are found within and in proximity to germinal centers in secondary lymphoid organs, and their memory compartment also circulates in the blood. Our knowledge on the biology of TFH cells has increased significantly during the past decade, largely as a result of mouse studies. However, recent studies on human TFH cells isolated from lymphoid organ and blood samples and recent observations on the developmental mechanism of human TFH cells have revealed both similarities and differences between human and mouse TFH cells. Here we present the similarities and differences between mouse and human lymphoid organ-resident TFH cells and discuss the role of TFH cells in response to vaccines and in disease pathogenesis.
Collapse
|
36
|
Niwa R, Satoh M. The Current Status and Prospects of Antibody Engineering for Therapeutic Use: Focus on Glycoengineering Technology. J Pharm Sci 2015; 104:930-41. [DOI: 10.1002/jps.24316] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 11/09/2014] [Accepted: 12/02/2014] [Indexed: 12/31/2022]
|
37
|
Intrinsic transgene immunogenicity gears CD8(+) T-cell priming after rAAV-mediated muscle gene transfer. Mol Ther 2014; 23:697-706. [PMID: 25492560 DOI: 10.1038/mt.2014.235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/02/2014] [Indexed: 01/18/2023] Open
Abstract
Antitransgene CD8(+) T-cell responses are an important hurdle after recombinant adeno-associated virus (rAAV) vector-mediated gene transfer. Indeed, depending on the mutational genotype of the host, transgene amino-acid sequences of foreign origin can elicit deleterious cellular and humoral responses. We compared here two different major histocompatibility complex (MHC) class I epitopes of an engineered ovalbumin transgene delivered in muscle tissue by rAAV1 vector and found very different strength of CD8 responses, muscle destruction being correlated with the course of the immunodominant response. We further demonstrate that robust CD8(+) T-cell priming can occur through the cross-presentation pathway but requires the presence of either a strong MHC class II epitope or antibodies to the transgene product. Finally, manipulating transgene subcellular localization, we found that provided we avoid transgene expression in antigen presenting cells, the poorly accessible cytosolic form of ovalbumin transgene lacking strong MHC II epitope, evades CD8(+) T-cell priming and remains permanently expressed in muscle with no immune cell infiltration. Our results demonstrate that the intrinsic immunogenicity of transgenes delivered with rAAV vector in muscle can be manipulated in a rational manner to avoid adverse immune responses.
Collapse
|
38
|
Furlan SN, Mandraju R, Brewer T, Roybal K, Troutman TD, Hu W, Palm NW, Unni A, Pasare C. Enhancement of anti-tumor CD8 immunity by IgG1-mediated targeting of Fc receptors. MAbs 2014; 6:108-18. [PMID: 24284965 DOI: 10.4161/mabs.27052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DCs) function as professional antigen presenting cells and are critical for linking innate immune responses to the induction of adaptive immunity. Many current cancer DC vaccine strategies rely on differentiating DCs, feeding them tumor antigens ex vivo, and infusing them into patients. Importantly, this strategy relies on prior knowledge of suitable “tumor-specific” antigens to prime an effective anti-tumor response. DCs express a variety of receptors specific for the Fc region of immunoglobulins, and antigen uptake via Fc receptors is highly efficient and facilitates antigen presentation to T cells. Therefore, we hypothesized that expression of the mouse IgG1 Fc region on the surface of tumors would enhance tumor cell uptake by DCs and other myeloid cells and promote the induction of anti-tumor T cell responses. To test this, we engineered a murine lymphoma cell line expressing surface IgG1 Fc and discovered that such tumor cells were taken up rapidly by DCs, leading to enhanced cross-presentation of tumor-derived antigen to CD8+ T cells. IgG1-Fc tumors failed to grow in vivo and prophylactic vaccination of mice with IgG1-Fc tumors resulted in rejection of unmanipulated tumor cells. Furthermore, IgG1-Fc tumor cells were able to slow the growth of an unmanipulated primary tumor when used as a therapeutic tumor vaccine. Our data demonstrate that engagement of Fc receptors by tumors expressing the Fc region of IgG1 is a viable strategy to induce efficient and protective anti-tumor CD8+ T cell responses without prior knowledge of tumor-specific antigens.
Collapse
|
39
|
Baker K, Rath T, Pyzik M, Blumberg RS. The Role of FcRn in Antigen Presentation. Front Immunol 2014; 5:408. [PMID: 25221553 PMCID: PMC4145246 DOI: 10.3389/fimmu.2014.00408] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/12/2014] [Indexed: 01/06/2023] Open
Abstract
Immunoglobulins are unique molecules capable of simultaneously recognizing a diverse array of antigens and themselves being recognized by a broad array of receptors. The abundance specifically of the IgG subclass and the variety of signaling receptors to which it binds render this an important immunomodulatory molecule. In addition to the classical Fcγ receptors that bind IgG at the cell surface, the neonatal Fc receptor (FcRn) is a lifelong resident of the endolysosomal system of most hematopoietic cells where it determines the intracellular fate of both IgG and IgG-containing immune complexes (IgG IC). Cross-linking of FcRn by multivalent IgG IC within antigen presenting cells such as dendritic cells initiates specific mechanisms that result in trafficking of the antigen-bearing IgG IC into compartments from which the antigen can successfully be processed into peptide epitopes compatible with loading onto both major histocompatibility complex class I and II molecules. In turn, this enables the synchronous activation of both CD4(+) and CD8(+) T cell responses against the cognate antigen, thereby bridging the gap between the humoral and cellular branches of the adaptive immune response. Critically, FcRn-driven T cell priming is efficient at very low doses of antigen due to the exquisite sensitivity of the IgG-mediated antigen delivery system through which it operates. FcRn-mediated antigen presentation has important consequences in tissue compartments replete with IgG and serves not only to determine homeostatic immune activation at a variety of sites but also to induce inflammatory responses upon exposure to antigens perceived as foreign. Therapeutically targeting the pathway by which FcRn enables T cell activation in response to IgG IC is thus a highly attractive prospect not only for the treatment of diseases that are driven by immune complexes but also for manipulating local immune responses against defined antigens such as those present during infections and cancer.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nueremberg, Erlangen, Germany
| | - Michal Pyzik
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Digestive Diseases Center, Boston, MA, USA
| |
Collapse
|
40
|
Wiede F, Ziegler A, Zehn D, Tiganis T. PTPN2 restrains CD8⁺ T cell responses after antigen cross-presentation for the maintenance of peripheral tolerance in mice. J Autoimmun 2014; 53:105-14. [PMID: 24997008 DOI: 10.1016/j.jaut.2014.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 05/12/2014] [Accepted: 05/23/2014] [Indexed: 02/07/2023]
Abstract
Antigen cross-presentation by dendritic cells is crucial for priming cytotoxic CD8(+) T cells to invading pathogens and tumour antigens, as well as mediating peripheral tolerance to self-antigens. The protein tyrosine phosphatase N2 (PTPN2) attenuates T cell receptor (TCR) signalling and tunes CD8(+) T cell responses in vivo. In this study we have examined the role of PTPN2 in the maintenance of peripheral tolerance after the cross-presentation of pancreatic β-cell antigens. The transfer of OVA-specific OT-I CD8(+) T cells (C57BL/6) into RIP-mOVA recipients expressing OVA in pancreatic β-cells only results in islet destruction when OVA-specific CD4(+) T cells are co-transferred. Herein we report that PTPN2-deficient OT-I CD8(+) T cells transferred into RIP-mOVA recipients acquire CTL activity and result in β cell destruction and the development of diabetes in the absence of CD4(+) help. These studies identify PTPN2 as a critical mediator of peripheral T cell tolerance limiting CD8(+) T cell responses after the cross-presentation of self-antigens. Our findings reveal a mechanism by which PTPN2 SNPs might convert a tolerogenic CD8(+) T cell response into one capable of causing the destruction of pancreatic β-cells. Moreover, our results provide insight into potential approaches for enhancing T cell-mediated immunity and/or T cell adoptive tumour immunotherapy.
Collapse
Affiliation(s)
- Florian Wiede
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Alexandra Ziegler
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Dietmar Zehn
- Swiss Vaccine Research Institute, Epalinges, Switzerland
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
41
|
Drujont L, Carretero-Iglesia L, Bouchet-Delbos L, Beriou G, Merieau E, Hill M, Delneste Y, Cuturi MC, Louvet C. Evaluation of the therapeutic potential of bone marrow-derived myeloid suppressor cell (MDSC) adoptive transfer in mouse models of autoimmunity and allograft rejection. PLoS One 2014; 9:e100013. [PMID: 24927018 PMCID: PMC4057339 DOI: 10.1371/journal.pone.0100013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/21/2014] [Indexed: 12/17/2022] Open
Abstract
Therapeutic use of immunoregulatory cells represents a promising approach for the treatment of uncontrolled immunity. During the last decade, myeloid-derived suppressor cells (MDSC) have emerged as novel key regulatory players in the context of tumor growth, inflammation, transplantation or autoimmunity. Recently, MDSC have been successfully generated in vitro from naive mouse bone marrow cells or healthy human PBMCs using minimal cytokine combinations. In this study, we aimed to evaluate the potential of adoptive transfer of such cells to control auto- and allo-immunity in the mouse. Culture of bone marrow cells with GM-CSF and IL-6 consistently yielded a majority of CD11b+Gr1hi/lo cells exhibiting strong inhibition of CD8+ T cell proliferation in vitro. However, adoptive transfer of these cells failed to alter antigen-specific CD8+ T cell proliferation and cytotoxicity in vivo. Furthermore, MDSC could not prevent the development of autoimmunity in a stringent model of type 1 diabetes. Rather, loading the cells prior to injection with a pancreatic neo-antigen peptide accelerated the development of the disease. Contrastingly, in a model of skin transplantation, repeated injection of MDSC or single injection of LPS-activated MDSC resulted in a significant prolongation of allograft survival. The beneficial effect of MDSC infusions on skin graft survival was paradoxically not explained by a decrease of donor-specific T cell response but associated with a systemic over-activation of T cells and antigen presenting cells, prominently in the spleen. Taken together, our results indicate that in vitro generated MDSC bear therapeutic potential but will require additional in vitro factors or adjunct immunosuppressive treatments to achieve safe and more robust immunomodulation upon adoptive transfer.
Collapse
Affiliation(s)
- Lucile Drujont
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Laura Carretero-Iglesia
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Laurence Bouchet-Delbos
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Gaelle Beriou
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Emmanuel Merieau
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Marcelo Hill
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Yves Delneste
- UMR Inserm 892 CNRS 6299, Université d’Angers, CHU Angers, Laboratoire d’Immunologie et Allergologie, Angers, France
| | - Maria Cristina Cuturi
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - Cedric Louvet
- ITUN, Inserm UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
- * E-mail:
| |
Collapse
|
42
|
Platzer B, Stout M, Fiebiger E. Antigen cross-presentation of immune complexes. Front Immunol 2014; 5:140. [PMID: 24744762 PMCID: PMC3978348 DOI: 10.3389/fimmu.2014.00140] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 03/19/2014] [Indexed: 12/23/2022] Open
Abstract
The ability of dendritic cells (DCs) to cross-present tumor antigens has long been a focus of interest to physicians, as well as basic scientists, that aim to establish efficient cell-based cancer immune therapy. A prerequisite for exploiting this pathway for therapeutic purposes is a better understanding of the mechanisms that underlie the induction of tumor-specific cytotoxic T-lymphocyte (CTL) responses when initiated by DCs via cross-presentation. The ability of humans DC to perform cross-presentation is of utmost interest, as this cell type is a main target for cell-based immunotherapy in humans. The outcome of a cross-presentation event is guided by the nature of the antigen, the form of antigen uptake, and the subpopulation of DCs that performs presentation. Generally, CD8α+ DCs are considered to be the most potent cross-presenting DCs. This paradigm, however, only applies to soluble antigens. During adaptive immune responses, immune complexes form when antibodies interact with their specific epitopes on soluble antigens. Immunoglobulin G (IgG) immune complexes target Fc-gamma receptors on DCs to shuttle exogenous antigens efficiently into the cross-presentation pathway. This receptor-mediated cross-presentation pathway is a well-described route for the induction of strong CD8+ T cell responses. IgG-mediated cross-presentation is intriguing because it permits the CD8− DCs, which are commonly considered to be weak cross-presenters, to efficiently cross-present. Engaging multiple DC subtypes for cross-presentation might be a superior strategy to boost CTL responses in vivo. We here summarize our current understanding of how DCs use IgG-complexed antigens for the efficient induction of CTL responses. Because of its importance for human cell therapy, we also review the recent advances in the characterization of cross-presentation properties of human DC subsets.
Collapse
Affiliation(s)
- Barbara Platzer
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Madeleine Stout
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Edda Fiebiger
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
43
|
Lehmann B, Schwab I, Böhm S, Lux A, Biburger M, Nimmerjahn F. FcγRIIB: a modulator of cell activation and humoral tolerance. Expert Rev Clin Immunol 2014; 8:243-54. [DOI: 10.1586/eci.12.5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc Natl Acad Sci U S A 2013; 110:11103-8. [PMID: 23690610 DOI: 10.1073/pnas.1305569110] [Citation(s) in RCA: 510] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mobilization of the T-cell response against cancer has the potential to achieve long-lasting cures. However, it is not known how to harness antigen-presenting cells optimally to achieve an effective antitumor T-cell response. In this study, we show that anti-CD47 antibody-mediated phagocytosis of cancer by macrophages can initiate an antitumor T-cell immune response. Using the ovalbumin model antigen system, anti-CD47 antibody-mediated phagocytosis of cancer cells by macrophages resulted in increased priming of OT-I T cells [cluster of differentiation 8-positive (CD8(+))] but decreased priming of OT-II T cells (CD4(+)). The CD4(+) T-cell response was characterized by a reduction in forkhead box P3-positive (Foxp3(+)) regulatory T cells. Macrophages following anti-CD47-mediated phagocytosis primed CD8(+) T cells to exhibit cytotoxic function in vivo. This response protected animals from tumor challenge. We conclude that anti-CD47 antibody treatment not only enables macrophage phagocytosis of cancer but also can initiate an antitumor cytotoxic T-cell immune response.
Collapse
|
45
|
Baker K, Rath T, Lencer WI, Fiebiger E, Blumberg RS. Cross-presentation of IgG-containing immune complexes. Cell Mol Life Sci 2013; 70:1319-1334. [PMID: 22847331 PMCID: PMC3609906 DOI: 10.1007/s00018-012-1100-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 07/09/2012] [Accepted: 07/17/2012] [Indexed: 12/23/2022]
Abstract
IgG is a molecule that functionally combines facets of both innate and adaptive immunity and therefore bridges both arms of the immune system. On the one hand, IgG is created by adaptive immune cells, but can be generated by B cells independently of T cell help. On the other hand, once secreted, IgG can rapidly deliver antigens into intracellular processing pathways, which enable efficient priming of T cell responses towards epitopes from the cognate antigen initially bound by the IgG. While this process has long been known to participate in CD4(+) T cell activation, IgG-mediated delivery of exogenous antigens into a major histocompatibility complex (MHC) class I processing pathway has received less attention. The coordinated engagement of IgG with IgG receptors expressed on the cell-surface (FcγR) and within the endolysosomal system (FcRn) is a highly potent means to deliver antigen into processing pathways that promote cross-presentation of MHC class I and presentation of MHC class II-restricted epitopes within the same dendritic cell. This review focuses on the mechanisms by which IgG-containing immune complexes mediate such cross-presentation and the implications that this understanding has for manipulation of immune-mediated diseases that depend upon or are due to the activities of CD8(+) T cells.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Wayne I. Lencer
- Division of Gastroenterology and Nutrition, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115 USA
- Harvard Digestive Diseases Center, Boston, MA 02115 USA
| | - Edda Fiebiger
- Division of Gastroenterology and Nutrition, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115 USA
- Harvard Digestive Diseases Center, Boston, MA 02115 USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
- Harvard Digestive Diseases Center, Boston, MA 02115 USA
| |
Collapse
|
46
|
Campbell CT, Gulley JL, Oyelaran O, Hodge JW, Schlom J, Gildersleeve JC. Serum antibodies to blood group A predict survival on PROSTVAC-VF. Clin Cancer Res 2013; 19:1290-9. [PMID: 23362327 PMCID: PMC3594414 DOI: 10.1158/1078-0432.ccr-12-2478] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE There is evidence that therapeutic cancer vaccines can lengthen survival for some patients with cancer, but responses vary widely from one person to another. Methods to predict clinical outcomes will advance the field and provide new insights into critical determinants of in vivo efficacy. EXPERIMENTAL DESIGN This retrospective study included 141 subjects from phase II trials of PROSTVAC-VF, a poxvirus-based cancer vaccine currently in phase III clinical trials for advanced prostate cancer. A glycan microarray was used to profile prevaccination antiglycan antibody populations in sera as potential biomarkers for PROSTVAC-VF. The screen for predictive biomarkers identified antiglycan antibodies that consistently stratified subjects into groups with different Kaplan-Meier survival estimates. Because of the potential for overfitting, a permutation test was used to estimate the false discovery rate. RESULTS Prevaccination antibody levels to blood group A trisaccharide (BG-Atri) were found to have a statistically significant correlation with survival. Long-term survival was approximately doubled in subjects with abundant anti-BG-Atri immunoglobulin M (IgM) relative to subjects with little or no preexisting IgM for BG-Atri. This survival correlation was specific to vaccine treatment, as no correlation was observed in control patients immunized with wild-type poxviruses lacking the key tumor antigen, prostate-specific antigen (PSA). Moreover, anti-BG-Atri IgM levels were not correlated with general measures of disease severity, such as PSA levels, Gleason score, or Halabi predicted survival. CONCLUSION In addition to reporting a new potentially predictive biomarker for PROSTVAC-VF, this study highlights the use of glycan microarray technology for improving our understanding of vaccine immunology. Clin Cancer Res; 19(5); 1290-9. ©2012 AACR.
Collapse
Affiliation(s)
- Christopher T. Campbell
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702
| | - James L. Gulley
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Oyindasola Oyelaran
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jeffrey C. Gildersleeve
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702
| |
Collapse
|
47
|
Ly LV, Sluijter M, van der Burg SH, Jager MJ, van Hall T. Effective cooperation of monoclonal antibody and peptide vaccine for the treatment of mouse melanoma. THE JOURNAL OF IMMUNOLOGY 2012. [PMID: 23203930 DOI: 10.4049/jimmunol.1200135] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
mAbs binding to tumor-associated surface Ags are therapeutically applied in a range of malignancies. Therapeutic vaccination only recently met with clinical success, and the first cancer vaccine received U.S. Food and Drug Administration approval last year. To improve current protocols, we combined peptide vaccines with mAb to the tyrosinase-related protein (TRP)-1 surface Ag for the treatment of B16F10 skin melanoma. Vaccine formulations with synthetic long peptides failed to elicit strong CD8 T cell responses to self-differentiation Ags gp100 and TRP-2, whereas altered peptide sequences recruited gp100-specific CD8 T cells from the endogenous repertoire with frequencies of 40%. However, these high frequencies were reached too late; large, progressively growing melanomas had already emerged. Addition of the TRP-1-directed mAb TA99 to the treatment protocol mediated eradication of s.c. lesions. The mode of action of the Ab did not depend on complement factor C3 and did not lead to improved Ag presentation and CD8 T cell immunity; rather, it recruited FcγR-bearing innate immune cells during early tumor control, thereby creating a window of time for the generation of protective cellular immunity. These data support the concept of combination therapy, in which passive transfer of mAbs is supplemented with cancer peptide vaccines. Moreover, we advocate that tumor Ag-specific T cell immunity directed against self-proteins can be exploited from the endogenous repertoire.
Collapse
Affiliation(s)
- Long V Ly
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
48
|
Abstract
A classic understanding of the interplay between B and T cell components of the immune system that drive autoimmunity, where B cells provide an effector function, is represented by systemic lupus erythematosus (SLE), an autoimmune condition characterised by the production of auto-antibodies. In SLE, CD4+T cells provide cognate help to self-reactive B cells, which in turn produce pathogenic auto-antibodies (1). Thus, B cells act as effectors by producing auto-antibody aided by T cell help such that B and T cell interactions are unidirectional. However, this paradigm of B and T cell interactions is challenged by new clinical data demonstrating that B cell depletion is effective for T cell mediated autoimmune diseases including type I diabetes mellitus (T1D) (2), rheumatoid arthritis (3), and multiple sclerosis (4). These clinical data indicate a model whereby B cells can influence the developing autoimmune T cell response, and therefore act as effectors, in ways that extend beyond the production of autoantibody (5). In this review by largely focusing on type I diabetes we will develop a hypothesis that bi-directional B and T interactions control the course of autoimmunity.
Collapse
Affiliation(s)
- Eliana Mariño
- Centre of Immunology and Inflammation, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | | |
Collapse
|
49
|
Towards curative cancer immunotherapy: overcoming posttherapy tumor escape. Clin Dev Immunol 2012; 2012:124187. [PMID: 22778760 PMCID: PMC3386616 DOI: 10.1155/2012/124187] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/06/2012] [Indexed: 02/07/2023]
Abstract
The past decade has witnessed the evolvement of cancer immunotherapy as an increasingly effective therapeutic modality, evidenced by the approval of two immune-based products by the FDA, that is, the cancer vaccine Provenge (sipuleucel-T) for prostate cancer and the antagonist antibody against cytotoxic T-lymphocyte antigen-4 (CTLA-4) ipilimumab for advanced melanoma. In addition, the clinical evaluations of a variety of promising immunotherapy drugs are well under way. Benefiting from more efficacious immunotherapeutic agents and treatment strategies, a number of recent clinical studies have achieved unprecedented therapeutic outcomes in some patients with certain types of cancers. Despite these advances, however, the efficacy of most cancer immunotherapies currently under clinical development has been modest. A recurring scenario is that therapeutic maneuvers initially led to measurable antitumor immune responses in cancer patients but ultimately failed to improve patient outcomes. It is increasingly recognized that tumor cells can antagonize therapy-induced immune attacks through a variety of counterregulation mechanisms, which represent a fundamental barrier to the success of cancer immunotherapy. Herein we summarize the findings from some recent preclinical and clinical studies, focusing on how tumor cells advance their survival and expansion by hijacking therapy-induced immune effector mechanisms that would otherwise mediate their destruction.
Collapse
|
50
|
Czyzyk J, Henegariu O, Preston-Hurlburt P, Baldzizhar R, Fedorchuk C, Esplugues E, Bottomly K, Gorus FK, Herold K, Flavell RA. Enhanced anti-serpin antibody activity inhibits autoimmune inflammation in type 1 diabetes. THE JOURNAL OF IMMUNOLOGY 2012; 188:6319-27. [PMID: 22593614 DOI: 10.4049/jimmunol.1200467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intracellular (clade B) OVA-serpin protease inhibitors play an important role in tissue homeostasis by protecting cells from death in response to hypo-osmotic stress, heat shock, and other stimuli. It is not known whether these serpins influence immunological tolerance and the risk for autoimmune diseases. We found that a fraction of young autoimmune diabetes-prone NOD mice had elevated levels of autoantibodies against a member of clade B family known as serpinB13. High levels of anti-serpinB13 Abs were accompanied by low levels of anti-insulin autoantibodies, reduced numbers of islet-associated T cells, and delayed onset of diabetes. Exposure to anti-serpinB13 mAb alone also decreased islet inflammation, and coadministration of this reagent and a suboptimal dose of anti-CD3 mAb accelerated recovery from diabetes. In a fashion similar to that discovered in the NOD model, a deficiency in humoral activity against serpinB13 was associated with early onset of human type 1 diabetes. These findings suggest that, in addition to limiting exposure to proteases within the cell, clade B serpins help to maintain homeostasis by inducing protective humoral immunity.
Collapse
Affiliation(s)
- Jan Czyzyk
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|