1
|
Bandola-Simon J, Ito Y, Wucherpfennig KW, Roche PA. Defective removal of invariant chain peptides from MHC class II suppresses tumor antigen presentation and promotes tumor growth. Cell Rep 2025; 44:115150. [PMID: 39752250 PMCID: PMC11886875 DOI: 10.1016/j.celrep.2024.115150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 02/01/2025] Open
Abstract
Tumor-draining lymph node dendritic cells (DCs) are poor stimulators of tumor antigen-specific CD4 T cells; however, the mechanism behind this defect is unclear. We now show that, in tumor-draining lymph node DCs, a large proportion of major histocompatibility complex class II (MHC-II) molecules retains the class II-associated invariant chain peptide (CLIP) fragment of the invariant chain bound to the MHC-II peptide binding groove due to reduced expression of the peptide editor H2-M and enhanced activity of the CLIP-generating proteinase cathepsin S. The net effect of this is that MHC-II molecules are unable to efficiently bind antigenic peptides. DCs in mice expressing a mutation in the invariant chain sequence that results in enhanced MHC-II-CLIP accumulation are poor stimulators of CD4 T cells and have diminished anti-tumor responses. Our data reveal a previously unknown mechanism of immune evasion in which enhanced expression of MHC-II-CLIP complexes on tumor-draining lymph node DCs limits MHC-II availability for tumor peptides.
Collapse
MESH Headings
- Histocompatibility Antigens Class II/metabolism
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/genetics
- Animals
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigen Presentation/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Mice
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Mice, Inbred C57BL
- CD4-Positive T-Lymphocytes/immunology
- Peptides/metabolism
- Peptides/immunology
- Lymph Nodes/immunology
- Neoplasms/immunology
- Neoplasms/pathology
- Cell Line, Tumor
- Humans
Collapse
Affiliation(s)
- Joanna Bandola-Simon
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshinaga Ito
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Childs BA, Kim J, Patel RR, Vandergriff TW, Goff HW, Wang RC. Whole-Exome Sequencing Identifies Novel and Previously Reported Mutations in a Case of Intravascular B-Cell Lymphoma. Am J Dermatopathol 2024; 46:890-893. [PMID: 39288753 DOI: 10.1097/dad.0000000000002824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Affiliation(s)
- Beth A Childs
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
| | - Jiwoong Kim
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX; and
| | - Ravi R Patel
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
| | | | - Heather W Goff
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
| | - Richard C Wang
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
- Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
3
|
Kim S, Jeong H, Ahn HK, Han B, Lee KC, Song YK, Lim S, Yim J, Koh J, Jeon YK. Increased CCL2/CCR2 axis promotes tumor progression by increasing M2 macrophages in MYC/BCL2 double-expressor DLBCL. Blood Adv 2024; 8:5773-5788. [PMID: 39293078 PMCID: PMC11605354 DOI: 10.1182/bloodadvances.2024013699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
ABSTRACT The pathogenesis of myelocytomatosis oncogene (MYC) and B-cell lymphoma 2 (BCL2) double-expressor diffuse large B-cell lymphoma (DE-DLBCL) remains unclear. To investigate how MYC and BCL2 contribute to tumor aggressiveness, we analyzed tumors from 14 patients each with DE-DLBCL and non-DE-DLBCL using whole transcriptome sequencing. Validation was performed using publicly available data sets, tumor tissues from 126 patients, DLBCL cell lines, and a syngeneic mouse lymphoma model. Our transcriptome analysis revealed significantly elevated messenger RNA levels of C-C motif chemokine ligand 2 (CCL2) and C-C chemokine receptor type 2 (CCR2) in DE-DLBCLs when compared with non-DE-DLBCLs (adjusted P value < .05). Transcriptomic analysis of public data sets and immunohistochemistry corroborated these findings, indicating increased levels of M2 macrophages but a reduction in T-cell infiltration in DE-DLBCLs when compared with non-DE-DLBCLs (all P < .05). CCR2 expression was observed mainly in tumor-infiltrating macrophages and not in DLBCL cells. Increased expression of CCL2 and CCR2 was significantly associated with a poor prognosis in patients with DLBCL. In the in vitro analyses, MYChigh/BCL2high DLBCL cells showed higher CCL2 expression and secretion than MYClow/BCL2low cells. MYC and BCL2 increased CCL2 expression and secretion by upregulation of nuclear factor κB p65 in DLBCL cells, and CCL2 promoted M2 polarization of macrophages. In a mouse lymphoma model, CCL2 contributed to the immunosuppressive microenvironment and tumor growth of MYChigh/BCL2high tumors. We demonstrated that the increased CCL2/CCR2 axis confers aggressiveness to DE-DLBCL by increasing M2 polarization and can be a potential therapeutic target.
Collapse
MESH Headings
- Receptors, CCR2/metabolism
- Receptors, CCR2/genetics
- Humans
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Animals
- Proto-Oncogene Proteins c-myc/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Mice
- Macrophages/metabolism
- Gene Expression Regulation, Neoplastic
- Disease Progression
- Cell Line, Tumor
- Tumor Microenvironment
- Female
- Male
Collapse
Affiliation(s)
- Sehui Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyein Jeong
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Hyun Kyung Ahn
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Bogyeong Han
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki-Chang Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Keun Song
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sojung Lim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeemin Yim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Seoul Metropolitan Government, Seoul National University Boramae Hospital, Seoul, Republic of Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Republic of Korea
| |
Collapse
|
4
|
Zhou Y, Lou J, Tian Y, Ding J, Wang X, Tang B. How lactate affects immune strategies in lymphoma. Front Mol Biosci 2024; 11:1480884. [PMID: 39464313 PMCID: PMC11502318 DOI: 10.3389/fmolb.2024.1480884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Tumor cells undergo metabolic reprogramming through shared pathways, resulting in a hypoxic, acidic, and highly permeable internal tumor microenvironment (TME). Lactate, once only regarded as a waste product of glycolysis, has an inseparable dual role with tumor immunity. It can not only provide a carbon source for immune cells to enhance immunity but also help the immune escape through a variety of ways. Lymphoma also depends on the proliferation signal of TME. This review focuses on the dynamic process of lactate metabolism and immune function changes in lymphoma and aims to comprehensively summarize and explore which genes, transcription factors, and pathways affect the biological changes and functions of immune cells. To deeply understand the complex and multifaceted role of lactate metabolism and immunity in lymphoma, the combination of lactate targeted therapy and classical immunotherapy will be a promising development direction in the future.
Collapse
Affiliation(s)
- Yuehan Zhou
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinzhan Lou
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuqin Tian
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinlei Ding
- Department of Thoracic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaobo Wang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Tang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Fei F, Rong L, Jiang N, Wayne AS, Xie J. Targeting HLA-DR loss in hematologic malignancies with an inhibitory chimeric antigen receptor. Mol Ther 2022; 30:1215-1226. [PMID: 34801727 PMCID: PMC8899520 DOI: 10.1016/j.ymthe.2021.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/19/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptor natural killer (CAR-NK) cells have remarkable cytotoxicity against hematologic malignancies; however, they may also attack normal cells sharing the target antigen. Since human leukocyte antigen DR (HLA-DR) is reportedly lost or downregulated in a substantial proportion of hematologic malignancies, presumably a mechanism to escape immune surveillance, we hypothesize that the anti-cancer specificity of CAR-NK cells can be enhanced by activating them against cancer antigens while inhibiting them against HLA-DR. Here, we report the development of an anti-HLA-DR inhibitory CAR (iCAR) that can effectively suppress NK cell activation against HLA-DR-expressing cells. We show that dual CAR-NK cells, which co-express the anti-CD19 or CD33 activating CAR and the anti-HLA-DR iCAR, can preferentially target HLA-DR-negative cells over HLA-DR-positive cells in vitro. We find that the HLA-DR-mediated inhibition is positively correlated with both iCAR and HLA-DR densities. We also find that HLA-DR-expressing surrounding cells do not affect the target selectivity of dual CAR-NK cells. Finally, we confirm that HLA-DR-positive cells are resistant to dual CAR-NK cell-mediated killing in a xenograft mouse model. Our approach holds great promise for enhancing CAR-NK and CAR-T cell specificity against malignancies with HLA-DR loss.
Collapse
Affiliation(s)
- Fan Fei
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Liang Rong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Nan Jiang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Alan S. Wayne
- Cancer and Blood Disease Institute, Division of Hematology-Oncology, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jianming Xie
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
6
|
Structural aspects of the MHC expression control system. Biophys Chem 2022; 284:106781. [PMID: 35228036 PMCID: PMC8941990 DOI: 10.1016/j.bpc.2022.106781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/04/2022] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
The major histocompatibility complex (MHC) spans innate and adaptive immunity by presenting antigenic peptides to CD4+ and CD8+ T cells. Multiple transcription factors form an enhanceosome complex on the MHC promoter and recruit transcriptional machinery to activate gene transcription. Immune signals such as interferon-γ (IFN-γ) control MHC level by up-regulating components of the enhanceosome complex. As MHC plays crucial roles in immune regulation, alterations in the MHC enhanceosome structure will alter the pace of rapid immune responses at the transcription level and lead to various diseases related to the immune system. In this review, we discuss the current understanding of the MHC enhanceosome, with a focus on the structures of MHC enhanceosome components and the molecular basis of MHC enhanceosome assembly.
Collapse
|
7
|
Toloza MJ, Bestach Y, Lincango-Yupanki M, Bordone J, Mariano R, Tarqui M, Pérez M, Aranguren PN, Enrico A, Larripa IB, Belli CB. Expression dynamics of the immune mediators ARG1, TBET, CIITA, IL10 and TGFB1 in chronic myeloid leukaemia patients during the first year of imatinib therapy. Gene 2021; 813:146110. [PMID: 34902507 DOI: 10.1016/j.gene.2021.146110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/24/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022]
Abstract
The use of tyrosine kinase inhibitors seems to restore the broadly compromised immune system described in chronic myeloid leukaemia (CML) patients at diagnosis leading to a re-activation of the effector-mediated immune surveillance. Here, we describe the expression dynamics of immune factors during the first year on imatinib therapy. Gene expression was evaluated in 132 peripheral blood samples from 79 CML patients, including 34 who were serially followed. An aliquot of the stored sample used to monitor BCR-ABL1 levels was retro-transcribed to cDNA and gene expression was quantified by real-time PCR. An elevated expression of ARG1 was observed at diagnosis, while TBET, CIITA, IL10 and TGFB1 were significantly decreased. Once on therapy, each gene displayed a particular behaviour. ARG1 normalized to control levels at 3 months only in optimal molecular responders and was identified as the major contributor to the difference among patients. TBET reached normal levels after 12 months in optimal responders and non-responders, regardless the Th1-response previously associated, and CIITA continued downregulated. IL10 and TGFB1 achieved normal levels early at 3 months in both groups, afterwards IL10 was sustained while TGFB1 was slightly increased after 1 year in responders. Our findings are in agreement with an immune re-activation after imatinib initiation; however, some immune mediators may require a longer exposition. The follow-up of novel and reliable biomarkers, such as ARG1, one of the principal mechanisms of myeloid-derived-suppressor cells to inhibit immune system, may be useful to deepen the characterization of early responder patients.
Collapse
MESH Headings
- Adult
- Antineoplastic Agents/pharmacology
- Arginase/genetics
- Arginase/metabolism
- Biomarkers, Pharmacological/analysis
- Biomarkers, Pharmacological/blood
- Female
- Fusion Proteins, bcr-abl/genetics
- Gene Expression
- Humans
- Imatinib Mesylate/therapeutic use
- Immunologic Factors/therapeutic use
- Interleukin-10/blood
- Interleukin-10/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Male
- Middle Aged
- Nuclear Proteins/blood
- Nuclear Proteins/genetics
- Protein Kinase Inhibitors/pharmacology
- Trans-Activators/blood
- Trans-Activators/genetics
- Transcriptome/genetics
- Transforming Growth Factor beta1/blood
- Transforming Growth Factor beta1/genetics
Collapse
Affiliation(s)
- María Jazmín Toloza
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina.
| | - Yesica Bestach
- Laboratorio de Inmunofarmacología, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Marco Lincango-Yupanki
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Javier Bordone
- Servicio de Hematología, Hospital de Alta Complejidad en red "El Cruce", Argentina
| | - Romina Mariano
- Servicio de Hematología, Hospital "San Martín de Paraná", Argentina
| | - Melissa Tarqui
- Servicio de Hematología, Hospital Italiano de La Plata, Argentina
| | - Mariel Pérez
- Servicio de Hematología, Hospital Interzonal General de Agudos "Prof. Dr. Rodolfo Rossi", La Plata, Argentina
| | | | - Alicia Enrico
- Servicio de Hematología, Hospital Italiano de La Plata, Argentina
| | - Irene B Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Carolina B Belli
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| |
Collapse
|
8
|
Abstract
Diffuse large B-cell lymphomas (DLBCL)s, the most common type of Non-Hodgkin’s Lymphoma, constitute a heterogeneous group of disorders including different disease sites, strikingly diverse molecular features and a profound variability in the clinical behavior. Molecular studies and clinical trials have partially revealed the underlying causes for this variability and have made possible the recognition of some molecular variants susceptible of specific therapeutic approaches. The main histogenetic groups include the germinal center, activated B cells, thymic B cells and terminally differentiated B cells, a basic scheme where the large majority of DLBCL cases can be ascribed. The nodal/extranodal origin, specific mutational changes and microenvironment peculiarities provide additional layers of complexity. Here, we summarize the status of the knowledge and make some specific proposals for addressing the future development of targeted therapy for DLBC cases.
Collapse
|
9
|
Apollonio B, Ioannou N, Papazoglou D, Ramsay AG. Understanding the Immune-Stroma Microenvironment in B Cell Malignancies for Effective Immunotherapy. Front Oncol 2021; 11:626818. [PMID: 33842331 PMCID: PMC8027510 DOI: 10.3389/fonc.2021.626818] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022] Open
Abstract
Cancers, including lymphomas, develop in complex tissue environments where malignant cells actively promote the creation of a pro-tumoral niche that suppresses effective anti-tumor effector T cell responses. Research is revealing that the tumor microenvironment (TME) differs between different types of lymphoma, covering inflamed environments, as exemplified by Hodgkin lymphoma, to non-inflamed TMEs as seen in chronic lymphocytic leukemia (CLL) or diffuse-large B-cell lymphoma (DLBCL). In this review we consider how T cells and interferon-driven inflammatory signaling contribute to the regulation of anti-tumor immune responses, as well as sensitivity to anti-PD-1 immune checkpoint blockade immunotherapy. We discuss tumor intrinsic and extrinsic mechanisms critical to anti-tumor immune responses, as well as sensitivity to immunotherapies, before adding an additional layer of complexity within the TME: the immunoregulatory role of non-hematopoietic stromal cells that co-evolve with tumors. Studying the intricate interactions between the immune-stroma lymphoma TME should help to design next-generation immunotherapies and combination treatment strategies to overcome complex TME-driven immune suppression.
Collapse
Affiliation(s)
- Benedetta Apollonio
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Nikolaos Ioannou
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Despoina Papazoglou
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Alan G Ramsay
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
10
|
Csizmar CM, Ansell SM. Engaging the Innate and Adaptive Antitumor Immune Response in Lymphoma. Int J Mol Sci 2021; 22:3302. [PMID: 33804869 PMCID: PMC8038124 DOI: 10.3390/ijms22073302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has emerged as a powerful therapeutic strategy for many malignancies, including lymphoma. As in solid tumors, early clinical trials have revealed that immunotherapy is not equally efficacious across all lymphoma subtypes. For example, immune checkpoint inhibition has a higher overall response rate and leads to more durable outcomes in Hodgkin lymphomas compared to non-Hodgkin lymphomas. These observations, combined with a growing understanding of tumor biology, have implicated the tumor microenvironment as a major determinant of treatment response and prognosis. Interactions between lymphoma cells and their microenvironment facilitate several mechanisms that impair the antitumor immune response, including loss of major histocompatibility complexes, expression of immunosuppressive ligands, secretion of immunosuppressive cytokines, and the recruitment, expansion, and skewing of suppressive cell populations. Accordingly, treatments to overcome these barriers are being rapidly developed and translated into clinical trials. This review will discuss the mechanisms of immune evasion, current avenues for optimizing the antitumor immune response, clinical successes and failures of lymphoma immunotherapy, and outstanding hurdles that remain to be addressed.
Collapse
Affiliation(s)
| | - Stephen M. Ansell
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Augé H, Notarantonio AB, Morizot R, Quinquenel A, Fornecker LM, Hergalant S, Feugier P, Broséus J. Microenvironment Remodeling and Subsequent Clinical Implications in Diffuse Large B-Cell Histologic Variant of Richter Syndrome. Front Immunol 2020; 11:594841. [PMID: 33381116 PMCID: PMC7767850 DOI: 10.3389/fimmu.2020.594841] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Richter Syndrome (RS) is defined as the development of an aggressive lymphoma in the context of Chronic Lymphocytic Leukemia (CLL), with a Diffuse Large B-Cell Lymphoma (DLBCL) histology in 95% cases. RS genomic landscape shares only a few features with de novo DLBCLs and is marked by a wide spectrum of cytogenetic abnormalities. Little is known about RS microenvironment. Therapeutic options and efficacy are limited, leading to a 12 months median overall survival. The new targeted treatments usually effective in CLL fail to obtain long-term remissions in RS. Methods We reviewed available PubMed literature about RS genomics, PD-1/PD-L1 (Programmed Death 1/Programmed Death Ligand 1) pathway triggering and subsequent new therapeutic options. Results Data from about 207 patients from four landmark papers were compiled to build an overview of RS genomic lesions and point mutations. A number of these abnormalities may be involved in tumor microenvironment reshaping. T lymphocyte exhaustion through PD-L1 overexpression by tumor cells and subsequent PD-1/PD-L1 pathway triggering is frequently reported in solid cancers. This immune checkpoint inhibitor is also described in B lymphoid malignancies, particularly CLL: PD-1 expression is reported in a subset of prolymphocytes from the CLL lymph node proliferation centers. However, there is only few data about PD-1/PD-L1 pathway in RS. In RS, PD-1 expression is a hallmark of recently described « Regulatory B-cells », which interact with tumor microenvironment by producing inhibiting cytokines such as TGF-β and IL-10, impairing T lymphocytes anti-tumoral function. Based upon the discovery of high PD-1 expression on tumoral B lymphocyte from RS, immune checkpoint blockade therapies such as anti-PD-1 antibodies have been tested on small RS cohorts and provided heterogeneous but encouraging results. Conclusion RS genetic landscape and immune evasion mechanisms are being progressively unraveled. New protocols using targeted treatments such as checkpoint inhibitors as single agents or in combination with immunochemotherapy are currently being evaluated.
Collapse
Affiliation(s)
- Hélène Augé
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risque Environnementaux (N-GERE), Université de Lorraine, Nancy, France.,Université de Lorraine, CHRU-Nancy, service d'hématologie clinique, pôle spécialités médicales, Nancy, France
| | - Anne-Béatrice Notarantonio
- Université de Lorraine, CHRU-Nancy, service d'hématologie clinique, pôle spécialités médicales, Nancy, France.,UMR7365 Ingénierie Moléculaire et Physiopathologie Articulaire (IMOPA), CNRS, Université de Lorraine, Nancy, France
| | - Romain Morizot
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risque Environnementaux (N-GERE), Université de Lorraine, Nancy, France.,Université de Lorraine, CHRU-Nancy, service d'hématologie clinique, pôle spécialités médicales, Nancy, France
| | - Anne Quinquenel
- Département d'hématologie, Université de Reims Champagne-Ardenne, Reims, France.,Département d'hématologie clinique, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Luc-Matthieu Fornecker
- Université de Strasbourg, Inserm, IRFAC/UMR-S1113, Strasbourg, France.,Département d'hématologie clinique, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Sébastien Hergalant
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risque Environnementaux (N-GERE), Université de Lorraine, Nancy, France
| | - Pierre Feugier
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risque Environnementaux (N-GERE), Université de Lorraine, Nancy, France.,Université de Lorraine, CHRU-Nancy, service d'hématologie clinique, pôle spécialités médicales, Nancy, France
| | - Julien Broséus
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risque Environnementaux (N-GERE), Université de Lorraine, Nancy, France.,Université de Lorraine, CHRU-Nancy, service d'hématologie biologique, pôle laboratoires, Nancy, France
| |
Collapse
|
12
|
Evolving insights into the genomic complexity and immune landscape of diffuse large B-cell lymphoma: opportunities for novel biomarkers. Mod Pathol 2020; 33:2422-2436. [PMID: 32620919 DOI: 10.1038/s41379-020-0616-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Recently, comprehensive genomic analyses have allowed a better molecular characterization of diffuse large B-cell lymphoma (DLBCL), offering novel opportunities in patient risk stratification and management. In the era of precision medicine, this has allowed us to move closer toward a more promising therapeutic outcome in the setting of DLBCL. In this review, we highlight the newly reported heterogeneous mutational landscapes of DLBCL (from two whole-exome sequencing studies, and from a more recent work targeting a 293-gene of a hematologic malignancy-designed panel. Altogether, these studies provide further evidence of the clinical applicability of genomic tests. We also briefly review established biomarkers in DLBCL (e.g., MYC and TP53), and our understanding of the germinal center cell reaction, including its epigenetic regulation, emphasizing some of the key epigenetic modifiers that play a role in lymphomagenesis, with available therapeutic targets. In addition, we present current data regarding the role of immune landscapes in DLBCL (inflamed versus non-inflamed), how the recently defined molecular DLBCL subtypes may affect the cellular composition of the tumor microenvironment and the function of the immune cells, and how this new knowledge may result in promising therapeutic approaches in the near future.
Collapse
|
13
|
The immune landscape and response to immune checkpoint blockade therapy in lymphoma. Blood 2020; 135:523-533. [PMID: 31790142 DOI: 10.1182/blood.2019000847] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
The clinical development of effective cancer immunotherapies, along with advances in genomic analysis, has led to the identification of tumor environmental features that predict for sensitivity to immune checkpoint blockade therapy (CBT). Early-phase clinical trial results have demonstrated the remarkable effectiveness of CBT in specific lymphoma subtypes, including classical Hodgkin lymphoma and primary mediastinal B-cell lymphoma. Conversely, CBT has been relatively disappointing in follicular lymphoma and diffuse large B-cell lymphoma. These clinical observations, coupled with important scientific discoveries, have uncovered salient features of the lymphoma microenvironment that correlate with immunotherapy response in patients. For example, classical Hodgkin lymphoma is characterized by an inflammatory environment, genetic alterations that facilitate escape from immune attack, and sensitivity to PD-1 blockade therapy. On the other hand, for lymphomas in which measures of immune surveillance are lacking, including follicular lymphoma and most diffuse large B-cell lymphomas, anti-PD-1 therapy has been less effective. An improved understanding of the immune landscapes of these lymphomas is needed to define subsets that might benefit from CBT. In this article, we describe the immune environments associated with major B-cell lymphomas with an emphasis on the immune escape pathways orchestrated by these diseases. We also discuss how oncogenic alterations in lymphoma cells may affect the cellular composition of the immune environment and ultimately, vulnerability to CBT. Finally, we highlight key areas for future investigation, including the need for the development of biomarkers that predict for sensitivity to CBT in lymphoma patients.
Collapse
|
14
|
Nesic M, El-Galaly TC, Bøgsted M, Pedersen IS, Dybkær K. Mutational landscape of immune surveillance genes in diffuse large B-cell lymphoma. Expert Rev Hematol 2020; 13:655-668. [PMID: 32293210 DOI: 10.1080/17474086.2020.1755958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Immune surveillance is the dynamic process whereby the immune system identifies and kills tumor cells based on their aberrant expression of stress-related surface molecules or presentation of tumor neoantigens. It plays a crucial role in controlling the initiation and progression of hematologic cancers such as leukemia and lymphoma, and it has been reported that diffuse large B-cell lymphoma (DLBCL) fails to express specific cell-surface molecules that are necessary for the recognition and elimination of tumor cells. AREAS COVERED This review is based on a systematic search strategy to identify relevant literature in the PubMed and Embase databases. Ten candidate genes are identified based on mutational frequency, and functions with detailed mapping performed for hotspot alterations that may have a functional impact on malignant transformation and decreased immune surveillance efficacy. EXPERT OPINION Ongoing development of technology and bioinformatics tools combined with data from large clinical cohorts have the potential to define the mutational landscape associated with immune surveillance in DLBCL. Specific functional studies are required to make an unambiguous link between genetic aberrations and biological impact on impaired immune surveillance.
Collapse
Affiliation(s)
- Marijana Nesic
- Department of Hematology, Aalborg University Hospital , Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital , Aalborg, Denmark
| | - Tarec Christoffer El-Galaly
- Department of Hematology, Aalborg University Hospital , Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital , Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University , Aalborg, Denmark
| | - Martin Bøgsted
- Department of Hematology, Aalborg University Hospital , Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital , Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University , Aalborg, Denmark
| | - Inge Søkilde Pedersen
- Department of Clinical Medicine, Aalborg University , Aalborg, Denmark.,Department of Molecular Diagnostics, Aalborg University Hospital , Aalborg, Denmark
| | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital , Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital , Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University , Aalborg, Denmark
| |
Collapse
|
15
|
Understanding and Managing Large B Cell Lymphoma Relapses after Chimeric Antigen Receptor T Cell Therapy. Biol Blood Marrow Transplant 2019; 25:e344-e351. [PMID: 31279751 DOI: 10.1016/j.bbmt.2019.06.036] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 02/06/2023]
Abstract
Most patients with large cell lymphoma are cured with frontline chemoimmunotherapy. For individuals with refractory disease and those who relapse after conventional therapies, chimeric antigen receptor (CAR) T cells are an important treatment option and have led to remissions in otherwise refractory patients. In the pivotal trials, durable responses were achieved in approximately 40% to 50% of patients treated with axicabtagene ciloleucel, tisagenlecleucel, or lisocabtagene maraleucel, indicating that many patients will require subsequent treatment. Failure after CAR T cell therapy is caused by a variety of factors that can be divided into 3 broad categories: tumor intrinsic factors, other host factors, and inadequacies of the CAR T cells. Within this framework, this article reviews possible mechanisms of treatment failures and, based on the timing of relapse, considers potential salvage therapies and opportunities for future clinical studies.
Collapse
|
16
|
Mulder TA, Wahlin BE, Österborg A, Palma M. Targeting the Immune Microenvironment in Lymphomas of B-Cell Origin: From Biology to Clinical Application. Cancers (Basel) 2019; 11:cancers11070915. [PMID: 31261914 PMCID: PMC6678966 DOI: 10.3390/cancers11070915] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023] Open
Abstract
In lymphomas of B-cell origin, cancer cells orchestrate an inflammatory microenvironment of immune and stromal cells that sustain the tumor cell survival and growth, known as a tumor microenvironment (TME). The features of the TME differ between the different lymphoma types, ranging from extremely inflammatory, such as in Hodgkin lymphoma, to anergic, leading to immune deficiency and susceptibility to infections, such as in chronic lymphocytic leukemia. Understanding the characteristic features of the TME as well as the interactions between cancer and TME cells has given insight into the pathogenesis of most lymphomas and contributed to identify novel therapeutic targets. Here, we summarize the preclinical data that contributed to clarifying the role of the immune cells in the TME of different types of lymphomas of B-cell origin, and explain how the understanding of the biological background has led to new clinical applications. Moreover, we provide an overview of the clinical results of trials that assessed the safety and efficacy of drugs directly targeting TME immune cells in lymphoma patients.
Collapse
Affiliation(s)
- Tom A Mulder
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Björn E Wahlin
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Marzia Palma
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
17
|
Mlynarczyk C, Fontán L, Melnick A. Germinal center-derived lymphomas: The darkest side of humoral immunity. Immunol Rev 2019; 288:214-239. [PMID: 30874354 PMCID: PMC6518944 DOI: 10.1111/imr.12755] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
One of the unusual features of germinal center (GC) B cells is that they manifest many hallmarks of cancer cells. Accordingly, most B-cell neoplasms originate from the GC reaction, and characteristically display abundant point mutations, structural genomic lesions, and clonal diversity from the genetic and epigenetic standpoints. The dominant biological theme of GC-derived lymphomas is mutation of genes involved in epigenetic regulation and immune receptor signaling, which come into play at critical transitional stages of the GC reaction. Hence, mechanistic studies of these mutations reveal fundamental insight into the biology of the normal and malignant GC B cell. The BCL6 transcription factor plays a central role in establishing the GC phenotype in B cells, and most lymphomas are dependent on BCL6 to maintain survival, proliferation, and perhaps immune evasion. Many lymphoma mutations have the commonality of enhancing the oncogenic functions of BCL6, or overcoming some of its tumor suppressive effects. Herein, we discuss how unique features of the GC reaction create vulnerabilities that select for particular lymphoma mutations. We examine the interplay between epigenetic programming, metabolism, signaling, and immune regulatory mechanisms in lymphoma, and discuss how these are leading to novel precision therapy strategies to treat lymphoma patients.
Collapse
Affiliation(s)
- Coraline Mlynarczyk
- Department of MedicineDivision of Hematology & Medical OncologyWeill Cornell MedicineNew York CityNew York
| | - Lorena Fontán
- Department of MedicineDivision of Hematology & Medical OncologyWeill Cornell MedicineNew York CityNew York
| | - Ari Melnick
- Department of MedicineDivision of Hematology & Medical OncologyWeill Cornell MedicineNew York CityNew York
| |
Collapse
|
18
|
de Charette M, Houot R. Hide or defend, the two strategies of lymphoma immune evasion: potential implications for immunotherapy. Haematologica 2018; 103:1256-1268. [PMID: 30006449 PMCID: PMC6068015 DOI: 10.3324/haematol.2017.184192] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
Evading immune eradication is a prerequisite for neoplastic progression and one of the hallmarks of cancer. Here, we review the different immune escape strategies of lymphoma and classify them into two main mechanisms. First, lymphoma cells may “hide” to become invisible to the immune system. This can be achieved by losing or downregulating MHC and/or molecules involved in antigen presentation (including antigen processing machinery and adhesion molecules), thereby preventing their recognition by the immune system. Second, lymphoma cells may “defend” themselves to become resistant to immune eradication. This can be achieved in several ways: by becoming resistant to apoptosis, by expressing inhibitory ligands that deactivate immune cells and/or by inducing an immunosuppressive (humoral and cellular) microenvironment. These immune escape mechanisms may have therapeutic implications. Their identification may be used to guide “personalized immunotherapy” for lymphoma.
Collapse
Affiliation(s)
| | - Roch Houot
- CHU Rennes, Service Hématologie Clinique, F-35033, France .,INSERM, U1236, F-35043, France
| |
Collapse
|
19
|
Zhou XA, Louissaint A, Wenzel A, Yang J, Martinez-Escala ME, Moy AP, Morgan EA, Paxton CN, Hong B, Andersen EF, Guitart J, Behdad A, Cerroni L, Weinstock DM, Choi J. Genomic Analyses Identify Recurrent Alterations in Immune Evasion Genes in Diffuse Large B-Cell Lymphoma, Leg Type. J Invest Dermatol 2018; 138:2365-2376. [PMID: 29857068 DOI: 10.1016/j.jid.2018.04.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 02/08/2023]
Abstract
Cutaneous diffuse large B-cell lymphomas (DLBCLs) are aggressive lymphomas with a poor prognosis. To elucidate their genetic bases, we analyzed exome sequencing of 37 cutaneous DLBCLs, including 31 DLBCLs, leg type (DLBCL-LT) and 6 cutaneous DLBCLs-not otherwise specified (DLBCL-NOS). As reported previously, 77% of DLBCL-LT harbor NF-κB-activating MYD88 mutations. In nearly all MYD88-wild-type DLBCL-LT, we found cancer-promoting mutations that either activate the NF-κB pathway through alternative genes (NFKBIE or REL) or activate other canonical cancer pathways (BRAF, MED12, PIK3R1, and STAT3). After NF-κB, the second most commonly mutated pathway putatively enables immune evasion via mutations predicted to downregulate antigen processing (B2M, CIITA, HLA) or T-cell co-stimulation (CD58). DLBCL-LT have little genetic overlap with the genetically heterogeneous DLBCL-NOS. Instead, they resemble primary central nervous system and testicular large B-cell lymphomas (primary central nervous system lymphomas and primary testicular lymphomas). Like primary central nervous system lymphomas/primary testicular lymphomas, 40% of DLBCL-LT (vs. 0% of DLBCLs-not otherwise specified) harbored PDL1/PDL2 translocations, which lead to overexpression of PD-L1 or PD-L2 in 50% of the cases. Collectively, these data broaden our understanding of cutaneous DLBCLs and suggest novel therapeutic approaches (e.g., BRAF or PI3K inhibitors). Additionally, they suggest novel treatment paradigms, wherein DLBCL-LT can be targeted with strategies (e.g., immune checkpoint blockers) currently being developed for genomically similar primary central nervous system lymphomas/primary testicular lymphomas.
Collapse
Affiliation(s)
- Xiaolong Alan Zhou
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Abner Louissaint
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alexander Wenzel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jingyi Yang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | | | - Andrea P Moy
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Elizabeth A Morgan
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christian N Paxton
- ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, Utah, USA
| | - Bo Hong
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Erica F Andersen
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Joan Guitart
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Amir Behdad
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lorenzo Cerroni
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jaehyuk Choi
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
20
|
Bandola-Simon J, Roche PA. Dysfunction of antigen processing and presentation by dendritic cells in cancer. Mol Immunol 2018; 113:31-37. [PMID: 29628265 DOI: 10.1016/j.molimm.2018.03.025] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/31/2018] [Accepted: 03/29/2018] [Indexed: 12/23/2022]
Abstract
The ability to mount an effective anti-tumor immune response requires coordinate control of CD4 T cell and CD8 T cell function by antigen presenting cells (APCs). Unfortunately, tumors create an immunosuppressive microenvironment that helps protect tumor cells from immune recognition. In many cases this defect can be traced back to a failure of APCs (most importantly dendritic cells (DCs)) to recognize, process, and present tumor antigens to T cells. In this review, we will summarize work addressing the role of different DC subsets in anti-tumor immunity and the various mechanisms used by tumor cells to suppress the ability of APCs to stimulate potent anti-tumor T cell responses.
Collapse
Affiliation(s)
- Joanna Bandola-Simon
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States.
| |
Collapse
|
21
|
Nijland M, Veenstra RN, Visser L, Xu C, Kushekhar K, van Imhoff GW, Kluin PM, van den Berg A, Diepstra A. HLA dependent immune escape mechanisms in B-cell lymphomas: Implications for immune checkpoint inhibitor therapy? Oncoimmunology 2017; 6:e1295202. [PMID: 28507804 PMCID: PMC5414870 DOI: 10.1080/2162402x.2017.1295202] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/31/2017] [Accepted: 02/08/2017] [Indexed: 02/09/2023] Open
Abstract
Antigen presentation by tumor cells in the context of Human Leukocyte Antigen (HLA) is generally considered to be a prerequisite for effective immune checkpoint inhibitor therapy. We evaluated cell surface HLA class I, HLA class II and cytoplasmic HLA-DM staining by immunohistochemistry (IHC) in 389 classical Hodgkin lymphomas (cHL), 22 nodular lymphocyte predominant Hodgkin lymphomas (NLPHL), 137 diffuse large B-cell lymphomas (DLBCL), 39 primary central nervous system lymphomas (PCNSL) and 19 testicular lymphomas. We describe a novel mechanism of immune escape in which loss of HLA-DM expression results in aberrant membranous invariant chain peptide (CLIP) expression in HLA class II cell surface positive lymphoma cells, preventing presentation of antigenic peptides. In HLA class II positive cases, HLA-DM expression was lost in 49% of cHL, 0% of NLPHL, 14% of DLBCL, 3% of PCNSL and 0% of testicular lymphomas. Considering HLA class I, HLA class II and HLA-DM together, 88% of cHL, 10% of NLPHL, 62% of DLBCL, 77% of PCNSL and 87% of testicular lymphoma cases had abnormal HLA expression patterns. In conclusion, an HLA expression pattern incompatible with normal antigen presentation is common in cHL, DLBCL, PCNSL and testicular lymphoma. Retention of CLIP in HLA class II caused by loss of HLA-DM is a novel immune escape mechanism, especially prevalent in cHL. Aberrant HLA expression should be taken into account when evaluating efficacy of checkpoint inhibitors in B-cell lymphomas.
Collapse
Affiliation(s)
- Marcel Nijland
- Department of Hematology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Rianne N Veenstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Lydia Visser
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Chuanhui Xu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Kushi Kushekhar
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Gustaaf W van Imhoff
- Department of Hematology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Philip M Kluin
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| |
Collapse
|
22
|
de Charette M, Marabelle A, Houot R. Turning tumour cells into antigen presenting cells: The next step to improve cancer immunotherapy? Eur J Cancer 2016; 68:134-147. [PMID: 27755997 DOI: 10.1016/j.ejca.2016.09.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022]
Abstract
Downregulation/loss of the antigen presentation is a major immune escape mechanism in cancer. It allows tumour cells to become 'invisible' and avoid immune attack by antitumour T cells. In tumour harbouring properties of professional antigen presenting cells (i.e. tumour B cells in lymphoma), downregulation/loss of the antigen presentation may also prevent direct priming of naïve T cells by tumour cells. Here, we review treatments that may induce/restore antigen presentation by the tumour cells. These treatments may increase the generation of antitumour T cells and/or their capacity to recognise and eliminate tumour cells. By forcing tumour cells to present their antigens, these treatments may sensitise patients to T cell-based immunotherapies, including checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Aurélien Marabelle
- Gustave Roussy, Université Paris-Saclay, Département d'Innovation Thérapeutique et d'Essais Précoces, Villejuif, F-94805, France; INSERM U1015, Villejuif, F-94805, France
| | - Roch Houot
- CHU Rennes, Service Hématologie Clinique, F-35033, Rennes, France; INSERM, U917, F-35043, Rennes, France.
| |
Collapse
|
23
|
HLA-G and MHC Class II Protein Expression in Diffuse Large B-Cell Lymphoma. Arch Immunol Ther Exp (Warsz) 2015; 64:225-40. [PMID: 26667793 DOI: 10.1007/s00005-015-0372-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 09/14/2015] [Indexed: 02/01/2023]
Abstract
The expression of human leukocyte antigen-G (HLA-G) and HLA class II protein was studied by immunohistochemical staining of lymph nodes from 148 patients with diffuse large B-cell lymphoma (DLBCL) and related to the clinical course of the disease. Negative HLA-G expression was associated with a lower probability of achieving a complete remission (p = 0.04). Patients with negative HLA-G expression tended towards a lower 3-year overall survival (OS) rate compared to those with positive expression of HLA-G (p = 0.08). When restricting the analysis to patients receiving chemotherapy with rituximab, the estimated 3-year OS rate of patients with positive HLA-G expression was 73.3 % compared with 47.5 % (p = 0.03) in those with negative expression. Patients with negative HLA class II expression presented a lower 3-year OS rate compared to subjects with positive expression (p = 0.04). The loss of HLA class II expression (p = 0.05) and belonging to the intermediate high/high IPI risk group (p = 0.001) independently increased the risk of death. HLA class II expression also retained its prognostic value in patients receiving rituximab; the 3-year OS rate was 65.3 % in patients with positive HLA class II expression versus 29.6 % (p = 0.04) in subjects that had loss of HLA class II expression. To our knowledge, for the first time, the expression of HLA-G protein in DLBCL and its association with the clinical course of the disease was demonstrated. Moreover, the link between losing HLA class II protein expression and poor survival of patients treated with immunochemotherapy was confirmed.
Collapse
|
24
|
Kridel R, Steidl C, Gascoyne RD. Tumor-associated macrophages in diffuse large B-cell lymphoma. Haematologica 2015; 100:143-5. [PMID: 25638802 DOI: 10.3324/haematol.2015.124008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Robert Kridel
- Department of Lymphoid Cancer Research, BC Cancer Agency and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Christian Steidl
- Department of Lymphoid Cancer Research, BC Cancer Agency and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Randy D Gascoyne
- Department of Lymphoid Cancer Research, BC Cancer Agency and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
25
|
Testoni M, Zucca E, Young KH, Bertoni F. Genetic lesions in diffuse large B-cell lymphomas. Ann Oncol 2015; 26:1069-1080. [PMID: 25605746 PMCID: PMC4542576 DOI: 10.1093/annonc/mdv019] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/05/2014] [Accepted: 12/15/2014] [Indexed: 01/04/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma in adults, accounting for 35%-40% of all cases. The combination of the anti-CD20 monoclonal antibody rituximab with anthracycline-based combination chemotherapy (R-CHOP, rituximab with cyclophosphamide, doxorubicin, vincristine and prednisone) lead to complete remission in most and can cure more than half of patients with DLBCL. The diversity in clinical presentation, as well as the pathologic and biologic heterogeneity, suggests that DLBCL comprises several disease entities that might ultimately benefit from different therapeutic approaches. In this review, we summarize the current literature focusing on the genetic lesions identified in DLBCL.
Collapse
Affiliation(s)
- M Testoni
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona
| | - E Zucca
- Lymphoma Unit, IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - K H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - F Bertoni
- Lymphoma and Genomics Research Program, IOR Institute of Oncology Research, Bellinzona; Lymphoma Unit, IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.
| |
Collapse
|
26
|
Cycon KA, Mulvaney K, Rimsza LM, Persky D, Murphy SP. Histone deacetylase inhibitors activate CIITA and MHC class II antigen expression in diffuse large B-cell lymphoma. Immunology 2013; 140:259-72. [PMID: 23789844 DOI: 10.1111/imm.12136] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 12/29/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL), the most common form of non-Hodgkin's lymphoma (NHL) diagnosed in the USA, consists of at least two distinct subtypes: germinal centre B (GCB) and activated B-cell (ABC). Decreased MHC class II (MHCII) expression on the tumours in both DLBCL subtypes directly correlates with significant decreases in patient survival. One common mechanism accounting for MHCII down-regulation in DLBCL is reduced expression of the MHC class II transactivator (CIITA), the master regulator of MHCII transcription. Furthermore, reduced CIITA expression in ABC DLBCL correlates with the presence of the transcriptional repressor positive regulatory domain-I-binding factor-1 (PRDI-BF1). However, the mechanisms underlying down-regulation of CIITA in GCB DLBCL are currently unclear. In this study, we demonstrate that neither PRDI-BF1 nor CpG hypermethylation at the CIITA promoters are responsible for decreased CIITA in GCB DLBCL. In contrast, histone modifications associated with an open chromatin conformation and active transcription were significantly lower at the CIITA promoters in CIITA(-) GCB cells compared with CIITA(+) B cells, which suggests that epigenetic mechanisms contribute to repression of CIITA transcription. Treatment of CIITA(-) or CIITA(low) GCB cells with several different histone deacetylase inhibitors (HDACi) activated modest CIITA and MHCII expression. However, CIITA and MHCII levels were significantly higher in these cells after exposure to the HDAC-1-specific inhibitor MS-275. These results suggest that CIITA transcription is repressed in GCB DLBCL cells through epigenetic mechanisms involving HDACs, and that HDACi treatment can alleviate repression. These observations may have important implications for patient therapy.
Collapse
|
27
|
Puvvada S, Kendrick S, Rimsza L. Molecular classification, pathway addiction, and therapeutic targeting in diffuse large B cell lymphoma. Cancer Genet 2013; 206:257-65. [PMID: 24080457 DOI: 10.1016/j.cancergen.2013.07.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 11/16/2022]
Abstract
The rapid emergence of molecularly based techniques to detect changes in the genetic landscape of diffuse large B cell lymphoma (DLBCL), including gene expression, DNA and RNA sequencing, and epigenetic profiling, has significantly influenced the understanding and therapeutic targeting of DLBCL. In this review, we briefly discuss the new methods used in the study of DLBCL. We describe the influence of the generated data on DLBCL classification and the identification of new entities and altered cell survival strategies, with a focus on the renewed interest in some classic oncogenic pathways that are currently targeted for new therapy. Finally, we examine the molecular genomic studies that revealed the importance of the tumor microenvironment in the pathogenesis of DLBCL.
Collapse
Affiliation(s)
- Soham Puvvada
- Department of Medicine, Division of Hematology-Oncology, University of Arizona, Tucson, AZ, USA.
| | | | | |
Collapse
|
28
|
Zhe S, Naqvi SAZ, Yang Y, Qi Y. Joint network and node selection for pathway-based genomic data analysis. Bioinformatics 2013; 29:1987-96. [PMID: 23749986 PMCID: PMC3722525 DOI: 10.1093/bioinformatics/btt335] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Motivation: By capturing various biochemical interactions, biological pathways provide insight into underlying biological processes. Given high-dimensional microarray or RNA-sequencing data, a critical challenge is how to integrate them with rich information from pathway databases to jointly select relevant pathways and genes for phenotype prediction or disease prognosis. Addressing this challenge can help us deepen biological understanding of phenotypes and diseases from a systems perspective. Results: In this article, we propose a novel sparse Bayesian model for joint network and node selection. This model integrates information from networks (e.g. pathways) and nodes (e.g. genes) by a hybrid of conditional and generative components. For the conditional component, we propose a sparse prior based on graph Laplacian matrices, each of which encodes detailed correlation structures between network nodes. For the generative component, we use a spike and slab prior over network nodes. The integration of these two components, coupled with efficient variational inference, enables the selection of networks as well as correlated network nodes in the selected networks. Simulation results demonstrate improved predictive performance and selection accuracy of our method over alternative methods. Based on three expression datasets for cancer study and the KEGG pathway database, we selected relevant genes and pathways, many of which are supported by biological literature. In addition to pathway analysis, our method is expected to have a wide range of applications in selecting relevant groups of correlated high-dimensional biomarkers. Availability: The code can be downloaded at www.cs.purdue.edu/homes/szhe/software.html. Contact:alanqi@purdue.edu
Collapse
Affiliation(s)
- Shandian Zhe
- Department of Computer Science, Department of Biology, and Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | |
Collapse
|
29
|
Procházka V, Jarošová M, Prouzová Z, Nedomová R, Papajík T, Indrák K. Immune Escape Mechanisms in Diffuse Large B-Cell Lymphoma. ACTA ACUST UNITED AC 2012. [DOI: 10.5402/2012/208903] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most frequent subtype of non-Hodgkin lymphomas in Western countries. Implementation of immunotherapy using monoclonal antibodies to therapeutic protocols has led to dramatic improvements in overall survival. DLBCL became a model of a successful immunochemotherapy concept. Despite this fact, there is still a proportion of patients who do not respond to or relapse early after treatment. Growing evidence suggests that host antitumor immunity is suppressed by lymphoma cells in many ways. First, host cytotoxic T cells are directly suppressed by interaction with programmed cell death (PD) ligand on lymphoma cell surface and a similar mechanism enhances the activity of suppressive regulatory T cells (Tregs). Second, tumor cells escape host cytotoxic cells due to lower immunogenicity caused by reduced expression of HLA antigens. Both mechanisms have an origin in primary genetic events in lymphomagenesis. Rearrangement of MHC class II transcriptional activator (CIITA) gene and amplification of Janus kinase (JAK2) gene lead to enhanced expression of PD ligands 1 and 2, higher proliferation activity, and lower expression of HLA. This paper summarizes current knowledge about clinically relevant immune escape mechanisms in DLBCL.
Collapse
Affiliation(s)
- V. Procházka
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, I. P. Pavlova 6, Olomouc 77520, Czech Republic
| | - M. Jarošová
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, I. P. Pavlova 6, Olomouc 77520, Czech Republic
| | - Z. Prouzová
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - R. Nedomová
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, I. P. Pavlova 6, Olomouc 77520, Czech Republic
| | - T. Papajík
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, I. P. Pavlova 6, Olomouc 77520, Czech Republic
| | - K. Indrák
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, I. P. Pavlova 6, Olomouc 77520, Czech Republic
| |
Collapse
|
30
|
Jiang Y, Soong TD, Wang L, Melnick AM, Elemento O. Genome-wide detection of genes targeted by non-Ig somatic hypermutation in lymphoma. PLoS One 2012; 7:e40332. [PMID: 22808135 PMCID: PMC3395700 DOI: 10.1371/journal.pone.0040332] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 06/07/2012] [Indexed: 12/29/2022] Open
Abstract
The processes of somatic hypermutation (SHM) and class switch recombination introduced by activation-induced cytosine deaminase (AICDA) at the Immunoglobulin (Ig) loci are key steps for creating a pool of diversified antibodies in germinal center B cells (GCBs). Unfortunately, AICDA can also accidentally introduce mutations at bystander loci, particularly within the 5′ regulatory regions of proto-oncogenes relevant to diffuse large B cell lymphomas (DLBCL). Since current methods for genomewide sequencing such as Exon Capture and RNAseq only target mutations in coding regions, to date non-Ig promoter SHMs have been studied only in a handful genes. We designed a novel approach integrating bioinformatics tools with next generation sequencing technology to identify regulatory loci targeted by SHM genome-wide. We observed increased numbers of SHM associated sequence variant hotspots in lymphoma cells as compared to primary normal germinal center B cells. Many of these SHM hotspots map to genes that have not been reported before as mutated, including BACH2, BTG2, CXCR4, CIITA, EBF1, PIM2, and TCL1A, etc., all of which have potential roles in B cell survival, differentiation, and malignant transformation. In addition, using BCL6 and BACH2 as examples, we demonstrated that SHM sites identified in these 5′ regulatory regions greatly altered their transcription activities in a reporter assay. Our approach provides a first cost-efficient, genome-wide method to identify regulatory mutations and non-Ig SHM hotspots.
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Cell Differentiation/genetics
- Cell Line, Tumor
- Chromatin Immunoprecipitation
- Gene Expression Regulation, Neoplastic
- Genes, Neoplasm/genetics
- Genes, Regulator/genetics
- Genome, Human/genetics
- Histones/metabolism
- Humans
- Lymphoma/genetics
- Lymphoma/immunology
- Lymphoma/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Methylation
- Polymorphism, Single Nucleotide/genetics
- Promoter Regions, Genetic/genetics
- Sequence Analysis, DNA
- Somatic Hypermutation, Immunoglobulin/genetics
Collapse
Affiliation(s)
- Yanwen Jiang
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - T. David Soong
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
| | - Ling Wang
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Ari M. Melnick
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail: (AM); (OE)
| | - Olivier Elemento
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail: (AM); (OE)
| |
Collapse
|
31
|
Perry AM, Mitrovic Z, Chan WC. Biological Prognostic Markers in Diffuse Large B-Cell Lymphoma. Cancer Control 2012; 19:214-26. [DOI: 10.1177/107327481201900306] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Anamarija M. Perry
- Department of Pathology and Microbiology at the Nebraska Medical Center, Omaha, Nebraska
| | - Zdravko Mitrovic
- Department of Internal Medicine, Clinical Hospital Dubrava, Zagreb, Croatia
| | - Wing C. Chan
- Department of Pathology and Microbiology at the Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
32
|
Rao S, Lana S, Eickhoff J, Marcus E, Avery P, Morley P, Avery A. Class II Major Histocompatibility Complex Expression and Cell Size Independently Predict Survival in Canine B-Cell Lymphoma. J Vet Intern Med 2011; 25:1097-105. [DOI: 10.1111/j.1939-1676.2011.0767.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
33
|
Majumder P, Boss JM. DNA methylation dysregulates and silences the HLA-DQ locus by altering chromatin architecture. Genes Immun 2011; 12:291-9. [PMID: 21326318 PMCID: PMC3107363 DOI: 10.1038/gene.2010.77] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The MHC-II locus encodes a cluster of highly polymorphic genes HLA-DR, -DQ, and -DP that are co-expressed in mature B lymphocytes. Two cell lines were established over 30 years ago from a patient diagnosed with acute lymphocytic leukemia. Laz221 represented the leukemic cells of the patient; whereas Laz388 represented the normal B cells of the patient. Whereas Laz388 expressed both HLA-DR and HLA-DQ surface and gene products, Laz221 expressed only HLA-DR genes. The discordant expression of HLA-DR and HLA-DQ genes was due to epigenetic silencing of the HLA-DQ region CTCF-binding insulators that separate the MHC-II subregions by DNA methylation. These epigenetic modifications resulted in the loss of binding of the insulator protein CTCF to the HLA-DQ flanking insulator regions and the MHC-II specific transcription factors to the HLA-DQ promoter regions. These events led to the inability of the HLA-DQ promoter regions to interact with flanking insulators that control HLA-DQ expression. Inhibition of DNA methylation by treatment with 5’deoxyazacytidine reversed each of these changes and restored expression of the HLA-DQ locus. These results highlight the consequence of disrupting an insulator within the MHC-II region and may be a normal developmental mechanism or one used by tumor cells to escape immune surveillance.
Collapse
Affiliation(s)
- P Majumder
- Department of Microbiology and Immunology, Emory University School Of Medicine, Atlanta, GA 30322, USA
| | | |
Collapse
|
34
|
Diepstra A. MHC class II as a therapeutic target in B-cell lymphomas: the CIITA road to paradise? Leuk Lymphoma 2010; 50:1740-1. [PMID: 19863170 DOI: 10.3109/10428190903350454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Arjan Diepstra
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands.
| |
Collapse
|
35
|
Wilkinson ST, Fernandez DR, Murphy SP, Braziel RM, Campo E, Chan WC, Delabie J, Gascoyne RD, Staudt LM, Jaffe ES, Rosenwald A, Rimsza LM. Decreased major histocompatibility complex class II expression in diffuse large B-cell lymphoma does not correlate with CpG methylation of class II transactivator promoters III and IV. Leuk Lymphoma 2010; 50:1875-8. [PMID: 19814686 DOI: 10.3109/10428190903297531] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
36
|
Coordinate loss of MHC class II expression in the diffuse large B cell lymphoma cell line OCI-Ly2 is due to a novel mutation in RFX-AP. Immunogenetics 2009; 62:109-16. [PMID: 20024540 DOI: 10.1007/s00251-009-0418-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 11/26/2009] [Indexed: 10/20/2022]
Abstract
Loss of major histocompatibility complex class II (MHCII) antigen expression on diffuse large B cell lymphoma (DLBCL) corresponds closely with significant decreases in patient survival. However, the mechanisms accounting for MHCII loss in DLBCL have not been thoroughly characterized to date. In this report, we demonstrate that coordinate loss of MHCII expression in OCI-Ly2 DLBCL cells is associated with an 11-base deletion in the cDNA encoding RFX-AP, one of the subunits of the heterotrimeric regulatory factor X (RFX) that is required for activating MHCII transcription. This deletion results in a frameshift in the RFX-AP protein beginning at amino acid 234 and, therefore, in the loss of C-terminal amino acids that are required for function. Stable transfection of OCI-Ly2 DLBCL cells with an expression vector for wild-type RFX-AP restores MHCII expression, which strongly suggests that the defect in RFX-AP accounts for MHCII loss in these cells.
Collapse
|