1
|
Anderson GSF, Chapman MA. T cell-redirecting therapies in hematological malignancies: Current developments and novel strategies for improved targeting. Mol Ther 2024; 32:2856-2891. [PMID: 39095991 PMCID: PMC11403239 DOI: 10.1016/j.ymthe.2024.07.028] [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: 03/25/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
T cell-redirecting therapies (TCRTs), such as chimeric antigen receptor (CAR) or T cell receptor (TCR) T cells and T cell engagers, have emerged as a highly effective treatment modality, particularly in the B and plasma cell-malignancy setting. However, many patients fail to achieve deep and durable responses; while the lack of truly unique tumor antigens, and concurrent on-target/off-tumor toxicities, have hindered the development of TCRTs for many other cancers. In this review, we discuss the recent developments in TCRT targets for hematological malignancies, as well as novel targeting strategies that aim to address these, and other, challenges.
Collapse
Affiliation(s)
| | - Michael A Chapman
- MRC Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK; Addenbrooke's Hospital, Cambridge Universities Foundation Trust, Cambridge CB2 0QQ, UK.
| |
Collapse
|
2
|
Zhang J, Xu X, Deng H, Liu L, Xiang Y, Feng J. Overcoming cancer drug-resistance calls for novel strategies targeting abnormal alternative splicing. Pharmacol Ther 2024; 261:108697. [PMID: 39025436 DOI: 10.1016/j.pharmthera.2024.108697] [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: 03/02/2024] [Revised: 05/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Abnormal gene alternative splicing (AS) events are strongly associated with cancer progression. Here, we summarize AS events that contribute to the development of drug resistance and classify them into three categories: alternative cis-splicing (ACS), alternative trans-splicing (ATS), and alternative back-splicing (ABS). The regulatory mechanisms underlying AS processes through cis-acting regulatory elements and trans-acting factors are comprehensively described, and the distinct functions of spliced variants, including linear spliced variants derived from ACS, chimeric spliced variants arising from ATS, and circRNAs generated through ABS, are discussed. The identification of dysregulated spliced variants, which contribute to drug resistance and hinder effective cancer treatment, suggests that abnormal AS processes may together serve as a precise regulatory mechanism enabling drug-resistant cancer cell survival or, alternatively, represent an evolutionary pathway for cancer cells to adapt to changes in the external environment. Moreover, this review summarizes recent advancements in treatment approaches targeting AS-associated drug resistance, focusing on cis-acting regulatory elements, trans-acting factors, and specific spliced variants. Collectively, gaining an in-depth understanding of the mechanisms underlying aberrant alternative splicing events and developing strategies to target this process hold great promise for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Xinyu Xu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Hongwei Deng
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yuancai Xiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou city, Sichuan 646000, China.
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
3
|
Bauer M, Schöbel CM, Wickenhauser C, Seliger B, Jasinski-Bergner S. Deciphering the role of alternative splicing in neoplastic diseases for immune-oncological therapies. Front Immunol 2024; 15:1386993. [PMID: 38736877 PMCID: PMC11082354 DOI: 10.3389/fimmu.2024.1386993] [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: 02/16/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Alternative splicing (AS) is an important molecular biological mechanism regulated by complex mechanisms involving a plethora of cis and trans-acting elements. Furthermore, AS is tissue specific and altered in various pathologies, including infectious, inflammatory, and neoplastic diseases. Recently developed immuno-oncological therapies include monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells targeting, among others, immune checkpoint (ICP) molecules. Despite therapeutic successes have been demonstrated, only a limited number of patients showed long-term benefit from these therapies with tumor entity-related differential response rates were observed. Interestingly, splice variants of common immunotherapeutic targets generated by AS are able to completely escape and/or reduce the efficacy of mAb- and/or CAR-based tumor immunotherapies. Therefore, the analyses of splicing patterns of targeted molecules in tumor specimens prior to therapy might help correct stratification, thereby increasing therapy success by antibody panel selection and antibody dosages. In addition, the expression of certain splicing factors has been linked with the patients' outcome, thereby highlighting their putative prognostic potential. Outstanding questions are addressed to translate the findings into clinical application. This review article provides an overview of the role of AS in (tumor) diseases, its molecular mechanisms, clinical relevance, and therapy response.
Collapse
Affiliation(s)
- Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chiara-Maria Schöbel
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
- Department of Good Manufacturing Practice (GMP) Development & Advanced Therapy Medicinal Products (ATMP) Design, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute for Medical Immunology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Simon Jasinski-Bergner
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
| |
Collapse
|
4
|
Mraz M. 5'-UTR mRNA splicing determines CD20 levels. Blood 2023; 142:1676-1678. [PMID: 37971763 DOI: 10.1182/blood.2023022305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Affiliation(s)
- Marek Mraz
- CEITEC Masaryk University and University Hospital Brno
| |
Collapse
|
5
|
Ang Z, Paruzzo L, Hayer KE, Schmidt C, Torres Diz M, Xu F, Zankharia U, Zhang Y, Soldan S, Zheng S, Falkenstein CD, Loftus JP, Yang SY, Asnani M, King Sainos P, Pillai V, Chong E, Li MM, Tasian SK, Barash Y, Lieberman PM, Ruella M, Schuster SJ, Thomas-Tikhonenko A. Alternative splicing of its 5'-UTR limits CD20 mRNA translation and enables resistance to CD20-directed immunotherapies. Blood 2023; 142:1724-1739. [PMID: 37683180 PMCID: PMC10667349 DOI: 10.1182/blood.2023020400] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/04/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Aberrant skipping of coding exons in CD19 and CD22 compromises the response to immunotherapy in B-cell malignancies. Here, we showed that the MS4A1 gene encoding human CD20 also produces several messenger RNA (mRNA) isoforms with distinct 5' untranslated regions. Four variants (V1-4) were detected using RNA sequencing (RNA-seq) at distinct stages of normal B-cell differentiation and B-lymphoid malignancies, with V1 and V3 being the most abundant. During B-cell activation and Epstein-Barr virus infection, redirection of splicing from V1 to V3 coincided with increased CD20 positivity. Similarly, in diffuse large B-cell lymphoma, only V3, but not V1, correlated with CD20 protein levels, suggesting that V1 might be translation-deficient. Indeed, the longer V1 isoform contained upstream open reading frames and a stem-loop structure, which cooperatively inhibited polysome recruitment. By modulating CD20 isoforms with splice-switching morpholino oligomers, we enhanced CD20 expression and anti-CD20 antibody rituximab-mediated cytotoxicity in a panel of B-cell lines. Furthermore, reconstitution of CD20-knockout cells with V3 mRNA led to the recovery of CD20 positivity, whereas V1-reconstituted cells had undetectable levels of CD20 protein. Surprisingly, in vitro CD20-directed chimeric antigen receptor T cells were able to kill both V3- and V1-expressing cells, but the bispecific T-cell engager mosunetuzumab was only effective against V3-expressing cells. To determine whether CD20 splicing is involved in immunotherapy resistance, we performed RNA-seq on 4 postmosunetuzumab follicular lymphoma relapses and discovered that in 2 of them, the downregulation of CD20 was accompanied by a V3-to-V1 shift. Thus, splicing-mediated mechanisms of epitope loss extend to CD20-directed immunotherapies.
Collapse
Affiliation(s)
- Zhiwei Ang
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Luca Paruzzo
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
- Division of Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Katharina E. Hayer
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Carolin Schmidt
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Manuel Torres Diz
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Feng Xu
- Division of Genomic Diagnostic, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Urvi Zankharia
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA
| | - Yunlin Zhang
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Samantha Soldan
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA
| | - Sisi Zheng
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Joseph P. Loftus
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Scarlett Y. Yang
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Mukta Asnani
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Vinodh Pillai
- Division of Hematopathology, Children's Hospital of Philadelphia, Philadelphia, PA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Emeline Chong
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Marilyn M. Li
- Division of Genomic Diagnostic, Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Hematopathology, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sarah K. Tasian
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Yoseph Barash
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Paul M. Lieberman
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
- Division of Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Stephen J. Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
- Division of Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
6
|
Ang Z, Paruzzo L, Hayer KE, Schmidt C, Torres Diz M, Xu F, Zankharia U, Zhang Y, Soldan S, Zheng S, Falkenstein CD, Loftus JP, Yang SY, Asnani M, King Sainos P, Pillai V, Chong E, Li MM, Tasian SK, Barash Y, Lieberman PM, Ruella M, Schuster SJ, Thomas-Tikhonenko A. Alternative splicing of its 5'-UTR limits CD20 mRNA translation and enables resistance to CD20-directed immunotherapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.19.529123. [PMID: 37645778 PMCID: PMC10461923 DOI: 10.1101/2023.02.19.529123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Aberrant skipping of coding exons in CD19 and CD22 compromises responses to immunotherapy for B-cell malignancies. Here, we show that the MS4A1 gene encoding human CD20 also produces several mRNA isoforms with distinct 5' untranslated regions (5'-UTR). Four variants (V1-4) were detectable by RNA-seq in distinct stages of normal B-cell differentiation and B-lymphoid malignancies, with V1 and V3 being the most abundant by far. During B-cell activation and Epstein-Barr virus infection, redirection of splicing from V1 to V3 coincided with increased CD20 positivity. Similarly, in diffuse large B-cell lymphoma only V3, but not V1, correlated with CD20 protein levels, suggesting that V1 might be translation-deficient. Indeed, the longer V1 isoform was found to contain upstream open reading frames (uORFs) and a stem-loop structure, which cooperatively inhibited polysome recruitment. By modulating CD20 isoforms with splice-switching Morpholino oligomers, we enhanced CD20 expression and anti-CD20 antibody rituximab-mediated cytotoxicity in a panel of B-cell lines. Furthermore, reconstitution of CD20-knockout cells with V3 mRNA led to the recovery of CD20 positivity, while V1-reconstituted cells had undetectable levels of CD20 protein. Surprisingly, in vitro CD20-directed CAR T cells were able to kill both V3- and V1-expressing cells, but the bispecific T cell engager mosunetuzumab was only effective against V3-expressing cells. To determine whether CD20 splicing is involved in immunotherapy resistance, we performed RNA-seq on four post-mosunetuzumab follicular lymphoma relapses and discovered that in two of them downregulation of CD20 was accompanied by the V3-to-V1 shift. Thus, splicing-mediated mechanisms of epitope loss extend to CD20-directed immunotherapies. Key Points In normal & malignant human B cells, CD20 mRNA is alternatively spliced into four 5'-UTR isoforms, some of which are translation-deficient.The balance between translation-deficient and -competent isoforms modulates CD20 protein levels & responses to CD20-directed immunotherapies. Explanation of Novelty We discovered that in normal and malignant B-cells, CD20 mRNA is alternatively spliced to generate four distinct 5'-UTRs, including the longer translation-deficient V1 variant. Cells predominantly expressing V1 were still sensitive to CD20-targeting chimeric antigen receptor T-cells. However, they were resistant to the bispecific anti-CD3/CD20 antibody mosunetuzumab, and the shift to V1 were observed in CD20-negative post-mosunetuzumab relapses of follicular lymphoma.
Collapse
|
7
|
Han N, Liu Z. Targeting alternative splicing in cancer immunotherapy. Front Cell Dev Biol 2023; 11:1232146. [PMID: 37635865 PMCID: PMC10450511 DOI: 10.3389/fcell.2023.1232146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Tumor immunotherapy has made great progress in cancer treatment but still faces several challenges, such as a limited number of targetable antigens and varying responses among patients. Alternative splicing (AS) is an essential process for the maturation of nearly all mammalian mRNAs. Recent studies show that AS contributes to expanding cancer-specific antigens and modulating immunogenicity, making it a promising solution to the above challenges. The organoid technology preserves the individual immune microenvironment and reduces the time/economic costs of the experiment model, facilitating the development of splicing-based immunotherapy. Here, we summarize three critical roles of AS in immunotherapy: resources for generating neoantigens, targets for immune-therapeutic modulation, and biomarkers to guide immunotherapy options. Subsequently, we highlight the benefits of adopting organoids to develop AS-based immunotherapies. Finally, we discuss the current challenges in studying AS-based immunotherapy in terms of existing bioinformatics algorithms and biological technologies.
Collapse
Affiliation(s)
- Nan Han
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhaoqi Liu
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
8
|
Effer B, Perez I, Ulloa D, Mayer C, Muñoz F, Bustos D, Rojas C, Manterola C, Vergara-Gómez L, Dappolonnio C, Weber H, Leal P. Therapeutic Targets of Monoclonal Antibodies Used in the Treatment of Cancer: Current and Emerging. Biomedicines 2023; 11:2086. [PMID: 37509725 PMCID: PMC10377242 DOI: 10.3390/biomedicines11072086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is one of the leading global causes of death and disease, and treatment options are constantly evolving. In this sense, the use of monoclonal antibodies (mAbs) in immunotherapy has been considered a fundamental aspect of modern cancer therapy. In order to avoid collateral damage, it is indispensable to identify specific molecular targets or biomarkers of therapy and/or diagnosis (theragnostic) when designing an appropriate immunotherapeutic regimen for any type of cancer. Furthermore, it is important to understand the currently employed mAbs in immunotherapy and their mechanisms of action in combating cancer. To achieve this, a comprehensive understanding of the biology of cancer cell antigens, domains, and functions is necessary, including both those presently utilized and those emerging as potential targets for the design of new mAbs in cancer treatment. This review aims to provide a description of the therapeutic targets utilized in cancer immunotherapy over the past 5 years, as well as emerging targets that hold promise as potential therapeutic options in the application of mAbs for immunotherapy. Additionally, the review explores the mechanisms of actin of the currently employed mAbs in immunotherapy.
Collapse
Affiliation(s)
- Brian Effer
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Isabela Perez
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Daniel Ulloa
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Carolyn Mayer
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Francisca Muñoz
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Diego Bustos
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Claudio Rojas
- Programa de Doctorado en Ciencias Médicas, Universidad de la Frontera, Temuco 4811230, Chile
- Centro de Estudios Morfológicos y Quirúrgicos de La, Universidad de La Frontera, Temuco 4811230, Chile
| | - Carlos Manterola
- Programa de Doctorado en Ciencias Médicas, Universidad de la Frontera, Temuco 4811230, Chile
- Centro de Estudios Morfológicos y Quirúrgicos de La, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis Vergara-Gómez
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Camila Dappolonnio
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Helga Weber
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Pamela Leal
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
- Department of Agricultural Sciences and Natural Resources, Faculty of Agricultural and Forestry Science, Universidad de La Frontera, Temuco 4810296, Chile
| |
Collapse
|
9
|
Feng L, Gao X, Jiao Z, Wang Z, Min F. BTK inhibitor combined with anti-PD-1 monoclonal antibody for the treatment of CD20-negative primary central nervous system lymphoma: A case report. Oncol Lett 2022; 25:48. [PMID: 36644138 PMCID: PMC9811622 DOI: 10.3892/ol.2022.13634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/11/2022] [Indexed: 12/23/2022] Open
Abstract
CD20-negative diffuse large B-cell lymphoma (DLBCL) is a rare type of lymphoproliferative disorder characterized by a high degree of aggressiveness, a tendency for extranodal invasion and chemotherapeutic resistance. CD20-negative DLBCL originating from the nervous system is rarer. In primary central nervous system lymphoma (PCNSL), >90% of cases are histologically classified as DLBCL. The present study reports the case of a 65-year-old female with CD20-negative PCNSL, whose primary clinical symptom was a persistent headache. Serum tests for human immunodeficiency virus, Epstein-Barr virus-DNA, human herpesvirus 8, hepatitis B and hepatitis C were negative. Cranial magnetic resonance imaging suggested multiple intracranial occupancies. The neoplastic cells were found to be positive for CD19, CD79α, Bcl-2 (~92%) and c-Myc (~50%), while showing negative results for CD20, CD138, programmed cell death protein 1 (PD-1) and programmed cell death receptor 1 ligand 1 (PD-L1). The Ki-67 proliferation index was >80%. In the tumor microenvironment, <10% of the tumor-associated macrophages expressed PD-L1. The number of PD-1-positive tumor-infiltrating lymphocytes was 30-40 cells according to high-power field microscopy. The patient's disease progressed during methotrexate-based treatment, leading to a change in the treatment regimen to the Bruton tyrosine kinase inhibitor, zanubrutinib, combined with the anti-PD-1 monoclonal antibody tislelizumab. After two courses of the combined treatment, the patient achieved complete remission (CR) and continued to receive consolidation treatment. In the 20 months of follow-up since CR was achieved, the patient's general condition was good and the disease was in continuous remission. The present case report and literature review show that a combination of drugs targeting different mechanisms may be used to treat PCNSL to prolong patient survival time. The mechanism of the enhanced efficacy of a combination of the two drugs may be related to the enhancement of antitumor T-cell immune responses and reversal of T-cell immune metabolic dysfunctions by the inhibition of glycolysis.
Collapse
Affiliation(s)
- Lan Feng
- Department of Hematology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Xiaohui Gao
- Department of Hematology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Zhiyun Jiao
- Department of Radiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Zheng Wang
- Department of Pathology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Fenglin Min
- Department of Hematology, Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou, Jiangsu 225012, P.R. China,Correspondence to: Dr Fenglin Min, Department of Hematology, Yangzhou Hospital of Traditional Chinese Medicine, 575 Wengchang Middle Road, Hanjiang, Yangzhou, Jiangsu 225012, P.R. China, E-mail:
| |
Collapse
|
10
|
Pavlasova G, Mraz M. The regulation and function of CD20: an "enigma" of B-cell biology and targeted therapy. Haematologica 2021; 105:1494-1506. [PMID: 32482755 PMCID: PMC7271567 DOI: 10.3324/haematol.2019.243543] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/15/2020] [Indexed: 12/26/2022] Open
Abstract
The introduction of anti-CD20 monoclonal antibodies such as rituximab, ofatumumab, or obinutuzumab improved the therapy of B-cell malignancies even though the precise physiological role and regulation of CD20 remains unclear. Furthermore, CD20 expression is highly variable between different B-cell malignancies, patients with the same malignancy, and even between intraclonal subpopulations in an individual patient. Several epigenetic (EZH2, HDAC1/2, HDAC1/4, HDAC6, complex Sin3A-HDAC1) and transcription factors (USF, OCT1/2, PU.1, PiP, ELK1, ETS1, SP1, NFκB, FOXO1, CREM, SMAD2/3) regulating CD20 expression (encoded by MS4A1) have been characterized. CD20 is induced in the context of microenvironmental interactions by CXCR4/SDF1 (CXCL12) chemokine signaling and the molecular function of CD20 has been linked to the signaling propensity of B-cell receptor (BCR). CD20 has also been shown to interact with multiple other surface proteins on B cells (such as CD40, MHCII, CD53, CD81, CD82, and CBP). Current efforts to combine anti-CD20 monoclonal antibodies with BCR signaling inhibitors targeting BTK or PI3K (ibrutinib, acalabrutinib, idelalisib, duvelisib) or BH3-mimetics (venetoclax) lead to the necessity to better understand both the mechanisms of regulation and the biological functions of CD20. This is underscored by the observation that CD20 is decreased in response to the "BCR inhibitor" ibrutinib which largely prevents its successful combination with rituximab. Several small molecules (such as histone deacetylase inhibitors, DNA methyl-transferase inhibitors, aurora kinase A/B inhibitors, farnesyltransferase inhibitors, FOXO1 inhibitors, and bryostatin-1) are being tested to upregulate cell-surface CD20 levels and increase the efficacy of anti-CD20 monoclonal antibodies. Herein, we review the current understanding of CD20 function, and the mechanisms of its regulation in normal and malignant B cells, highlighting the therapeutic implications.
Collapse
Affiliation(s)
- Gabriela Pavlasova
- Central European Institute of Technology, Masaryk University, Brno.,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marek Mraz
- Central European Institute of Technology, Masaryk University, Brno .,Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
11
|
Deng K, Yao J, Huang J, Ding Y, Zuo J. Abnormal alternative splicing promotes tumor resistance in targeted therapy and immunotherapy. Transl Oncol 2021; 14:101077. [PMID: 33774500 PMCID: PMC8039720 DOI: 10.1016/j.tranon.2021.101077] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Abnormal alternative splicing is involve in abnormal expression of genes in cancer. Abnormal alternative splicing events promote malignant progression of cancer. Abnormal alternative splicing develops tumor resistance to targeted therapy by changing the target point and signal transduction pathway. Abnormal alternative splicing develops tumor resistance to immunotherapy by changing cell surface antigens and protein structure.
Abnormally alternative splicing events are common hallmark of diverse types of cancers. Splicing variants with aberrant functions play an important role in cancer development. Most importantly, a growing body of evidence has supported that alternative splicing might play a significant role in the therapeutic resistance of tumors. Targeted therapy and immunotherapy are the future directions of tumor therapy; however, the loss of antigen targets on the tumor cells surface and alterations in drug efficacy have resulted in the failure of targeted therapy and immunotherapy. Interestingly, abnormal alternative splicing, as a strategy to regulate gene expression, is reportedly involved in the reprogramming of cell signaling pathways and epitopes on the tumor cell surface by changing splicing patterns of genes, thus rendering tumors resisted to targeted therapy and immunotherapy. Accordingly, increased knowledge regarding abnormal alternative splicing in tumors may help predict therapeutic resistance during targeted therapy and immunotherapy and lead to novel therapeutic approaches in cancer. Herein, we provide a brief synopsis of abnormal alternative splicing events in cancer progression and therapeutic resistance.
Collapse
Affiliation(s)
- Kun Deng
- The Laboratory of translational medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang, Hunan 421001, P R China
| | - Jingwei Yao
- The Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421002, P R China
| | - Jialu Huang
- The Laboratory of translational medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang, Hunan 421001, P R China
| | - Yubo Ding
- The Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421002, P R China
| | - Jianhong Zuo
- The Laboratory of translational medicine, Hengyang Medical School, University of South China, 28 Changsheng Road, Hengyang, Hunan 421001, P R China; The Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421002, P R China; Clinical Laboratory, The Third Affiliated Hospital of University of South China, Hengyang, Hunan 421900, China.
| |
Collapse
|
12
|
Omics-wide quantitative B-cell infiltration analyses identify GPR18 for human cancer prognosis with superiority over CD20. Commun Biol 2020; 3:234. [PMID: 32398659 PMCID: PMC7217858 DOI: 10.1038/s42003-020-0964-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor-infiltrating B lymphocyte (TIL-B), and TIL-B-related biomarkers have clinical prognostic values for human cancers. CD20 (encoded by MS4A1) is a widely used TIL-B biomarker. Using TCGA-quantitative multiomics datasets, we first cross-compare prognostic powers of intratumoral CD20 protein, mRNA and TIL-B levels in pan-cancers. Here, we show that MS4A1 and TIL-B are consistently prognostic in 5 cancers (head and neck, lung, cervical, kidney and low-grade glioma), while unexpectedly, CD20 protein levels lack quantitative correlations with MS4A1/TIL-B levels and demonstrate limited prognosticity. Subsequent bioinformatics discovery for TIL-B prognostic gene identifies a single gene, GPR18 with stand-alone prognosticity across 9 cancers (superior over CD20), with further validations in multiple non-TCGA cohorts. GPR18's immune signature denotes major B-cell-T-cell interactions, with its intratumoral expression strongly tied to a "T-cell active", likely cytolytic, status across human cancers, suggesting its functional link to cytolytic T-cell activity in cancer. GPR18 merits biological and clinical utility assessments over CD20.
Collapse
|
13
|
Panjwani MK, Atherton MJ, MaloneyHuss MA, Haran KP, Xiong A, Gupta M, Kulikovsaya I, Lacey SF, Mason NJ. Establishing a model system for evaluating CAR T cell therapy using dogs with spontaneous diffuse large B cell lymphoma. Oncoimmunology 2019; 9:1676615. [PMID: 32002286 PMCID: PMC6959441 DOI: 10.1080/2162402x.2019.1676615] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/29/2019] [Indexed: 11/19/2022] Open
Abstract
Multiple rodent and primate preclinical studies have advanced CAR T cells into the clinic. However, no single model accurately reflects the challenges of effective CAR T therapy in human cancer patients. To evaluate the effectiveness of next-generation CAR T cells that aim to overcome barriers to durable tumor elimination, we developed a system to evaluate CAR T cells in pet dogs with spontaneous cancer. Here we report on this system and the results of a pilot trial using CAR T cells to treat canine diffuse large B cell lymphoma (DLBCL). We designed and manufactured CD20-targeting, second-generation canine CAR T cells for functional evaluation in vitro and in vivo using lentivectors to parallel human CAR T cell manufacturing. A first-in-species trial of five dogs with DLBCL treated with CAR T was undertaken. Canine CAR T cells functioned in an antigen-specific manner and killed CD20+ targets. Circulating CAR T cells were detectable post-infusion, however, induction of canine anti-mouse antibodies (CAMA) was associated with CAR T cell loss. Specific selection pressure on CD20+ tumors was observed following CAR T cell therapy, culminating in antigen escape and emergence of CD20-disease. Patient survival times correlated with ex vivo product expansion. Altering product manufacturing improved transduction efficiency and skewed toward a memory-like phenotype of canine CAR T cells. Manufacturing of functional canine CAR T cells using a lentivector is feasible. Comparable challenges to effective CAR T cell therapy exist, indicating their relevance in informing future human clinical trial design.
Collapse
Affiliation(s)
- M Kazim Panjwani
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew J Atherton
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martha A MaloneyHuss
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kumudhini P Haran
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ailian Xiong
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Minnal Gupta
- Translational and Correlative Studies Laboratory (TCSL), Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irina Kulikovsaya
- Translational and Correlative Studies Laboratory (TCSL), Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simon F Lacey
- Translational and Correlative Studies Laboratory (TCSL), Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicola J Mason
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Milcent B, Josseaume N, Riller Q, Giglioli I, Rabia E, Deligne C, Latouche JB, Hamieh M, Couture A, Toutirais O, Lone YC, Jeger-Madiot R, Graff-Dubois S, Amorim S, Loiseau P, Toubert A, Brice P, Thieblemont C, Teillaud JL, Sibéril S. Presence of T cells directed against CD20-derived peptides in healthy individuals and lymphoma patients. Cancer Immunol Immunother 2019; 68:1561-1572. [PMID: 31494742 PMCID: PMC6805815 DOI: 10.1007/s00262-019-02389-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
Preclinical and clinical studies have suggested that cancer treatment with antitumor antibodies induces a specific adaptive T cell response. A central role in this process has been attributed to CD4+ T cells, but the relevant T cell epitopes, mostly derived from non-mutated self-antigens, are largely unknown. In this study, we have characterized human CD20-derived epitopes restricted by HLA-DR1, HLA-DR3, HLA-DR4, and HLA-DR7, and investigated whether T cell responses directed against CD20-derived peptides can be elicited in human HLA-DR-transgenic mice and human samples. Based on in vitro binding assays to recombinant human MHC II molecules and on in vivo immunization assays in H-2 KO/HLA-A2+-DR1+ transgenic mice, we have identified 21 MHC II-restricted long peptides derived from intracellular, membrane, or extracellular domains of the human non-mutated CD20 protein that trigger in vitro IFN-γ production by PBMCs and splenocytes from healthy individuals and by PBMCs from follicular lymphoma patients. These CD20-derived MHC II-restricted peptides could serve as a therapeutic tool for improving and/or monitoring anti-CD20 T cell activity in patients treated with rituximab or other anti-CD20 antibodies.
Collapse
Affiliation(s)
- Benoit Milcent
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Nathalie Josseaume
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Quentin Riller
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Ilenia Giglioli
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Emilia Rabia
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Claire Deligne
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Jean-Baptiste Latouche
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Mohamad Hamieh
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Alexandre Couture
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Olivier Toutirais
- Unicaen, Inserm 1237, Physiopathology and Imaging of Neurological Disorders, Normandie University, Caen, France.,French Blood Service (Etablissement Français du Sang, EFS), Caen, France
| | - Yu-Chun Lone
- Inserm U1014, Hôpital Paul Brousse, Villejuif, France
| | - Raphaël Jeger-Madiot
- Inserm U1135, CNRS ERL8255, Center for Immunology and Microbial Infection, Paris, France
| | - Stéphanie Graff-Dubois
- Inserm U1135, CNRS ERL8255, Center for Immunology and Microbial Infection, Paris, France
| | - Sandy Amorim
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France
| | - Pascale Loiseau
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot-Paris 7, Paris, France
| | - Antoine Toubert
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot-Paris 7, Paris, France
| | - Pauline Brice
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France
| | - Catherine Thieblemont
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France.,EA7324 Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Luc Teillaud
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France.,Laboratory "Immune Microenvironment and Immunotherapy", Sorbonne Université UMRS 1135, INSERM U.1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Sophie Sibéril
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France. .,Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, 15 rue de l'Ecole de Médecine, 75006, Paris, France.
| |
Collapse
|
15
|
Scialdone A, Hasni MS, Damm JK, Lennartsson A, Gullberg U, Drott K. The HDAC inhibitor valproate induces a bivalent status of the CD20 promoter in CLL patients suggesting distinct epigenetic regulation of CD20 expression in CLL in vivo. Oncotarget 2018; 8:37409-37422. [PMID: 28445158 PMCID: PMC5514918 DOI: 10.18632/oncotarget.16964] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/08/2017] [Indexed: 11/25/2022] Open
Abstract
Treatment with anti-CD20 antibodies is only moderately efficient in chronic lymphocytic leukemia (CLL), a feature which has been explained by the inherently low CD20 expression in CLL. It has been shown that CD20 is epigenetically regulated and that histone deacetylase inhibitors (HDACis) can increase CD20 expression in vitro in CLL. To assess whether HDACis can upregulate CD20 also in vivo in CLL, the HDACi valproate was given to three del13q/NOTCH1wt CLL patients and CD20 levels were analysed (the PREVAIL study). Valproate treatment resulted in expected global activating histone modifications suggesting HDAC inhibitory effects. However, although valproate induced expression of CD20 mRNA and protein in the del13q/NOTCH1wt I83-E95 CLL cell line, no such effects were observed in the patients studied. In contrast to the cell line, in patients valproate treatment resulted in transient recruitment of the transcriptional repressor EZH2 to the CD20 promoter, correlating to an increase of the repressive histone mark H3K27me3. This suggests that valproate-mediated induction of CD20 may be hampered by EZH2 mediated H3K27me3 in vivo in CLL. Moreover, valproate treatment resulted in induction of EZH2 and global H3K27me3 in patient cells, suggesting transcriptionally repressive effects of valproate in CLL. Our results suggest new in vivo mechanisms of HDACis which may have implications on the design of future clinical trials in B-cell malignancies.
Collapse
Affiliation(s)
- Annarita Scialdone
- Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | | | - Jesper Kofoed Damm
- Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Urban Gullberg
- Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Kristina Drott
- Department of Hematology and Transfusion Medicine, Lund University, Lund, Sweden.,Clinic of Oncology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
16
|
Magne J, Jenvrin A, Chauchet A, Casasnovas O, Donzel A, Jego L, Aral B, Guy J, Nadal N, Vernerey D, Callier P, Garnache-Ottou F, Ferrand C. Potential added value of a RT-qPCR method of SOX 11 expression, in the context of a multidisciplinary diagnostic assessment of B cell malignancies. Exp Hematol Oncol 2018; 7:5. [PMID: 29484276 PMCID: PMC5819690 DOI: 10.1186/s40164-018-0097-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/02/2018] [Indexed: 01/24/2023] Open
Abstract
Background Expression of SRY [sex-determining region Y]-box11 (SOX11) is specific to mantle cell lymphoma (MCL) and contributes, in conjunction with immunoglobulin variable heavy chain gene mutation status, to the identification of two forms of this disease. Methods The aim of this report was firstly, to design an easy and suitable RT-qPCR method to quantify SOX11 mRNA expression in mantle cell lymphoma and other B cell malignancies with the proper reference gene; secondly, to define the best threshold of relative quantity of SOX11 mRNA in order to reach the best compromise between sensitivity and specificity. Results For best discrimination of MCL and non-MCL groups we determined an area under the curve (AUC) of 0.9750 and a threshold of 1.76 with 100% sensitivity and 88% specificity. AUC and threshold values of respectively 0.91/1.346 [87% sensitivity, 80% specificity] and 0.9525/1.7120 [100% sensitivity, 88% specificity] for GAPDH and RPLP0 respectively denote that the RPLP0 reference gene alone is sufficient for PCR housekeeping gene. Conclusion This work describes an RT-qPCR assay for SOX11 expression in order to better characterize MCL at diagnosis. Further studies on larger cohorts are needed to evaluate this molecular tool, especially for the follow-up of minimal residual disease. Electronic supplementary material The online version of this article (10.1186/s40164-018-0097-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julien Magne
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | - Alizée Jenvrin
- INSERM, EFS BFC, UMR1098, Univ. Bourgogne Franche-Comté, Besançon, 25000 France
| | - Adrien Chauchet
- 3Department of Hematology, University Hospital of Besancon, 25000 Besançon, France
| | - Olivier Casasnovas
- 4Department of Hematology, University Hospital of Dijon, 21000 Dijon, France
| | - Anne Donzel
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | - Laurence Jego
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | - Bernard Aral
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | - Julien Guy
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | - Nathalie Nadal
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | - Dewi Vernerey
- Univ. Bourgogne Franche-Comté, EA 3181, 25000 Besançon, France
| | - Patrick Callier
- 1INSERM, UMR866, Faculté des Sciences de Santé, Univ. Bourgogne Franche-Comté, 21000 Dijon, France
| | | | - Christophe Ferrand
- INSERM, EFS BFC, UMR1098, Univ. Bourgogne Franche-Comté, Besançon, 25000 France.,Laboratoire de Thérapeutique Immuno-Moléculaire et cellulaire des cancers, INSERM UMR1098, Etablissement Français du Sang Bourgogne/Franche-Comté, 8, rue du Docteur Jean-François-Xavier Girod, 25020 Besançon, France
| |
Collapse
|
17
|
Katchi T, Liu D. Diagnosis and treatment of CD20 negative B cell lymphomas. Biomark Res 2017; 5:5. [PMID: 28191314 PMCID: PMC5297138 DOI: 10.1186/s40364-017-0088-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/03/2017] [Indexed: 01/25/2023] Open
Abstract
CD20 negative B cell non-Hodgkin lymphoma (NHL) is rare and accounts for approximately 1-2% of B cell lymphomas. CD20- negative NHL is frequently associated with extranodal involvement, atypical morphology, aggressive clinical behaviour, resistance to standard chemotherapy and poor prognosis. The most common types of these include plasmablastic lymphoma, primary effusion lymphoma, large B-cell lymphoma arising from HHV8-associated multicentric Castleman’s disease, and ALK+ large B cell lymphoma. This review provides an overview of the diagnostic and treatment modalities for CD20 negative B cell NHL.
Collapse
Affiliation(s)
- Tasleem Katchi
- Division of Hematology & Oncology, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| | - Delong Liu
- Division of Hematology & Oncology, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| |
Collapse
|
18
|
Gamonet C, Bole-Richard E, Delherme A, Aubin F, Toussirot E, Garnache-Ottou F, Godet Y, Ysebaert L, Tournilhac O, Dartigeas C, Larosa F, Deconinck E, Saas P, Borg C, Deschamps M, Ferrand C. Erratum to: New CD20 alternative splice variants: molecular identification and differential expression within hematological B cell malignancies. Exp Hematol Oncol 2015; 5:10. [PMID: 27081577 PMCID: PMC4831155 DOI: 10.1186/s40164-016-0038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 03/30/2016] [Indexed: 12/03/2022] Open
Affiliation(s)
- Clémentine Gamonet
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France
| | - Elodie Bole-Richard
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France
| | - Aurélia Delherme
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France
| | - François Aubin
- EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France
| | - Eric Toussirot
- EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France ; CHRU, Department of Rheumatology, Université de Franche-Comté EA 4266, INSERM CIC-1431, 25000 Besançon, France ; EA 4266, Université de Franche-Comté, Besançon, France
| | - Francine Garnache-Ottou
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France
| | - Yann Godet
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France
| | - Loïc Ysebaert
- Inserm U1037, Université Toulouse 3-ERL CNRS, CHU Purpan, Toulouse, France
| | - Olivier Tournilhac
- Hématologie Clinique, CHU Estaing, 1 Place Lucie Aubrac, 63003 Clermont-Ferrand Cedex 1, France
| | | | - Fabrice Larosa
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; Hematology Department, CHU Jean Minjoz, 25020 Besançon, France
| | - Eric Deconinck
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France ; Hematology Department, CHU Jean Minjoz, 25020 Besançon, France
| | - Philippe Saas
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France
| | - Christophe Borg
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; EA3181 et Service de Dermatologie, Université de Franche Comté, CHU de Besançon, Besançon, France
| | - Marina Deschamps
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France
| | - Christophe Ferrand
- INSERM UMR1098, Établissement Français du Sang Bourgogne Franche Comté, Université de Franche-Comté, SFR FED4234, 25020 Besançon, France ; Laboratoire de Thérapeutique Immuno-Moléculaire et cellulaire des cancers, INSERM UMR1098, Etablissement Français du Sang-Bourgogne/Franche-Comté, 8, rue du Docteur Jean-François-Xavier Girod, 25020 Besançon Cedex, France
| |
Collapse
|