1
|
Elbaz O, Shaat RM, Abd El Ghaffar HA, Shamaa S, Abdel-Masseih HM, Anber N, Mortada MI. The Prevalence of MYD88 L265P and TNFAIP3 Mutations and Their Correlations with Clinico-Hematological Profile in Egyptian Patients with Diffuse Large B Cell Lymphoma. Asian Pac J Cancer Prev 2023; 24:2485-2491. [PMID: 37505783 PMCID: PMC10676502 DOI: 10.31557/apjcp.2023.24.7.2485] [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: 04/11/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) is characterized by chronic active B-cell receptor signaling and a constitutive activation of the NF-KB pathway. MYD88 L265P mutation occurs as a driving force of NF-KB overactivity in ABC-DLBCL. Nonetheless, in cases of DLBCL, the MYD88 L265P mutation has not yet been investigated in association with the tumour necrosis factor alpha induced protein3 (TNFAIP3) mutation. OBJECTIVE To investigate the frequency of MYD88 and TNFAIP3 mutations in DLBCL and their association to the clinico-hematological profile. MATERIAL AND METHODS We used real-time polymerase chain reaction in order to search for MYD88 L265P and TNFAIP3 mutations in 100 DLBCL patients. RESULTS MYD88 L265P In 20% of cases, the CT heterozygous genotype was discovered. CT heterozygous genotype was more common in ABC type, stage IV, greater IPI groups, extra-nodal infiltration, and BM infiltration. It was also linked to a shorter OS. TNFAIP3 mutation GA heterozygous genotype was detected in 18% of the patients, with ABC-DLBCL subtype accounting for 77.8%. The GA heterozygous genotype was usually related with stage IV, extranodal infiltration, and a reduced life expectancy. CONCLUSION MYD88 L265P and to lesser extent TNFAIP3 mutations are major mutations in ABC- DLBCL and may be predictive factors for poor OS in ABC- DLBCL patients.
Collapse
Affiliation(s)
- Osama Elbaz
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Rana M. Shaat
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Hasan A. Abd El Ghaffar
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Sameh Shamaa
- Medical Oncology Unit, Department of Internal Medicine, Oncology Center, Faculty of Medicine, Mansoura University, Egypt.
| | - Hanaa M. Abdel-Masseih
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Nahla Anber
- Assistant Consultant of Biochemistry, Emergency Hospital, Mansoura University, Egypt.
| | - Metwaly Ibrahim Mortada
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| |
Collapse
|
2
|
Turi M, Anilkumar Sithara A, Hofmanová L, Žihala D, Radhakrishnan D, Vdovin A, Knápková S, Ševčíková T, Chyra Z, Jelínek T, Šimíček M, Gullà A, Anderson KC, Hájek R, Hrdinka M. Transcriptome Analysis of Diffuse Large B-Cell Lymphoma Cells Inducibly Expressing MyD88 L265P Mutation Identifies Upregulated CD44, LGALS3, NFKBIZ, and BATF as Downstream Targets of Oncogenic NF-κB Signaling. Int J Mol Sci 2023; 24:ijms24065623. [PMID: 36982699 PMCID: PMC10057398 DOI: 10.3390/ijms24065623] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
During innate immune responses, myeloid differentiation primary response 88 (MyD88) functions as a critical signaling adaptor protein integrating stimuli from toll-like receptors (TLR) and the interleukin-1 receptor (IL-1R) family and translates them into specific cellular outcomes. In B cells, somatic mutations in MyD88 trigger oncogenic NF-κB signaling independent of receptor stimulation, which leads to the development of B-cell malignancies. However, the exact molecular mechanisms and downstream signaling targets remain unresolved. We established an inducible system to introduce MyD88 to lymphoma cell lines and performed transcriptomic analysis (RNA-seq) to identify genes differentially expressed by MyD88 bearing the L265P oncogenic mutation. We show that MyD88L265P activates NF-κB signaling and upregulates genes that might contribute to lymphomagenesis, including CD44, LGALS3 (coding Galectin-3), NFKBIZ (coding IkBƺ), and BATF. Moreover, we demonstrate that CD44 can serve as a marker of the activated B-cell (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) and that CD44 expression is correlated with overall survival in DLBCL patients. Our results shed new light on the downstream outcomes of MyD88L265P oncogenic signaling that might be involved in cellular transformation and provide novel therapeutical targets.
Collapse
Affiliation(s)
- Marcello Turi
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Anjana Anilkumar Sithara
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Lucie Hofmanová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - David Žihala
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Dhwani Radhakrishnan
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Alexander Vdovin
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Sofija Knápková
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Tereza Ševčíková
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Zuzana Chyra
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Michal Šimíček
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Annamaria Gullà
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Kenneth Carl Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Matouš Hrdinka
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
- Correspondence:
| |
Collapse
|
3
|
Mitchell S, Tsui R, Tan ZC, Pack A, Hoffmann A. The NF-κB multidimer system model: A knowledge base to explore diverse biological contexts. Sci Signal 2023; 16:eabo2838. [PMID: 36917644 PMCID: PMC10195159 DOI: 10.1126/scisignal.abo2838] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023]
Abstract
The nuclear factor κB (NF-κB) system is critical for various biological functions in numerous cell types, including the inflammatory response, cell proliferation, survival, differentiation, and pathogenic responses. Each cell type is characterized by a subset of 15 NF-κB dimers whose activity is regulated in a stimulus-responsive manner. Numerous studies have produced different mathematical models that account for cell type-specific NF-κB activities. However, whereas the concentrations or abundances of NF-κB subunits may differ between cell types, the biochemical interactions that constitute the NF-κB signaling system do not. Here, we synthesized a consensus mathematical model of the NF-κB multidimer system, which could account for the cell type-specific repertoires of NF-κB dimers and their cell type-specific activation and cross-talk. Our review demonstrates that these distinct cell type-specific properties of NF-κB signaling can be explained largely as emergent effects of the cell type-specific expression of NF-κB monomers. The consensus systems model represents a knowledge base that may be used to gain insights into the control and function of NF-κB in diverse physiological and pathological scenarios and that describes a path for generating similar regulatory knowledge bases for other pleiotropic signaling systems.
Collapse
Affiliation(s)
- Simon Mitchell
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, CA 90095, USA
- Brighton and Sussex Medical School, Department of Clinical and Experimental Medicine, University of Sussex, Falmer, East Sussex, BN1 9PX, UK
| | - Rachel Tsui
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Zhixin Cyrillus Tan
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, CA 90095, USA
| | - Arran Pack
- Brighton and Sussex Medical School, Department of Clinical and Experimental Medicine, University of Sussex, Falmer, East Sussex, BN1 9PX, UK
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Venturutti L, Rivas MA, Pelzer BW, Flümann R, Hansen J, Karagiannidis I, Xia M, McNally DR, Isshiki Y, Lytle A, Teater M, Chin CR, Meydan C, Knittel G, Ricker E, Mason CE, Ye X, Pan-Hammarström Q, Steidl C, Scott DW, Reinhardt HC, Pernis AB, Béguelin W, Melnick AM. An Aged/Autoimmune B-cell Program Defines the Early Transformation of Extranodal Lymphomas. Cancer Discov 2023; 13:216-243. [PMID: 36264161 PMCID: PMC9839622 DOI: 10.1158/2159-8290.cd-22-0561] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/26/2022] [Accepted: 10/17/2022] [Indexed: 01/17/2023]
Abstract
A third of patients with diffuse large B-cell lymphoma (DLBCL) present with extranodal dissemination, which is associated with inferior clinical outcomes. MYD88L265P is a hallmark extranodal DLBCL mutation that supports lymphoma proliferation. Yet extranodal lymphomagenesis and the role of MYD88L265P in transformation remain mostly unknown. Here, we show that B cells expressing Myd88L252P (MYD88L265P murine equivalent) activate, proliferate, and differentiate with minimal T-cell costimulation. Additionally, Myd88L252P skewed B cells toward memory fate. Unexpectedly, the transcriptional and phenotypic profiles of B cells expressing Myd88L252P, or other extranodal lymphoma founder mutations, resembled those of CD11c+T-BET+ aged/autoimmune memory B cells (AiBC). AiBC-like cells progressively accumulated in animals prone to develop lymphomas, and ablation of T-BET, the AiBC master regulator, stripped mouse and human mutant B cells of their competitive fitness. By identifying a phenotypically defined prospective lymphoma precursor population and its dependencies, our findings pave the way for the early detection of premalignant states and targeted prophylactic interventions in high-risk patients. SIGNIFICANCE Extranodal lymphomas feature a very poor prognosis. The identification of phenotypically distinguishable prospective precursor cells represents a milestone in the pursuit of earlier diagnosis, patient stratification, and prophylactic interventions. Conceptually, we found that extranodal lymphomas and autoimmune disorders harness overlapping pathogenic trajectories, suggesting these B-cell disorders develop and evolve within a spectrum. See related commentary by Leveille et al. (Blood Cancer Discov 2023;4:8-11). This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Leandro Venturutti
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC V5Z1L3, Canada., Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z1L3, Canada., Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z7, Canada.,Corresponding authors: Leandro Venturutti, PhD. Centre for Lymphoid Cancer and Terry Fox Laboratory, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada. Phone: 604-675-8000; Fax: 604-877-0712; , Ari M. Melnick, MD. Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, 413 E 69th St, New York, NY, 10021, USA. Phone: 646-962-6725; Fax: 646-962-0576;
| | - Martin A. Rivas
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Benedikt W. Pelzer
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA., Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Faculty of Medicine and University Hospital of Cologne, Cologne D-50937, Germany
| | - Ruth Flümann
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50931, Germany., Max-Planck-Institute for Biology of Aging, Cologne 50931, Germany
| | - Julia Hansen
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50931, Germany., Max-Planck-Institute for Biology of Aging, Cologne 50931, Germany
| | - Ioannis Karagiannidis
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Min Xia
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Dylan R. McNally
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yusuke Isshiki
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Andrew Lytle
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC V5Z1L3, Canada
| | - Matt Teater
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Christopher R. Chin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA., Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA., The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and the WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA., The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and the WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Gero Knittel
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital of Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Edd Ricker
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Christopher E. Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA., The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and the WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xiaofei Ye
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Stockholm, Sweden
| | - Qiang Pan-Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Stockholm, Sweden
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC V5Z1L3, Canada., Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z7, Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC V5Z1L3, Canada., Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z7, Canada., Department of Medicine, University of British Columbia, Vancouver, BC V6T1Z7, Canada
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital of Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Alessandra B. Pernis
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Wendy Béguelin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ari M. Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA.,Corresponding authors: Leandro Venturutti, PhD. Centre for Lymphoid Cancer and Terry Fox Laboratory, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada. Phone: 604-675-8000; Fax: 604-877-0712; , Ari M. Melnick, MD. Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, 413 E 69th St, New York, NY, 10021, USA. Phone: 646-962-6725; Fax: 646-962-0576;
| |
Collapse
|
5
|
Pindzola GM, Razzaghi R, Tavory RN, Nguyen HT, Morris VM, Li M, Agarwal S, Huang B, Okada T, Reinhardt HC, Knittel G, Kashkar H, Young RM, Pittaluga S, Muppidi JR. Aberrant expansion of spontaneous splenic germinal centers induced by hallmark genetic lesions of aggressive lymphoma. Blood 2022; 140:1119-1131. [PMID: 35759728 PMCID: PMC9461474 DOI: 10.1182/blood.2022015926] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/18/2022] [Indexed: 11/20/2022] Open
Abstract
Unique molecular vulnerabilities have been identified in the aggressive MCD/C5 genetic subclass of diffuse large B-cell lymphoma (DLBCL). However, the premalignant cell-of-origin exhibiting MCD-like dependencies remains elusive. In this study, we examined animals carrying up to 4 hallmark genetic lesions found in MCD consisting of gain-of-function mutations in Myd88 and Cd79b, loss of Prdm1, and overexpression of BCL2. We discovered that expression of combinations of these alleles in vivo promoted a cell-intrinsic accumulation of B cells in spontaneous splenic germinal centers (GCs). As with MCD, these premalignant B cells were enriched for B-cell receptors (BCRs) with evidence of self-reactivity, displayed a de novo dependence on Tlr9, and were more sensitive to inhibition of Bruton's tyrosine kinase. Mutant spontaneous splenic GC B cells (GCB) showed increased proliferation and IRF4 expression. Mice carrying all 4 genetic lesions showed a >50-fold expansion of spontaneous splenic GCs exhibiting aberrant histologic features with a dark zone immunophenotype and went on to develop DLBCL in the spleen with age. Thus, by combining multiple hallmark genetic alterations associated with MCD, our study identifies aberrant spontaneous splenic GCBs as a likely cell-of-origin for this aggressive genetic subtype of lymphoma.
Collapse
Affiliation(s)
- Grace M Pindzola
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Raud Razzaghi
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rachel N Tavory
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hang T Nguyen
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Vivian M Morris
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Biology, Johns Hopkins University, Baltimore, MD
| | - Moyi Li
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Shreya Agarwal
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Bonnie Huang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Takaharu Okada
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hans C Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Gero Knittel
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Hamid Kashkar
- Institute for Molecular Immunology, Center for Molecular Medicine Cologne (CMMC), CECAD Research Center, Faculty of Medicine University Hospital Cologne, University of Cologne, Cologne, Germany; and
| | - Ryan M Young
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jagan R Muppidi
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
6
|
Alcoceba M, García-Álvarez M, Medina A, Maldonado R, González-Calle V, Chillón MC, Sarasquete ME, González M, García-Sanz R, Jiménez C. MYD88 Mutations: Transforming the Landscape of IgM Monoclonal Gammopathies. Int J Mol Sci 2022; 23:5570. [PMID: 35628381 PMCID: PMC9141891 DOI: 10.3390/ijms23105570] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023] Open
Abstract
The MYD88 gene has a physiological role in the innate immune system. Somatic mutations in MYD88, including the most common L265P, have been associated with the development of certain types of lymphoma. MYD88L265P is present in more than 90% of patients with Waldenström's macroglobulinemia (WM) and IgM monoclonal gammopathy of undetermined significance (IgM-MGUS). The absence of MYD88 mutations in WM patients has been associated with a higher risk of transformation into aggressive lymphoma, resistance to certain therapies (BTK inhibitors), and shorter overall survival. The MyD88 signaling pathway has also been used as a target for specific therapies. In this review, we summarize the clinical applications of MYD88 testing in the diagnosis, prognosis, follow-up, and treatment of patients. Although MYD88L265P is not specific to WM, few tumors present a single causative mutation in a recurrent position. The role of the oncogene in the pathogenesis of WM is still unclear, especially considering that the mutation can be found in normal B cells of patients, as recently reported. This may have important implications for early lymphoma detection in healthy elderly individuals and for the treatment response assessment based on a MYD88L265P analysis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.A.); (M.G.-Á.); (A.M.); (R.M.); (V.G.-C.); (M.C.C.); (M.E.S.); (M.G.); (C.J.)
| | | |
Collapse
|
7
|
Drandi D, Decruyenaere P, Ferrante M, Offner F, Vandesompele J, Ferrero S. Nucleic Acid Biomarkers in Waldenström Macroglobulinemia and IgM-MGUS: Current Insights and Clinical Relevance. Diagnostics (Basel) 2022; 12:diagnostics12040969. [PMID: 35454017 PMCID: PMC9028641 DOI: 10.3390/diagnostics12040969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 12/13/2022] Open
Abstract
Waldenström Macroglobulinemia (WM) is an indolent lymphoplasmacytic lymphoma, characterized by the production of excess immunoglobulin M monoclonal protein. WM belongs to the spectrum of IgM gammopathies, ranging from asymptomatic IgM monoclonal gammopathy of undetermined significance (IgM-MGUS), through IgM-related disorders and asymptomatic WM to symptomatic WM. In recent years, its complex genomic and transcriptomic landscape has been extensively explored, hereby elucidating the biological mechanisms underlying disease onset, progression and therapy response. An increasing number of mutations, cytogenetic abnormalities, and molecular signatures have been described that have diagnostic, phenotype defining or prognostic implications. Moreover, cell-free nucleic acid biomarkers are increasingly being investigated, benefiting the patient in a minimally invasive way. This review aims to provide an extensive overview of molecular biomarkers in WM and IgM-MGUS, considering current shortcomings, as well as potential future applications in a precision medicine approach.
Collapse
Affiliation(s)
- Daniela Drandi
- Department of Molecular Biotechnology and Health Sciences, Hematology Division, University of Torino, 10126 Torino, Italy; (M.F.); (S.F.)
- Correspondence: (D.D.); (P.D.)
| | - Philippe Decruyenaere
- Department of Hematology, Ghent University Hospital, 9000 Ghent, Belgium;
- OncoRNALab, Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Correspondence: (D.D.); (P.D.)
| | - Martina Ferrante
- Department of Molecular Biotechnology and Health Sciences, Hematology Division, University of Torino, 10126 Torino, Italy; (M.F.); (S.F.)
| | - Fritz Offner
- Department of Hematology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Jo Vandesompele
- OncoRNALab, Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Simone Ferrero
- Department of Molecular Biotechnology and Health Sciences, Hematology Division, University of Torino, 10126 Torino, Italy; (M.F.); (S.F.)
| |
Collapse
|
8
|
Kaushal A, Nooka AK, Carr AR, Pendleton KE, Barwick BG, Manalo J, McCachren SS, Gupta VA, Joseph NS, Hofmeister CC, Kaufman JL, Heffner LT, Ansell SM, Boise LH, Lonial S, Dhodapkar KM, Dhodapkar MV. Aberrant Extrafollicular B Cells, Immune Dysfunction, Myeloid Inflammation, and MyD88-Mutant Progenitors Precede Waldenstrom Macroglobulinemia. Blood Cancer Discov 2021; 2:600-615. [PMID: 34778800 PMCID: PMC8580616 DOI: 10.1158/2643-3230.bcd-21-0043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/07/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
Waldenstrom macroglobulinemia (WM) and its precursor IgM gammopathy are distinct disorders characterized by clonal mature IgM-expressing B-cell outgrowth in the bone marrow. Here, we show by high-dimensional single-cell immunogenomic profiling of patient samples that these disorders originate in the setting of global B-cell compartment alterations, characterized by expansion of genomically aberrant extrafollicular B cells of the nonmalignant clonotype. Alterations in the immune microenvironment preceding malignant clonal expansion include myeloid inflammation and naïve B- and T-cell depletion. Host response to these early lesions involves clone-specific T-cell immunity that may include MYD88 mutation-specific responses. Hematopoietic progenitors carry the oncogenic MYD88 mutations characteristic of the malignant WM clone. These data support a model for WM pathogenesis wherein oncogenic alterations and signaling in progenitors, myeloid inflammation, and global alterations in extrafollicular B cells create the milieu promoting extranodal pattern of growth in differentiated malignant cells. SIGNIFICANCE These data provide evidence that growth of the malignant clone in WM is preceded by expansion of extrafollicular B cells, myeloid inflammation, and immune dysfunction in the preneoplastic phase. These changes may be related in part to MYD88 oncogenic signaling in pre-B progenitor cells and suggest a novel model for WM pathogenesis. This article is highlighted in the In This Issue feature, p. 549.
Collapse
Affiliation(s)
- Akhilesh Kaushal
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | - Ajay K. Nooka
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Allison R. Carr
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | - Katherine E. Pendleton
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | | | - Julia Manalo
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia
| | - Samuel S. McCachren
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Vikas A. Gupta
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Nisha S. Joseph
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Craig C. Hofmeister
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jonathan L. Kaufman
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Leonard T. Heffner
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | - Lawrence H. Boise
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Sagar Lonial
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Kavita M. Dhodapkar
- Winship Cancer Institute, Emory University, Atlanta, Georgia.,Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.,Corresponding Authors: Madhav V. Dhodapkar, Winship Cancer Institute, Emory University, 1364 Clifton Road NE, Atlanta, GA 30322. E-mail: ; and Kavita M. Dhodapkar,
| | - Madhav V. Dhodapkar
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.,Corresponding Authors: Madhav V. Dhodapkar, Winship Cancer Institute, Emory University, 1364 Clifton Road NE, Atlanta, GA 30322. E-mail: ; and Kavita M. Dhodapkar,
| |
Collapse
|
9
|
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
|
10
|
Ouk C, Roland L, Gachard N, Poulain S, Oblet C, Rizzo D, Saintamand A, Lemasson Q, Carrion C, Thomas M, Balabanian K, Espéli M, Parrens M, Soubeyran I, Boulin M, Faumont N, Feuillard J, Vincent-Fabert C. Continuous MYD88 Activation Is Associated With Expansion and Then Transformation of IgM Differentiating Plasma Cells. Front Immunol 2021; 12:641692. [PMID: 34017329 PMCID: PMC8129569 DOI: 10.3389/fimmu.2021.641692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/14/2021] [Indexed: 11/19/2022] Open
Abstract
Activating mutations of MYD88 (MYD88L265P being the far most frequent) are found in most cases of Waldenström macroglobulinemia (WM) as well as in various aggressive B-cell lymphoma entities with features of plasma cell (PC) differentiation, such as activated B-cell type diffuse large B-cell lymphoma (DLBCL). To understand how MYD88 activation exerts its transformation potential, we developed a new mouse model in which the MYD88L252P protein, the murine ortholog of human MYD88L265P, is continuously expressed in CD19 positive B-cells together with the Yellow Fluorescent Protein (Myd88L252P mice). In bone marrow, IgM B and plasma cells were expanded with a CD138 expression continuum from IgMhigh CD138low to IgMlow CD138high cells and the progressive loss of the B220 marker. Serum protein electrophoresis (SPE) longitudinal analysis of 40 Myd88L252P mice (16 to 56 weeks old) demonstrated that ageing was first associated with serum polyclonal hyper gammaglobulinemia (hyper Ig) and followed by a monoclonal immunoglobulin (Ig) peak related to a progressive increase in IgM serum levels. All Myd88L252P mice exhibited spleen enlargement which was directly correlated with the SPE profile and was maximal for monoclonal Ig peaks. Myd88L252P mice exhibited very early increased IgM PC differentiation. Most likely due to an early increase in the Ki67 proliferation index, IgM lymphoplasmacytic (LP) and plasma cells continuously expanded with age being first associated with hyper Ig and then with monoclonal Ig peak. This peak was consistently associated with a spleen LP-like B-cell lymphoma. Clonal expression of both membrane and secreted µ chain isoforms was demonstrated at the mRNA level by high throughput sequencing. The Myd88L252P tumor transcriptomic signature identified both proliferation and canonical NF-κB p65/RelA activation. Comparison with MYD88L265P WM showed that Myd88L252P tumors also shared the typical lymphoplasmacytic transcriptomic signature of WM bone marrow purified tumor B-cells. Altogether these results demonstrate for the first time that continuous MYD88 activation is specifically associated with clonal transformation of differentiating IgM B-cells. Since MYD88L252P targets the IgM PC differentiation continuum, it provides an interesting preclinical model for development of new therapeutic approaches to both WM and aggressive MYD88 associated DLBCLs.
Collapse
Affiliation(s)
- Catherine Ouk
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Lilian Roland
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Nathalie Gachard
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Stéphanie Poulain
- UMR CANTHER « CANcer Heterogeneity, Plasticity and Resistance to THERapies » INSERM 1277-CNRS 9020 UMRS 12, University of Lille, Hematology Laboratory, Biology and Pathology Center, CHU de Lille, Lille, France
| | - Christelle Oblet
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - David Rizzo
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Alexis Saintamand
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Quentin Lemasson
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Claire Carrion
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Morgane Thomas
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Karl Balabanian
- Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, University of Paris, Paris, France
| | - Marion Espéli
- Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, University of Paris, Paris, France
| | - Marie Parrens
- Pathology Department, Hospital University Center of Bordeaux, Bordeaux, France
| | | | - Mélanie Boulin
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Nathalie Faumont
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Jean Feuillard
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| | - Christelle Vincent-Fabert
- UMR CNRS 7276/INSERM U1262 CRIBL, University of Limoges, and Hematology Laboratory of Dupuytren Hospital University Center (CHU) of Limoges, Limoges, France
| |
Collapse
|
11
|
Yu X, Li W, Deng Q, Liu H, Wang X, Hu H, Cao Y, Xu-Monette ZY, Li L, Zhang M, Lu Z, Young KH, Li Y. MYD88 L265P elicits mutation-specific ubiquitination to drive NF-κB activation and lymphomagenesis. Blood 2021; 137:1615-1627. [PMID: 33025009 PMCID: PMC7995293 DOI: 10.1182/blood.2020004918] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 09/10/2020] [Indexed: 01/03/2023] Open
Abstract
Myeloid differentiation primary response protein 88 (MYD88) is a critical universal adapter that transduces signaling from Toll-like and interleukin receptors to downstream nuclear factor-κB (NF-κB). MYD88L265P (leucine changed to proline at position 265) is a gain-of-function mutation that occurs frequently in B-cell malignancies such as Waldenstrom macroglobulinemia. In this study, E3 ligase RING finger protein family 138 (RNF138) catalyzed K63-linked nonproteolytic polyubiquitination of MYD88L265P, resulting in enhanced recruitment of interleukin-1 receptor-associated kinases and elevated NF-κB activation. However, RNF138 had little effect on wild-type MYD88 (MYD88WT). With either RNF138 knockdown or mutation on MYD88 ubiquitination sites, MYD88L265P did not constitutively activate NF-κB. A20, a negative regulator of NF-κB signaling, mediated K48-linked polyubiquitination of RNF138 for proteasomal degradation. Depletion of A20 further augmented MYD88L265P-mediated NF-κB activation and lymphoma growth. Furthermore, A20 expression correlated negatively with RNF138 expression and NF-κB activation in lymphomas with MYD88L265P and in those without. Strikingly, RNF138 expression correlated positively with NF-κB activation in lymphomas with MYD88L265P, but not in those without it. Our study revealed a novel mutation-specific biochemical reaction that drives B-cell oncogenesis, providing a therapeutic opportunity for targeting oncogenic MYD88L265P, while sparing MYD88WT, which is critical to innate immunity.
Collapse
Affiliation(s)
- Xinfang Yu
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Wei Li
- Department of Medicine, Baylor College of Medicine, Houston, TX
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qipan Deng
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Haidan Liu
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xu Wang
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Hui Hu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Medical Laboratory, Central Hospital of Wuhan, Wuhan, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Zijun Y Xu-Monette
- Division of Hematopathology, Department of Pathology, Duke University Medical Center, Durham, NC
| | - Ling Li
- Department of Oncology, the First Affiliated Hospital, Zhengzhou University, Zhenzhou, China; and
- Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, the First Affiliated Hospital, Zhengzhou University, Zhenzhou, China; and
- Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Zhongxin Lu
- Department of Medical Laboratory, Central Hospital of Wuhan, Wuhan, China
| | - Ken H Young
- Division of Hematopathology, Department of Pathology, Duke University Medical Center, Durham, NC
| | - Yong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
12
|
LUBAC accelerates B-cell lymphomagenesis by conferring resistance to genotoxic stress on B cells. Blood 2021; 136:684-697. [PMID: 32325488 DOI: 10.1182/blood.2019002654] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
The linear ubiquitin chain assembly complex (LUBAC) is a key regulator of NF-κB signaling. Activating single-nucleotide polymorphisms of HOIP, the catalytic subunit of LUBAC, are enriched in patients with activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL), and expression of HOIP, which parallels LUBAC activity, is elevated in ABC-DLBCL samples. Thus, to clarify the precise roles of LUBAC in lymphomagenesis, we generated a mouse model with augmented expression of HOIP in B cells. Interestingly, augmented HOIP expression facilitated DLBCL-like B-cell lymphomagenesis driven by MYD88-activating mutation. The developed lymphoma cells partly shared somatic gene mutations with human DLBCLs, with increased frequency of a typical AID mutation pattern. In vitro analysis revealed that HOIP overexpression protected B cells from DNA damage-induced cell death through NF-κB activation, and analysis of the human DLBCL database showed that expression of HOIP positively correlated with gene signatures representing regulation of apoptosis signaling, as well as NF-κB signaling. These results indicate that HOIP facilitates lymphomagenesis by preventing cell death and augmenting NF-κB signaling, leading to accumulation of AID-mediated mutations. Furthermore, a natural compound that specifically inhibits LUBAC was shown to suppress the tumor growth in a mouse transplantation model. Collectively, our data indicate that LUBAC is crucially involved in B-cell lymphomagenesis through protection against DNA damage-induced cell death and is a suitable therapeutic target for B-cell lymphomas.
Collapse
|
13
|
Schmidt K, Sack U, Graf R, Winkler W, Popp O, Mertins P, Sommermann T, Kocks C, Rajewsky K. B-Cell-Specific Myd88 L252P Expression Causes a Premalignant Gammopathy Resembling IgM MGUS. Front Immunol 2020; 11:602868. [PMID: 33343574 PMCID: PMC7747680 DOI: 10.3389/fimmu.2020.602868] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/19/2020] [Indexed: 12/20/2022] Open
Abstract
A highly recurrent somatic L265P mutation in the TIR domain of the signaling adapter MYD88 constitutively activates NF-κB. It occurs in nearly all human patients with Waldenström’s macroglobulinemia (WM), a B cell malignancy caused by IgM-expressing cells. Here, we introduced an inducible leucine to proline point mutation into the mouse Myd88 locus, at the orthologous position L252P. When the mutation was introduced early during B cell development, B cells developed normally. However, IgM-expressing plasma cells accumulated with age in spleen and bone, leading to more than 20-fold elevated serum IgM titers. When introduced into germinal center B cells in the context of an immunization, the Myd88L252P mutation caused prolonged persistence of antigen-specific serum IgM and elevated numbers of antigen-specific IgM plasma cells. Myd88L252P-expressing B cells switched normally, but plasma cells expressing other immunoglobulin isotypes did not increase in numbers, implying that IgM expression may be required for the observed cellular expansion. In order to test whether the Myd88L252P mutation can cause clonal expansions, we introduced it into a small fraction of CD19-positive B cells. In this scenario, five out of five mice developed monoclonal IgM serum paraproteins accompanied by an expansion of clonally related plasma cells that expressed mostly hypermutated VDJ regions. Taken together, our data suggest that the Myd88L252P mutation is sufficient to promote aberrant survival and expansion of IgM-expressing plasma cells which in turn can cause IgM monoclonal gammopathy of undetermined significance (MGUS), the premalignant condition that precedes WM.
Collapse
Affiliation(s)
- Kristin Schmidt
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ulrike Sack
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Robin Graf
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Wiebke Winkler
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Popp
- Proteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Philipp Mertins
- Proteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Sommermann
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christine Kocks
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Transgenics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Klaus Rajewsky
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
14
|
Sezaki M, Hayashi Y, Wang Y, Johansson A, Umemoto T, Takizawa H. Immuno-Modulation of Hematopoietic Stem and Progenitor Cells in Inflammation. Front Immunol 2020; 11:585367. [PMID: 33329562 PMCID: PMC7732516 DOI: 10.3389/fimmu.2020.585367] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Lifelong blood production is maintained by bone marrow (BM)-residing hematopoietic stem cells (HSCs) that are defined by two special properties: multipotency and self-renewal. Since dysregulation of either may lead to a differentiation block or extensive proliferation causing dysplasia or neoplasia, the genomic integrity and cellular function of HSCs must be tightly controlled and preserved by cell-intrinsic programs and cell-extrinsic environmental factors of the BM. The BM had been long regarded an immune-privileged organ shielded from immune insults and inflammation, and was thereby assumed to provide HSCs and immune cells with a protective environment to ensure blood and immune homeostasis. Recently, accumulating evidence suggests that hemato-immune challenges such as autoimmunity, inflammation or infection elicit a broad spectrum of immunological reactions in the BM, and in turn, influence the function of HSCs and BM environmental cells. Moreover, in analogy with the emerging concept of “trained immunity”, certain infection-associated stimuli are able to train HSCs and progenitors to produce mature immune cells with enhanced responsiveness to subsequent challenges, and in some cases, form an inflammatory or infectious memory in HSCs themselves. In this review, we will introduce recent findings on HSC and hematopoietic regulation upon exposure to various hemato-immune stimuli and discuss how these challenges can elicit either beneficial or detrimental outcomes on HSCs and the hemato-immune system, as well as their relevance to aging and hematologic malignancies.
Collapse
Affiliation(s)
- Maiko Sezaki
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Yoshikazu Hayashi
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Yuxin Wang
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Alban Johansson
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Terumasa Umemoto
- Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
15
|
Flümann R, Rehkämper T, Nieper P, Pfeiffer P, Holzem A, Klein S, Bhatia S, Kochanek M, Kisis I, Pelzer BW, Ahlert H, Hauer J, da Palma Guerreiro A, Ryan JA, Reimann M, Riabinska A, Wiederstein J, Krüger M, Deckert M, Altmüller J, Klatt AR, Frenzel LP, Pasqualucci L, Béguelin W, Melnick AM, Sander S, Montesinos-Rongen M, Brunn A, Lohneis P, Büttner R, Kashkar H, Borkhardt A, Letai A, Persigehl T, Peifer M, Schmitt CA, Reinhardt HC, Knittel G. An Autochthonous Mouse Model of Myd88- and BCL2-Driven Diffuse Large B-cell Lymphoma Reveals Actionable Molecular Vulnerabilities. Blood Cancer Discov 2020; 2:70-91. [PMID: 33447829 DOI: 10.1158/2643-3230.bcd-19-0059] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Based on gene expression profiles, diffuse large B cell lymphoma (DLBCL) is sub-divided into germinal center B cell-like (GCB) and activated B cell-like (ABC) DLBCL. Two of the most common genomic aberrations in ABC-DLBCL are mutations in MYD88, as well as BCL2 copy number gains. Here, we employ immune phenotyping, RNA-Seq and whole exome sequencing to characterize a Myd88 and Bcl2-driven mouse model of ABC-DLBCL. We show that this model resembles features of human ABC-DLBCL. We further demonstrate an actionable dependence of our murine ABC-DLBCL model on BCL2. This BCL2 dependence was also detectable in human ABC-DLBCL cell lines. Moreover, human ABC-DLBCLs displayed increased PD-L1 expression, compared to GCB-DLBCL. In vivo experiments in our ABC-DLBCL model showed that combined venetoclax and RMP1-14 significantly increased the overall survival of lymphoma bearing animals, indicating that this combination may be a viable option for selected human ABC-DLBCL cases harboring MYD88 and BCL2 aberrations.
Collapse
Affiliation(s)
- Ruth Flümann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tim Rehkämper
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Pascal Nieper
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Pauline Pfeiffer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Alessandra Holzem
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sebastian Klein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Pathology, Cologne, Germany
| | - Sanil Bhatia
- Heinrich Heine University Düsseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, Düsseldorf, Germany
| | - Moritz Kochanek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ilmars Kisis
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Benedikt W Pelzer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Heinz Ahlert
- Heinrich Heine University Düsseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, Düsseldorf, Germany
| | - Julia Hauer
- Department of Pediatrics, Pediatric Hematology and Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Alexandra da Palma Guerreiro
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jeremy A Ryan
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Maurice Reimann
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumor Immunology, and Molekulares Krebsforschungszentrum - MKFZ, Virchow Campus, Berlin, Germany
| | - Arina Riabinska
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Janica Wiederstein
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Marcus Krüger
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Martina Deckert
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Neuropathology, Cologne, Germany
| | - Janine Altmüller
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Andreas R Klatt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Clinical Chemistry, Cologne, Germany
| | - Lukas P Frenzel
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Laura Pasqualucci
- Department of Pathology and Cell Biology, Institute for Cancer Genetics and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, USA
| | - Wendy Béguelin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, USA
| | - Ari M Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, USA
| | - Sandrine Sander
- Adaptive Immunity and Lymphoma Group, German Cancer Research Center/National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Manuel Montesinos-Rongen
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Neuropathology, Cologne, Germany
| | - Anna Brunn
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Neuropathology, Cologne, Germany
| | - Philipp Lohneis
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Pathology, Cologne, Germany
| | - Reinhard Büttner
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Pathology, Cologne, Germany
| | - Hamid Kashkar
- Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Arndt Borkhardt
- Heinrich Heine University Düsseldorf, Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, Düsseldorf, Germany
| | - Anthony Letai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Thorsten Persigehl
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Radiology and Interventional Radiology, Cologne, Germany
| | - Martin Peifer
- Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,University of Cologne, Department of Translational Genomics, Cologne, Germany
| | - Clemens A Schmitt
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumor Immunology, and Molekulares Krebsforschungszentrum - MKFZ, Virchow Campus, Berlin, Germany.,Kepler Universitätsklinikum, Medical Department of Hematology and Oncology, Johannes Kepler University, Linz, Austria
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - Gero Knittel
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic I of Internal Medicine, Cologne, Germany.,Center for Integrated Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
16
|
Chakrabarti S, Visweswariah SS. Intramacrophage ROS Primes the Innate Immune System via JAK/STAT and Toll Activation. Cell Rep 2020; 33:108368. [PMID: 33176146 PMCID: PMC7662148 DOI: 10.1016/j.celrep.2020.108368] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/10/2019] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Tissue injury is one of the most severe environmental perturbations for a living organism. When damage occurs in adult Drosophila, there is a local response of the injured tissue and a coordinated action across different tissues to help the organism overcome the deleterious effect of an injury. We show a change in the transcriptome of hemocytes at the site of tissue injury, with pronounced activation of the Toll signaling pathway. We find that induction of the cytokine upd-3 and Toll receptor activation occur in response to injury alone, in the absence of a pathogen. Intracellular accumulation of hydrogen peroxide in hemocytes is essential for upd-3 induction and is facilitated by the diffusion of hydrogen peroxide through a channel protein Prip. Importantly, hemocyte activation and production of reactive oxygen species (ROS) at the site of a sterile injury provide protection to flies on subsequent infection, demonstrating training of the innate immune system.
Collapse
Affiliation(s)
- Sveta Chakrabarti
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India.
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
17
|
Visco C, Tanasi I, Quaglia FM, Ferrarini I, Fraenza C, Krampera M. Oncogenic Mutations of MYD88 and CD79B in Diffuse Large B-Cell Lymphoma and Implications for Clinical Practice. Cancers (Basel) 2020; 12:E2913. [PMID: 33050534 PMCID: PMC7600909 DOI: 10.3390/cancers12102913] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin's lymphoma in adults. Despite the recognition of transcriptional subtypes with distinct functional characteristics, patient outcomes have not been substantially altered since the advent of chemoimmunotherapy (CIT) twenty years ago. Recently, a few pivotal studies added to the disease heterogeneity by describing several activating mutations, which have been associated with disease presentation, B-cell function and behavior, and final outcome. DLBCL arises from antigen exposed B-cells, with the B-cell receptor (BCR) playing a central role. BCR-activity related mutations, such as CD79B and MYD88, are responsible for chronic activation of the BCR in a substantial subset of patients. These mutations, often coexisting in the same patient, have been found in a substantial subset of patients with immune-privileged (IP) sites DLBCLs, and are drivers of lymphoma development conferring tissue-specific homing properties. Both mutations have been associated with disease behavior, including tumor response either to CIT or to BCR-targeted therapy. The recognition of CD79B and MYD88 mutations will contribute to the heterogeneity of the disease, both in recognizing the BCR as a potential therapeutic target and in providing genetic tools for personalized treatment.
Collapse
Affiliation(s)
- Carlo Visco
- Correspondence: (C.V.); (I.T.); Tel.: +39-0458124797 (C.V.); +39-0458128418 (I.T.)
| | - Ilaria Tanasi
- Correspondence: (C.V.); (I.T.); Tel.: +39-0458124797 (C.V.); +39-0458128418 (I.T.)
| | | | | | | | | |
Collapse
|
18
|
Human MYD88L265P is insufficient by itself to drive neoplastic transformation in mature mouse B cells. Blood Adv 2020; 3:3360-3374. [PMID: 31698464 DOI: 10.1182/bloodadvances.2019000588] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022] Open
Abstract
MYD88 L265P is the most common mutation in lymphoplasmacytic lymphoma/Waldenström macroglobulinemia (LPL/WM) and one of the most frequent in poor-prognosis subtypes of diffuse large B-cell lymphoma (DLBCL). Although inhibition of the mutated MYD88 pathway has an adverse impact on LPL/WM and DLBCL cell survival, its role in lymphoma initiation remains to be clarified. We show that in mice, human MYD88L265P promotes development of a non-clonal, low-grade B-cell lymphoproliferative disorder with several clinicopathologic features that resemble human LPL/WM, including expansion of lymphoplasmacytoid cells, increased serum immunoglobulin M (IgM) concentration, rouleaux formation, increased number of mast cells in the bone marrow, and proinflammatory signaling that progresses sporadically to clonal, high-grade DLBCL. Murine findings regarding differences in the pattern of MYD88 staining and immune infiltrates in the bone marrows of MYD88 wild-type (MYD88WT) and MYD88L265P mice are recapitulated in the human setting, which provides insight into LPL/WM pathogenesis. Furthermore, histologic transformation to DLBCL is associated with acquisition of secondary genetic lesions frequently seen in de novo human DLBCL as well as LPL/WM-transformed cases. These findings indicate that, although the MYD88L265P mutation might be indispensable for the LPL/WM phenotype, it is insufficient by itself to drive malignant transformation in B cells and relies on other, potentially targetable cooperating genetic events for full development of lymphoma.
Collapse
|
19
|
Lymphoplasmacytic lymphoma in a patient with Birt-Hogg-Dubé syndrome. Int J Hematol 2020; 112:864-870. [PMID: 32789566 DOI: 10.1007/s12185-020-02970-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/28/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
Birt-Hogg-Dubé (BHD) syndrome is an autosomal dominant disease characterized by benign skin hamartomas, pulmonary cysts leading to spontaneous pneumothorax, and an increased risk of renal cancer. BHD syndrome is caused by germline mutations in the folliculin (FLCN) gene, a putative tumor suppressor, which result in loss of function of the folliculin protein and may cause cancer predisposition. In a 45-year-old woman with anemia, lymphadenopathy, and a history of recurrent spontaneous pneumothorax, 18F-FDG PET/CT detected diffuse and slight 18F-FDG accumulation in the bone marrow, enlarged spleen, and systemic multiple enlarged lymph nodes. Genetic examination identified a germline nonsense mutation [c.998C > G (p.Ser333*)] on exon 9 of FLCN. Pathological examination of the lymph node revealed a diffuse neoplastic proliferation of plasmacytoid lymphocytes. The neoplastic lymphoid cells were positive for CD20, CD138, and light chain kappa as per immunohistochemistry and mRNA in situ hybridization, and a MYD88 gene mutation [c.755T > C (p.L252P)] was identified. Accordingly, she was diagnosed with lymphoplasmacytic lymphoma concomitant with BHD syndrome. To the best of our knowledge, this is the first report describing the development of hematological malignancy in a patient with BHD syndrome. The FLCN mutation might contribute lymphomagenesis as an additional mutation cooperating with the MYD88 mutation.
Collapse
|
20
|
Shrimpton J, Care MA, Carmichael J, Walker K, Evans P, Evans C, de Tute R, Owen R, Tooze RM, Doody GM. TLR-mediated activation of Waldenström macroglobulinemia B cells reveals an uncoupling from plasma cell differentiation. Blood Adv 2020; 4:2821-2836. [PMID: 32574366 PMCID: PMC7322944 DOI: 10.1182/bloodadvances.2019001279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Waldenström macroglobulinemia (WM) is a rare malignancy in which clonal B cells infiltrate the bone marrow and give rise to a smaller compartment of neoplastic plasma cells that secrete monoclonal immunoglobulin M paraprotein. Recent studies into underlying mutations in WM have enabled a much greater insight into the pathogenesis of this lymphoma. However, there is considerably less characterization of the way in which WM B cells differentiate and how they respond to immune stimuli. In this study, we assess WM B-cell differentiation using an established in vitro model system. Using T-cell-dependent conditions, we obtained CD138+ plasma cells from WM samples with a frequency similar to experiments performed with B cells from normal donors. Unexpectedly, a proportion of the WM B cells failed to upregulate CD38, a surface marker that is normally associated with plasmablast transition and maintained as the cells proceed with differentiation. In normal B cells, concomitant Toll-like receptor 7 (TLR7) activation and B-cell receptor cross-linking drives proliferation, followed by differentiation at similar efficiency to CD40-mediated stimulation. In contrast, we found that, upon stimulation with TLR7 agonist R848, WM B cells failed to execute the appropriate changes in transcriptional regulators, identifying an uncoupling of TLR signaling from the plasma cell differentiation program. Provision of CD40L was sufficient to overcome this defect. Thus, the limited clonotypic WM plasma cell differentiation observed in vivo may result from a strict requirement for integrated activation.
Collapse
Affiliation(s)
- Jennifer Shrimpton
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
| | - Matthew A Care
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
| | - Jonathan Carmichael
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
| | - Kieran Walker
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
| | - Paul Evans
- Haematological Malignancy Diagnostic Service, St James's Institute of Oncology, Leeds, United Kingdom
| | - Charlotte Evans
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
- Haematological Malignancy Diagnostic Service, St James's Institute of Oncology, Leeds, United Kingdom
| | - Ruth de Tute
- Haematological Malignancy Diagnostic Service, St James's Institute of Oncology, Leeds, United Kingdom
| | - Roger Owen
- Haematological Malignancy Diagnostic Service, St James's Institute of Oncology, Leeds, United Kingdom
| | - Reuben M Tooze
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
- Haematological Malignancy Diagnostic Service, St James's Institute of Oncology, Leeds, United Kingdom
| | - Gina M Doody
- Division of Haematology and Immunology, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom; and
| |
Collapse
|
21
|
Nfkbie-deficiency leads to increased susceptibility to develop B-cell lymphoproliferative disorders in aged mice. Blood Cancer J 2020; 10:38. [PMID: 32170099 PMCID: PMC7070037 DOI: 10.1038/s41408-020-0305-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant NF-κB activation is a hallmark of most B-cell malignancies. Recurrent inactivating somatic mutations in the NFKBIE gene, which encodes IκBε, an inhibitor of NF-κB-inducible activity, are reported in several B-cell malignancies with highest frequencies in chronic lymphocytic leukemia and primary mediastinal B-cell lymphoma, and account for a fraction of NF-κB pathway activation. The impact of NFKBIE deficiency on B-cell development and function remains, however, largely unknown. Here, we show that Nfkbie-deficient mice exhibit an amplification of marginal zone B cells and an expansion of B1 B-cell subsets. In germinal center (GC)-dependent immune response, Nfkbie deficiency triggers expansion of GC B-cells through increasing cell proliferation in a B-cell autonomous manner. We also show that Nfkbie deficiency results in hyperproliferation of a B1 B-cell subset and leads to increased NF-κB activation in these cells upon Toll-like receptor stimulation. Nfkbie deficiency cooperates with mutant MYD88 signaling and enhances B-cell proliferation in vitro. In aged mice, Nfkbie absence drives the development of an oligoclonal indolent B-cell lymphoproliferative disorders, resembling monoclonal B-cell lymphocytosis. Collectively, these findings shed light on an essential role of IκBε in finely tuning B-cell development and function.
Collapse
|
22
|
Miao Y, Medeiros LJ, Li Y, Li J, Young KH. Genetic alterations and their clinical implications in DLBCL. Nat Rev Clin Oncol 2019; 16:634-652. [PMID: 31127191 DOI: 10.1038/s41571-019-0225-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a highly heterogeneous lymphoid neoplasm with variations in gene expression profiles and genetic alterations, which lead to substantial variations in clinical course and response to therapy. The advent of high-throughput genome sequencing platforms, and especially whole-exome sequencing, has helped to define the genetic landscape of DLBCL. In the past 10 years, these studies have identified many genetic alterations in DLBCL, some of which are specific to B cell lymphomas, whereas others can also be observed in other types of cancer. These aberrations result in altered activation of a wide range of signalling pathways and other cellular processes, including those involved in B cell differentiation, B cell receptor signalling, activation of the NF-κB pathway, apoptosis and epigenetic regulation. Further elaboration of the genetics of DLBCL will not only improve our understanding of disease pathogenesis but also provide further insight into disease classification, prognostication and therapeutic targets. In this Review, we describe the current understanding of the prevalence and causes of specific genetic alterations in DLBCL and their role in disease development and progression. We also summarize the available clinical data on therapies designed to target the aberrant pathways driven by these alterations.
Collapse
Affiliation(s)
- Yi Miao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
23
|
Zammit NW, Walters SN, Seeberger KL, O'Connell PJ, Korbutt GS, Grey ST. A20 as an immune tolerance factor can determine islet transplant outcomes. JCI Insight 2019; 4:131028. [PMID: 31581152 DOI: 10.1172/jci.insight.131028] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/25/2019] [Indexed: 01/05/2023] Open
Abstract
Islet transplantation can restore lost glycemic control in type 1 diabetes subjects but is restricted in its clinical application by a limiting supply of islets and the need for heavy immune suppression to prevent rejection. TNFAIP3, encoding the ubiquitin editing enzyme A20, regulates the activation of immune cells by raising NF-κB signaling thresholds. Here, we show that increasing A20 expression in allogeneic islet grafts resulted in permanent survival for ~45% of recipients, and > 80% survival when combined with subtherapeutic rapamycin. Allograft survival was dependent upon Tregs and was antigen specific, and grafts showed reduced expression of inflammatory factors. Transplantation of islets with A20 containing a loss-of-function variant (I325N) resulted in increased RIPK1 ubiquitination and NF-κB signaling, graft hyperinflammation, and acute allograft rejection. Overexpression of A20 in human islets potently reduced expression of inflammatory mediators, with no impact on glucose-stimulated insulin secretion. Therapeutic administration of A20 raises inflammatory signaling thresholds to favor immune tolerance and promotes islet allogeneic survival. Clinically, this would allow for reduced immunosuppression and support the use of alternate islet sources.
Collapse
Affiliation(s)
- Nathan W Zammit
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Stacey N Walters
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Karen L Seeberger
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney at Westmead Hospital, NSW Australia
| | - Gregory S Korbutt
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Shane T Grey
- Immunology Department, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| |
Collapse
|
24
|
Shi H, Zhou X, Chen B, Xiao J, Li Y, Zhou X, Zhou Q, Chen K, Wang Q. Clinical Relevance of the High Prevalence of MYD88 L265P Mutated Vitreoretinal Lymphoma Identified by Droplet Digital Polymerase Chain Reaction. Ocul Immunol Inflamm 2019; 29:448-455. [PMID: 31603365 DOI: 10.1080/09273948.2019.1657903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose: To investigate the frequency and clinical relevance of missense mutation at position 265 changing leucine to proline in the myeloid differentiation factor 88 gene (MYD88 L265P) in the vitreous of Chinese patients with vitreoretinal lymphoma (VRL) using droplet digital polymerase chain reaction (ddPCR).Methods: Vitreous fluid (VF) from 29 eyes of 20 VRL patients at the North Huashan Hospital were included. MYD88 L265P analysis of VF was performed using ddPCR. Associations between clinicopathologic characteristics and MYD88 mutation were analyzed using t-test or Fisher's exact test.Results: MYD88 L265P mutations were detected in 22 of 29 samples from 14 patients with diffuse large B-cell lymphomas and one patient with lymphoplasmacytoid lymphoma. However, no significant associations were found between MYD88 L265P mutation status and age, sex, lymphoma subtype or location of the primary lesion.Conclusion: The high prevalence of MYD88 L265P identified by ddPCR suggests that this method of evaluating the frequency of MYD88 L265P is a promising tool for accurate diagnosis of VRL.
Collapse
Affiliation(s)
- Huimin Shi
- Department of Ophthalmology, North Huashan Hospital, Fudan University, Shanghai, China
| | - Xian Zhou
- Department of Ophthalmology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bobin Chen
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianjiang Xiao
- Department of Ophthalmology, North Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Li
- Department of Ophthalmology, North Huashan Hospital, Fudan University, Shanghai, China
| | - Xianjin Zhou
- Department of Ophthalmology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Zhou
- Department of Ophthalmology, North Huashan Hospital, Fudan University, Shanghai, China
| | - Kun Chen
- Department of Laboratory Medicine, North Huashan Hospital, Fudan University, Shanghai, China
| | - Qingping Wang
- Department of Ophthalmology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Young RM, Phelan JD, Wilson WH, Staudt LM. Pathogenic B-cell receptor signaling in lymphoid malignancies: New insights to improve treatment. Immunol Rev 2019; 291:190-213. [PMID: 31402495 PMCID: PMC6693651 DOI: 10.1111/imr.12792] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022]
Abstract
Signals emanating from the B-cell receptor (BCR) promote proliferation and survival in diverse forms of B-cell lymphoma. Precision medicine strategies targeting the BCR pathway have been generally effective in treating lymphoma, but often fail to produce durable responses in diffuse large B-cell lymphoma (DLBCL), a common and aggressive cancer. New insights into DLBCL biology garnered from genomic analyses and functional proteogenomic studies have identified novel modes of BCR signaling in this disease. Herein, we describe the distinct roles of antigen-dependent and antigen-independent BCR signaling in different subtypes of DLBCL. We highlight mechanisms by which the BCR cooperates with TLR9 and mutant isoforms of MYD88 to drive sustained NF-κB activity in the activated B-cell-like (ABC) subtype of DLBCL. Finally, we discuss progress in detecting and targeting oncogenic BCR signaling to improve the survival of patients with lymphoma.
Collapse
MESH Headings
- Animals
- Autoantigens/immunology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Germinal Center/immunology
- Germinal Center/metabolism
- Germinal Center/pathology
- Humans
- Leukemia, Lymphoid/diagnosis
- Leukemia, Lymphoid/etiology
- Leukemia, Lymphoid/metabolism
- Leukemia, Lymphoid/therapy
- Lymphoma/diagnosis
- Lymphoma/etiology
- Lymphoma/metabolism
- Lymphoma/therapy
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Ryan M. Young
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD. 20892
| | - James D. Phelan
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD. 20892
| | - Wyndham H. Wilson
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD. 20892
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD. 20892
| |
Collapse
|
26
|
Disrupting myddosome assembly in diffuse large B‑cell lymphoma cells using the MYD88 dimerization inhibitor ST2825. Oncol Rep 2019; 42:1755-1766. [PMID: 31432184 PMCID: PMC6775815 DOI: 10.3892/or.2019.7282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL), the most common type of non-Hodgkins lymphoma, is classified into germinal center and activated B cell (ABC) subtypes. The myeloid differentiation primary response gene 88 (MYD88) L265P mutation is the most prevalent oncogenic mutation among patients with ABC DLBCL, the subtype that has the more inferior outcome. MYD88 oligomerization driven by the L265P mutant augments myddosome assembly and triggers the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, highlighting MYD88 oligomerization as a potential therapeutic target for this malignancy. The synthetic peptidomimetic compound ST2825, which has previously been used as an anti-inflammatory agent, has been reported to inhibit MYD88 dimerization. In the present study, the anticancer effects of ST2825 were investigated using L265P-expressing ABC DLBCL cell lines. Using confocal microscopy and high-molecular-weight fraction experiments, it was revealed that L265P-associated myddosome assembly was disrupted by ST2825. The results also revealed that disrupting myddosome assembly promoted the death of ABC DLBCL cells harboring the L265P mutation, as well as downregulating survival signals, including the inhibition of NF-κB and the suppression of IL-10 and interferon-β production. Further co-immunoprecipitation studies demonstrated that MYD88 bound to BTK in L265P-DLBCL cells, and that this binding was abrogated following ST2825 treatment. Furthermore, the combination of myddosome-assembly disruption and BTK or BCL-2 signaling inhibition led to synergistic ABC DLBCL cell death, and more robust inhibition of NF-κB activity or increased apoptosis, respectively. The results of the present study provide evidence that the synthetic peptidomimetic compound ST2825, which targets myddosome assembly, may serve as a pharmacological inhibitor. ST2825 has the potential for clinical use in patients with L265P DLBCL, and other B-cell neoplasms driven by activated MYD88 signaling.
Collapse
|
27
|
Single-cell MYD88 sequencing of isolated B cells from vitreous biopsies aids vitreoretinal lymphoma diagnosis. Blood 2019; 134:709-712. [PMID: 31292111 DOI: 10.1182/blood.2019000022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
28
|
Roos-Weil D, Decaudin C, Armand M, Della-Valle V, Diop MK, Ghamlouch H, Ropars V, Hérate C, Lara D, Durot E, Haddad R, Mylonas E, Damm F, Pflumio F, Stoilova B, Metzner M, Elemento O, Dessen P, Camara-Clayette V, Cosset FL, Verhoeyen E, Leblond V, Ribrag V, Cornillet-Lefebvre P, Rameau P, Azar N, Charlotte F, Morel P, Charbonnier JB, Vyas P, Mercher T, Aoufouchi S, Droin N, Guillouf C, Nguyen-Khac F, Bernard OA. A Recurrent Activating Missense Mutation in Waldenström Macroglobulinemia Affects the DNA Binding of the ETS Transcription Factor SPI1 and Enhances Proliferation. Cancer Discov 2019; 9:796-811. [PMID: 31018969 DOI: 10.1158/2159-8290.cd-18-0873] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 03/28/2019] [Accepted: 04/18/2019] [Indexed: 11/16/2022]
Abstract
The ETS-domain transcription factors divide into subfamilies based on protein similarities, DNA-binding sequences, and interaction with cofactors. They are regulated by extracellular clues and contribute to cellular processes, including proliferation and transformation. ETS genes are targeted through genomic rearrangements in oncogenesis. The PU.1/SPI1 gene is inactivated by point mutations in human myeloid malignancies. We identified a recurrent somatic mutation (Q226E) in PU.1/SPI1 in Waldenström macroglobulinemia, a B-cell lymphoproliferative disorder. It affects the DNA-binding affinity of the protein and allows the mutant protein to more frequently bind and activate promoter regions with respect to wild-type protein. Mutant SPI1 binding at promoters activates gene sets typically promoted by other ETS factors, resulting in enhanced proliferation and decreased terminal B-cell differentiation in model cell lines and primary samples. In summary, we describe oncogenic subversion of transcription factor function through subtle alteration of DNA binding leading to cellular proliferation and differentiation arrest. SIGNIFICANCE: The demonstration that a somatic point mutation tips the balance of genome-binding pattern provides a mechanistic paradigm for how missense mutations in transcription factor genes may be oncogenic in human tumors.This article is highlighted in the In This Issue feature, p. 681.
Collapse
Affiliation(s)
- Damien Roos-Weil
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Camille Decaudin
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Marine Armand
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Véronique Della-Valle
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - M'boyba K Diop
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,AMMICa, INSERM US23/CNRS UMS3655, Gustave Roussy, Villejuif, France
| | - Hussein Ghamlouch
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Virginie Ropars
- Institute for Integrative Biology of the Cell (I2BC), Institute Joliot, CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex, France
| | - Cécile Hérate
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Diane Lara
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Eric Durot
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Rima Haddad
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL, Université Paris Diderot Sorbonne Paris Cité, Fontenay-aux-Roses, France
| | - Elena Mylonas
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frederik Damm
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Francoise Pflumio
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL, Université Paris Diderot Sorbonne Paris Cité, Fontenay-aux-Roses, France
| | - Bilyana Stoilova
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine,NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine and Department of Haematology, Oxford University and Oxford University Hospitals NHS Foundation Trust, United Kingdom
| | - Marlen Metzner
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine,NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine and Department of Haematology, Oxford University and Oxford University Hospitals NHS Foundation Trust, United Kingdom
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Philippe Dessen
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,AMMICa, INSERM US23/CNRS UMS3655, Gustave Roussy, Villejuif, France
| | - Valérie Camara-Clayette
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,AMMICa, INSERM US23/CNRS UMS3655, Gustave Roussy, Villejuif, France
| | - François-Loïc Cosset
- CIRI-InternationalCenter for Infectiology Research, Team EVIR, Université de Lyon; INSERM, U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1; CNRS, UMR5308, Lyon, France
| | - Els Verhoeyen
- CIRI-InternationalCenter for Infectiology Research, Team EVIR, Université de Lyon; INSERM, U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1; CNRS, UMR5308, Lyon, France.,Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | - Vincent Ribrag
- INSERM U1170, Gustave Roussy, Villejuif, France.,DITEP Gustave Roussy, Villejuif, Paris, France
| | - Pascale Cornillet-Lefebvre
- Laboratoire d'hématologie, Pôle de biologie, CHU de Reims-Hôpital Robert Debré, Avenuedu Général Koenig, Reims, France
| | - Philippe Rameau
- AMMICa, INSERM US23/CNRS UMS3655, Gustave Roussy, Villejuif, France
| | - Nabih Azar
- Sorbonne Université, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | | | - Pierre Morel
- Centre Hospitalier Dr. Schaffner,Lens; Service d'Hématologie Clinique et Thérapie Cellulaire, CHU Amiens Picardie, Amiens cedex, France
| | - Jean-Baptiste Charbonnier
- Institute for Integrative Biology of the Cell (I2BC), Institute Joliot, CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex, France
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine,NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine and Department of Haematology, Oxford University and Oxford University Hospitals NHS Foundation Trust, United Kingdom
| | - Thomas Mercher
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Said Aoufouchi
- Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,CNRS UMR8200, Gustave Roussy, Villejuif, France
| | - Nathalie Droin
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France.,AMMICa, INSERM US23/CNRS UMS3655, Gustave Roussy, Villejuif, France
| | - Christel Guillouf
- INSERM U1170, Gustave Roussy, Villejuif, France.,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Florence Nguyen-Khac
- Sorbonne Université, Hôpital Pitié-Salpêtrière, APHP, Paris, France. .,Sorbonne Université, INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Olivier A Bernard
- INSERM U1170, Gustave Roussy, Villejuif, France. .,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Villejuif, France.,Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| |
Collapse
|
29
|
|
30
|
Weber ANR, Cardona Gloria Y, Çınar Ö, Reinhardt HC, Pezzutto A, Wolz OO. Oncogenic MYD88 mutations in lymphoma: novel insights and therapeutic possibilities. Cancer Immunol Immunother 2018; 67:1797-1807. [PMID: 30203262 PMCID: PMC11028221 DOI: 10.1007/s00262-018-2242-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Oncogenic MYD88 mutations, most notably the Leu 265 Pro (L265P) mutation, were recently identified as potential driver mutations in various B-cell non-Hodgkin Lymphomas (NHLs). The L265P mutation is now thought to be common to virtually all NHLs and occurs in between 4 and 90% of cases, depending on the entity. Since it is tumor-specific, the mutation, and the pathways it regulates, might serve as advantageous therapeutic targets for both conventional chemotherapeutic intervention, as well as immunotherapeutic strategies. Here, we review recent progress on elucidating the molecular and cellular processes affected by the L265P mutation of MYD88, describe a new in vivo model for MyD88 L265P-mediated oncogenesis, and summarize how these findings could be exploited therapeutically by specific targeting of signaling pathways. In addition, we summarize current and explore future possibilities for conceivable immunotherapeutic approaches, such as L265P-derived peptide vaccination, adoptive transfer of L265P-restricted T cells, and use of T-cell receptor-engineered T cells. With clinical trials regarding their efficacy rapidly expanding to NHLs, we also discuss potential combinations of immune checkpoint inhibitors with the described targeted chemotherapies of L265P signaling networks, and/or with the above immunological approaches as potential ways of targeting MYD88-mutated lymphomas in the future.
Collapse
Affiliation(s)
- Alexander N R Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| | - Yamel Cardona Gloria
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Özcan Çınar
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - H Christian Reinhardt
- Clinic I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Antonio Pezzutto
- Berlin Institute for Health Charité and Max-Delbrück Center, Campus Buch, Building 42-53, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité Medical School, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Olaf-Oliver Wolz
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| |
Collapse
|
31
|
Wenzl K, Manske MK, Sarangi V, Asmann YW, Greipp PT, Schoon HR, Braggio E, Maurer MJ, Feldman AL, Witzig TE, Slager SL, Ansell SM, Cerhan JR, Novak AJ. Loss of TNFAIP3 enhances MYD88 L265P-driven signaling in non-Hodgkin lymphoma. Blood Cancer J 2018; 8:97. [PMID: 30301877 PMCID: PMC6177394 DOI: 10.1038/s41408-018-0130-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/04/2018] [Accepted: 08/09/2018] [Indexed: 01/04/2023] Open
Abstract
MYD88 mutations are one of the most recurrent mutations in hematologic malignancies. However, recent mouse models suggest that MYD88L265P alone may not be sufficient to induce tumor formation. Interplay between MYD88L265P and other genetic events is further supported by the fact that TNFAIP3 (A20) inactivation often accompanies MYD88L265P. However, we are still lacking information about the consequence of MYD88L265P in combination with TNFAIP3 loss in human B cell lymphoma. Review of our genetic data on diffuse large B cell lymphoma (DLBCL) and Waldenstrom macroglobulinemia (WM), found that a large percentage of DLBCL and WM cases that have a MYD88 mutation also harbor a TNFAIP3 loss, 55% DLBCL and 28% of WM, respectively. To mimic this combination of genetic events, we used genomic editing technology to knock out TNFAIP3 in MYD88L265P non-Hodgkin's lymphoma (NHL) cell lines. Loss of A20 expression resulted in increased NF-κB and p38 activity leading to upregulation of the NF-κB target genes BCL2 and MYC. Furthermore, we detected the increased production of IL-6 and CXCL10 which led to an upregulation of the JAK/STAT pathway. Overall, these results suggest that MYD88L265P signaling can be enhanced by a second genetic alteration in TNFAIP3 and highlights a potential opportunity for therapeutic targeting.
Collapse
Affiliation(s)
- Kerstin Wenzl
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Patricia T Greipp
- Genomics Laboratory, Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Matthew J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
32
|
Magierowicz M, Tomowiak C, Leleu X, Poulain S. Working Toward a Genomic Prognostic Classification of Waldenström Macroglobulinemia: C-X-C Chemokine Receptor Type 4 Mutation and Beyond. Hematol Oncol Clin North Am 2018; 32:753-763. [PMID: 30190015 DOI: 10.1016/j.hoc.2018.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Waldenström macroglobulinemia is a rare indolent B-cell lymphoma. Whole-exome sequencing studies have improved our knowledge of the Waldenström macroglobulinemia mutational landscape. The MYD88 L265P mutation is present in nearly 90% of patients with Waldenström macroglobulinemia. CXCR4 mutations are identified in approximately 30% of MYD88L265P cases and have been associated with ibrutinib resistance in clinical trials. Mutations in CD79B, ARID1a, or TP53 were described at lower frequency. Deciphering the earliest initiating lesions and identifying the molecular alterations leading to disease progression currently represent important goals in the future to identify the most relevant targets for precision therapy in Waldenström macroglobulinemia.
Collapse
Affiliation(s)
- Marion Magierowicz
- Laboratory of Hematology, Biology and Pathology Center, CHU of Lille, Lille, France
| | - Cécile Tomowiak
- Department of Hematology, Hospital of the Miletrie, INSERM CIC 1402, CHU of Poitiers, Poitiers, France
| | - Xavier Leleu
- Department of Hematology, Hospital of the Miletrie, INSERM CIC 1402, CHU of Poitiers, Poitiers, France
| | - Stéphanie Poulain
- Laboratory of Hematology, Biology and Pathology Center, CHU of Lille, Lille, France; INSERM UMR S 1172, Team 4, Cancer Research Institute, Lille, France.
| |
Collapse
|
33
|
Yu X, Li W, Deng Q, Li L, Hsi ED, Young KH, Zhang M, Li Y. MYD88 L265P Mutation in Lymphoid Malignancies. Cancer Res 2018; 78:2457-2462. [PMID: 29703722 DOI: 10.1158/0008-5472.can-18-0215] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/13/2018] [Accepted: 03/06/2018] [Indexed: 11/16/2022]
Abstract
Next-generation sequencing has revealed cancer genomic landscapes, in which over 100 driver genes that, when altered by intragenic mutations, can promote oncogenesis. MYD88 is a driver gene found in hematologic B-cell malignancies. A missense mutation (L265P) changing leucine at position 265 to proline in MYD88 is found in ∼90% of Waldenström macroglobulinemia (WM) cases and in significant portions of activated B-cell diffuse large B-cell lymphomas and IgM monoclonal gammopathy of undetermined significance. Few cancers such as WM have a single amino acid substitution in one gene like MYD88 L265P that occurs in ∼90% of cases, making WM paradigmatic for study of a single causative mutation in oncogenesis. In this review, we summarize the frequency and cancer spectrum of MYD88 L265P and its downstream effects in lymphoid cancers. Malignant B cells with MYD88 L265P are likely transformed from IgM-producing B cells either in response to T-cell-independent antigens or in response to protein antigens before class switching. We also discuss therapeutic strategies that include targeting Bruton tyrosine kinase and other kinases, interfering with the assembly of MYD88 and its interacting partners, and MYD88 L265P-specific peptide-based immunotherapy. Cancer Res; 78(10); 2457-62. ©2018 AACR.
Collapse
Affiliation(s)
- Xinfang Yu
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan Province, China
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Wei Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Qipan Deng
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ling Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan Province, China
| | - Eric D Hsi
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mingzhi Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan Province, China.
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
34
|
Extracellular vesicle–mediated transfer of constitutively active MyD88L265P engages MyD88wt and activates signaling. Blood 2018; 131:1720-1729. [DOI: 10.1182/blood-2017-09-805499] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/13/2018] [Indexed: 01/19/2023] Open
Abstract
Key Points
MyD88L265P is present in the EVs secreted by WM cancer cells and triggers signaling in the recipient cells. MyD88-containing EVs shape the proinflammatory microenvironment in the bone marrow.
Collapse
|
35
|
Grondona P, Bucher P, Schulze-Osthoff K, Hailfinger S, Schmitt A. NF-κB Activation in Lymphoid Malignancies: Genetics, Signaling, and Targeted Therapy. Biomedicines 2018; 6:biomedicines6020038. [PMID: 29587428 PMCID: PMC6027339 DOI: 10.3390/biomedicines6020038] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 12/12/2022] Open
Abstract
The NF-κB transcription factor family plays a crucial role in lymphocyte proliferation and survival. Consequently, aberrant NF-κB activation has been described in a variety of lymphoid malignancies, including diffuse large B-cell lymphoma, Hodgkin lymphoma, and adult T-cell leukemia. Several factors, such as persistent infections (e.g., with Helicobacter pylori), the pro-inflammatory microenvironment of the cancer, self-reactive immune receptors as well as genetic lesions altering the function of key signaling effectors, contribute to constitutive NF-κB activity in these malignancies. In this review, we will discuss the molecular consequences of recurrent genetic lesions affecting key regulators of NF-κB signaling. We will particularly focus on the oncogenic mechanisms by which these alterations drive deregulated NF-κB activity and thus promote the growth and survival of the malignant cells. As the concept of a targeted therapy based on the mutational status of the malignancy has been supported by several recent preclinical and clinical studies, further insight in the function of NF-κB modulators and in the molecular mechanisms governing aberrant NF-κB activation observed in lymphoid malignancies might lead to the development of additional treatment strategies and thus improve lymphoma therapy.
Collapse
Affiliation(s)
- Paula Grondona
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Philip Bucher
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Klaus Schulze-Osthoff
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Stephan Hailfinger
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Anja Schmitt
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| |
Collapse
|
36
|
Sikora KA, Bennett JR, Vyncke L, Deng Z, Tsai WL, Pauwels E, Layh-Schmitt G, Brundidge A, Navid F, Zaal KJM, Hanson E, Gadina M, Staudt LM, Griffin TA, Tavernier J, Peelman F, Colbert RA. Germline gain-of-function myeloid differentiation primary response gene-88 (MYD88) mutation in a child with severe arthritis. J Allergy Clin Immunol 2018; 141:1943-1947.e9. [PMID: 29427642 DOI: 10.1016/j.jaci.2018.01.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 01/10/2018] [Accepted: 01/22/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Keith A Sikora
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md.
| | - Joshua R Bennett
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Laurens Vyncke
- VIB Medical Biotechnology Center, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Zuoming Deng
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Wanxia Li Tsai
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Ewald Pauwels
- Center for Molecular Modeling, Ghent University, Zwijnaarde, Belgium
| | - Gerlinde Layh-Schmitt
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - April Brundidge
- Office of Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Fatemeh Navid
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Kristien J M Zaal
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Eric Hanson
- Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Massimo Gadina
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Louis M Staudt
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Thomas A Griffin
- Levine Children's Hospital at Carolinas Medical Center, Charlotte, NC
| | - Jan Tavernier
- VIB Medical Biotechnology Center, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Frank Peelman
- VIB Medical Biotechnology Center, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Robert A Colbert
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
37
|
Zhao X, Zhang Z, Moreira D, Su YL, Won H, Adamus T, Dong Z, Liang Y, Yin HH, Swiderski P, Pillai RK, Kwak L, Forman S, Kortylewski M. B Cell Lymphoma Immunotherapy Using TLR9-Targeted Oligonucleotide STAT3 Inhibitors. Mol Ther 2018; 26:695-707. [PMID: 29433938 PMCID: PMC5910676 DOI: 10.1016/j.ymthe.2018.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 01/22/2023] Open
Abstract
Growing evidence links the aggressiveness of non-Hodgkin’s lymphoma, especially the activated B cell-like type diffuse large B cell lymphomas (ABC-DLBCLs) to Toll-like receptor 9 (TLR9)/MyD88 and STAT3 transcription factor signaling. Here, we describe a dual-function molecule consisting of a clinically relevant TLR9 agonist (CpG7909) and a STAT3 inhibitor in the form of a high-affinity decoy oligodeoxynucleotide (dODN). The CpG-STAT3dODN blocked STAT3 DNA binding and activity, thus reducing expression of downstream target genes, such as MYC and BCL2L1, in human and mouse lymphoma cells. We further demonstrated that injections (i.v.) of CpG-STAT3dODN inhibited growth of human OCI-Ly3 lymphoma in immunodeficient mice. Moreover, systemic CpG-STAT3dODN administration induced complete regression of the syngeneic A20 lymphoma, resulting in long-term survival of immunocompetent mice. Both TLR9 stimulation and concurrent STAT3 inhibition were critical for immune-mediated therapeutic effects, since neither CpG7909 alone nor CpG7909 co-injected with unconjugated STAT3dODN extended mouse survival. The CpG-STAT3dODN induced expression of genes critical to antigen-processing/presentation and Th1 cell activation while suppressing survival signaling. These effects resulted in the generation of lymphoma cell-specific CD8/CD4-dependent T cell immunity protecting mice from tumor rechallenge. Our results suggest that CpG-STAT3dODN as a systemic/local monotherapy or in combination with PD1 blockade can provide an opportunity for treating patients with B cell NHL.
Collapse
Affiliation(s)
- Xingli Zhao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300020, China
| | - Zhuoran Zhang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Dayson Moreira
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yu-Lin Su
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Haejung Won
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Tomasz Adamus
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Zhenyuan Dong
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; Toni Stephenson Lymphoma Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yong Liang
- DNA/RNA Synthesis Core Facility, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Hongwei H Yin
- Molecular Pathology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Piotr Swiderski
- DNA/RNA Synthesis Core Facility, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Raju K Pillai
- Molecular Pathology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Larry Kwak
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; Toni Stephenson Lymphoma Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Stephen Forman
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; Toni Stephenson Lymphoma Center, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
38
|
Hindi SM, Shin J, Gallot YS, Straughn AR, Simionescu-Bankston A, Hindi L, Xiong G, Friedland RP, Kumar A. MyD88 promotes myoblast fusion in a cell-autonomous manner. Nat Commun 2017; 8:1624. [PMID: 29158520 PMCID: PMC5696367 DOI: 10.1038/s41467-017-01866-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 10/20/2017] [Indexed: 12/27/2022] Open
Abstract
Myoblast fusion is an indispensable step for skeletal muscle development, postnatal growth, and regeneration. Myeloid differentiation primary response gene 88 (MyD88) is an adaptor protein that mediates Toll-like receptors and interleukin-1 receptor signaling. Here we report a cell-autonomous role of MyD88 in the regulation of myoblast fusion. MyD88 protein levels are increased during in vitro myogenesis and in conditions that promote skeletal muscle growth in vivo. Deletion of MyD88 impairs fusion of myoblasts without affecting their survival, proliferation, or differentiation. MyD88 regulates non-canonical NF-κB and canonical Wnt signaling during myogenesis and promotes skeletal muscle growth and overload-induced myofiber hypertrophy in mice. Ablation of MyD88 reduces myofiber size during muscle regeneration, whereas its overexpression promotes fusion of exogenous myoblasts to injured myofibers. Our study shows that MyD88 modulates myoblast fusion and suggests that augmenting its levels may be a therapeutic approach to improve skeletal muscle formation in degenerative muscle disorders.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Jonghyun Shin
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Yann S Gallot
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Alex R Straughn
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Adriana Simionescu-Bankston
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Lubna Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Guangyan Xiong
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Robert P Friedland
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
39
|
Amin AD, Peters TL, Li L, Rajan SS, Choudhari R, Puvvada SD, Schatz JH. Diffuse large B-cell lymphoma: can genomics improve treatment options for a curable cancer? Cold Spring Harb Mol Case Stud 2017; 3:a001719. [PMID: 28487884 PMCID: PMC5411687 DOI: 10.1101/mcs.a001719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gene-expression profiling and next-generation sequencing have defined diffuse large B-cell lymphoma (DLBCL), the most common lymphoma diagnosis, as a heterogeneous group of subentities. Despite ongoing explosions of data illuminating disparate pathogenic mechanisms, however, the five-drug chemoimmunotherapy combination R-CHOP remains the frontline standard treatment. This has not changed in 15 years, since the anti-CD20 monoclonal antibody rituximab was added to the CHOP backbone, which first entered use in the 1970s. At least a third of patients are not cured by R-CHOP, and relapsed or refractory DLBCL is fatal in ∼90%. Targeted small-molecule inhibitors against distinct molecular pathways activated in different subgroups of DLBCL have so far translated poorly into the clinic, justifying the ongoing reliance on R-CHOP and other long-established chemotherapy-driven combinations. New drugs and improved identification of biomarkers in real time, however, show potential to change the situation eventually, despite some recent setbacks. Here, we review established and putative molecular drivers of DLBCL identified through large-scale genomics, highlighting among other things the care that must be taken when differentiating drivers from passengers, which is influenced by the promiscuity of activation-induced cytidine deaminase. Furthermore, we discuss why, despite having so much genomic data available, it has been difficult to move toward personalized medicine for this umbrella disorder and some steps that may be taken to hasten the process.
Collapse
Affiliation(s)
- Amit Dipak Amin
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Tara L Peters
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Lingxiao Li
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Soumya Sundara Rajan
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ramesh Choudhari
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Soham D Puvvada
- Department of Medicine, Division of Hematology-Oncology, University of Arizona Comprehensive Cancer Center, Tucson, Arizona 85719, USA
| | - Jonathan H Schatz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
40
|
Wang JQ, Jeelall YS, Humburg P, Batchelor EL, Kaya SM, Yoo HM, Goodnow CC, Horikawa K. Synergistic cooperation and crosstalk between MYD88L265P and mutations that dysregulate CD79B and surface IgM. J Exp Med 2017; 214:2759-2776. [PMID: 28701369 PMCID: PMC5584117 DOI: 10.1084/jem.20161454] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 04/30/2017] [Accepted: 06/13/2017] [Indexed: 12/26/2022] Open
Abstract
Wang et al. show cooperation between MYD88L265P and CD79B mutations dysregulating B cell responses to self-antigen and differentiation into plasma cells. Their results reveal that CD79B and surface IgM constitute a rate-limiting checkpoint against MYD88L265P, explaining the co-occurrence of MYD88 and CD79B mutations in human lymphomas. CD79B and MYD88 mutations are frequently and simultaneously detected in B cell malignancies. It is not known if these mutations cooperate or how crosstalk occurs. Here we analyze the consequences of CD79B and MYD88L265P mutations individually and combined in normal activated mouse B lymphocytes. CD79B mutations alone increased surface IgM but did not enhance B cell survival, proliferation, or altered NF-κB responsive markers. Conversely, B cells expressing MYD88L265P decreased surface IgM coupled with accumulation of endoglycosidase H–sensitive IgM intracellularly, resembling the trafficking block in anergic B cells repeatedly stimulated by self-antigen. Mutation or overexpression of CD79B counteracted the effect of MYD88L265P. In B cells chronically stimulated by self-antigen, CD79B and MYD88L265P mutations in combination, but not individually, blocked peripheral deletion and triggered differentiation into autoantibody secreting plasmablasts. These results reveal that CD79B and surface IgM constitute a rate-limiting checkpoint against B cell dysregulation by MYD88L265P and provide an explanation for the co-occurrence of MYD88 and CD79B mutations in lymphomas.
Collapse
Affiliation(s)
- James Q Wang
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Yogesh S Jeelall
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Peter Humburg
- Garvan Institute of Medical Research, Sydney, Australia
| | - Emma L Batchelor
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Sarp M Kaya
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Hee Min Yoo
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | - Keisuke Horikawa
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
41
|
Role of plasma cells in Waldenström macroglobulinaemia. Pathology 2017; 49:337-345. [DOI: 10.1016/j.pathol.2017.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/26/2017] [Accepted: 02/27/2017] [Indexed: 12/13/2022]
|
42
|
Purroy N, Wu CJ. Coevolution of Leukemia and Host Immune Cells in Chronic Lymphocytic Leukemia. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026740. [PMID: 28096240 DOI: 10.1101/cshperspect.a026740] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cumulative studies on the dissection of changes in driver genetic lesions in cancer across the course of the disease have provided powerful insights into the adaptive mechanisms of tumors in response to the selective pressures of therapy and environmental changes. In particular, the advent of next-generation-sequencing (NGS)-based technologies and its implementation for the large-scale comprehensive analyses of cancers have greatly advanced our understanding of cancer as a complex dynamic system wherein genetically distinct subclones interact and compete during tumor evolution. Aside from genetic evolution arising from interactions intrinsic to the cell subpopulations within tumors, it is increasingly appreciated that reciprocal interactions between the tumor cell and cellular constituents of the microenvironment further exert selective pressures on specific clones that can impact the balance between tumor immunity and immunologic evasion and escape. Herein, we review the evidence supporting these concepts, with a particular focus on chronic lymphocytic leukemia (CLL), a disease that has been highly amenable to genomic interrogation and studies of clonal heterogeneity and evolution. Better knowledge of the basis for immune escape has an important clinical impact on prognostic stratification and on the pursuit of new therapeutic opportunities.
Collapse
Affiliation(s)
- Noelia Purroy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142.,Harvard Medical School, Boston, Massachusetts 02115
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142.,Harvard Medical School, Boston, Massachusetts 02115.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
43
|
Heterogeneity of Toll-like receptor 9 signaling in B cell malignancies and its potential therapeutic application. J Transl Med 2017; 15:51. [PMID: 28241765 PMCID: PMC5329966 DOI: 10.1186/s12967-017-1152-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/17/2017] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptor 9 (TLR9) is expressed in a variety of B-cell malignancies and works as a bridge between innate and adaptive immunity. CpG oligodeoxynucleotides (CpG ODNs), TLR9 agonists, are able to induce anticancer immune responses and exert direct effects against cancer cells, serving as cancer therapeutic agents. Therefore, TLR9 might be a potential therapeutic target for drug development. However, several new evidences have revealed that direct effects of TLR9 agonists on B-cell malignancies is controversial. For example, CpG ODNs can induce apoptosis in certain type of chronic lymphocytic leukemia and lymphoma cells, while induce proliferation in multiple myeloma and other types of lymphoma cells. In this review, we summarize current understanding of the heterogeneity in responses of normal and malignant B cells to TLR9 agonists, due to differences in TLR9 expression levels, genetic alterations (such as MyD88 mutation), and signaling pathway activation. Especially, the downstream molecules of NF-κB signaling pathway play an important role in the heterogeneous response. In order to provide possibilities for therapeutic manipulation of TLR9 agonists in the treatment of these disorders, the preclinical and clinical advances in using CpG ODNs alone and in combination therapies are also summarized in this review.
Collapse
|
44
|
Dubois S, Viailly PJ, Bohers E, Bertrand P, Ruminy P, Marchand V, Maingonnat C, Mareschal S, Picquenot JM, Penther D, Jais JP, Tesson B, Peyrouze P, Figeac M, Desmots F, Fest T, Haioun C, Lamy T, Copie-Bergman C, Fabiani B, Delarue R, Peyrade F, André M, Ketterer N, Leroy K, Salles G, Molina TJ, Tilly H, Jardin F. Biological and Clinical Relevance of Associated Genomic Alterations in MYD88 L265P and non-L265P-Mutated Diffuse Large B-Cell Lymphoma: Analysis of 361 Cases. Clin Cancer Res 2016; 23:2232-2244. [PMID: 27923841 DOI: 10.1158/1078-0432.ccr-16-1922] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/01/2016] [Accepted: 11/17/2016] [Indexed: 01/01/2023]
Abstract
Purpose:MYD88 mutations, notably the recurrent gain-of-function L265P variant, are a distinguishing feature of activated B-cell like (ABC) diffuse large B-cell lymphoma (DLBCL), leading to constitutive NFκB pathway activation. The aim of this study was to examine the distinct genomic profiles of MYD88-mutant DLBCL, notably according to the presence of the L265P or other non-L265P MYD88 variants.Experimental Design: A cohort of 361 DLBCL cases (94 MYD88 mutant and 267 MYD88 wild-type) was submitted to next-generation sequencing (NGS) focusing on 34 genes to analyze associated mutations and copy number variations, as well as gene expression profiling, and clinical and prognostic analyses.Results: Importantly, we highlighted different genomic profiles for MYD88 L265P and MYD88 non-L265P-mutant DLBCL, shedding light on their divergent backgrounds. Clustering analysis also segregated subgroups according to associated genetic alterations among patients with the same MYD88 mutation. We showed that associated CD79B and MYD88 L265P mutations act synergistically to increase NFκB pathway activation, although the majority of MYD88 L265P-mutant cases harbors downstream NFκB alterations, which can predict BTK inhibitor resistance. Finally, although the MYD88 L265P variant was not an independent prognostic factor in ABC DLBCL, associated CD79B mutations significantly improved the survival of MYD88 L265P-mutant ABC DLBCL in our cohort.Conclusions: This study highlights the relative heterogeneity of MYD88-mutant DLBCL, adding to the field's knowledge of the theranostic importance of MYD88 mutations, but also of associated alterations, emphasizing the usefulness of genomic profiling to best stratify patients for targeted therapy. Clin Cancer Res; 23(9); 2232-44. ©2016 AACR.
Collapse
Affiliation(s)
- Sydney Dubois
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Pierre-Julien Viailly
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.,LITIS EA 4108, Normandie Université, Rouen, France
| | - Elodie Bohers
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Philippe Bertrand
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Philippe Ruminy
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Vinciane Marchand
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | | | - Sylvain Mareschal
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | | | - Dominique Penther
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | | | | | | | | | | | | | - Corinne Haioun
- Unité Hémopathies Lymphoïdes, AP-HP Hôpital Henri Mondor, Créteil, France
| | | | | | - Bettina Fabiani
- Laboratoire de Pathologie, AP-HP Hôpital Saint Antoine, Paris, France
| | - Richard Delarue
- Department of Hematology, AP-HP Hôpital Necker, Paris, France
| | | | - Marc André
- CHU Dinant Godinne, UcL Namur, Yvoir, Belgium
| | | | - Karen Leroy
- Inserm U955 Team 09, AP-HP Hôpital Henri Mondor, Créteil, France
| | | | - Thierry J Molina
- Pathology, AP-HP Hôpital Necker, Université Paris Descartes, Paris, France
| | - Hervé Tilly
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Fabrice Jardin
- Inserm U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.
| |
Collapse
|
45
|
Genetic characterization of MYD88-mutated lymphoplasmacytic lymphoma in comparison with MYD88-mutated chronic lymphocytic leukemia. Leukemia 2016; 31:1355-1362. [DOI: 10.1038/leu.2016.330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/04/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022]
|
46
|
Spina V, Rossi D. Molecular pathogenesis of splenic and nodal marginal zone lymphoma. Best Pract Res Clin Haematol 2016; 30:5-12. [PMID: 28288716 DOI: 10.1016/j.beha.2016.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 12/18/2022]
Abstract
Genomic studies have improved our understanding of the biological basis of splenic (SMZL) and nodal (NMZL) marginal zone lymphoma by providing a comprehensive and unbiased view of the genes/pathways that are deregulated in these diseases. Consistent with the physiological involvement of NOTCH, NF-κB, B-cell receptor and toll-like receptor signaling in mature B-cells differentiation into the marginal zone B-cells, many oncogenic mutations of genes involved in these pathways have been identified in SMZL and NMZL. Beside genetic lesions, also epigenetic and post-transcriptional modifications contribute to the deregulation of marginal zone B-cell differentiation pathways in SMZL and NMZL. This review describes the progress in understanding the molecular mechanism underlying SMZL and NMZL, including molecular and post-transcriptional modifications, and discusses how information gained from these efforts has provided new insights on potential targets of diagnostic, prognostic and therapeutic relevance in SMZL and NMZL.
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- Cell Differentiation
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphoma, B-Cell, Marginal Zone/diagnosis
- Lymphoma, B-Cell, Marginal Zone/genetics
- Lymphoma, B-Cell, Marginal Zone/metabolism
- Lymphoma, B-Cell, Marginal Zone/therapy
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Protein Processing, Post-Translational
- Splenic Neoplasms/diagnosis
- Splenic Neoplasms/genetics
- Splenic Neoplasms/metabolism
- Splenic Neoplasms/therapy
Collapse
Affiliation(s)
- Valeria Spina
- Hematology, Institute of Oncology Research and Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Davide Rossi
- Hematology, Institute of Oncology Research and Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.
| |
Collapse
|
47
|
Monlish DA, Bhatt ST, Schuettpelz LG. The Role of Toll-Like Receptors in Hematopoietic Malignancies. Front Immunol 2016; 7:390. [PMID: 27733853 PMCID: PMC5039188 DOI: 10.3389/fimmu.2016.00390] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/15/2016] [Indexed: 12/02/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors that shape the innate immune system by identifying pathogen-associated molecular patterns and host-derived damage-associated molecular patterns. TLRs are widely expressed on both immune cells and non-immune cells, including hematopoietic stem and progenitor cells, effector immune cell populations, and endothelial cells. In addition to their well-known role in the innate immune response to acute infection or injury, accumulating evidence supports a role for TLRs in the development of hematopoietic and other malignancies. Several hematopoietic disorders, including lymphoproliferative disorders and myelodysplastic syndromes, which possess a high risk of transformation to leukemia, have been linked to aberrant TLR signaling. Furthermore, activation of TLRs leads to the induction of a number of proinflammatory cytokines and chemokines, which can promote tumorigenesis by driving cell proliferation and migration and providing a favorable microenvironment for tumor cells. Beyond hematopoietic malignancies, the upregulation of a number of TLRs has been linked to promoting tumor cell survival, proliferation, and metastasis in a variety of cancers, including those of the colon, breast, and lung. This review focuses on the contribution of TLRs to hematopoietic malignancies, highlighting the known direct and indirect effects of TLR signaling on tumor cells and their microenvironment. In addition, the utility of TLR agonists and antagonists as potential therapeutics in the treatment of hematopoietic malignancies is discussed.
Collapse
Affiliation(s)
- Darlene A Monlish
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Sima T Bhatt
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Laura G Schuettpelz
- Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
48
|
Batool M, Anwar MA, Choi S. Toll-like receptors targeting technology for the treatment of lymphoma. Expert Opin Drug Discov 2016; 11:1047-1059. [PMID: 27602749 DOI: 10.1080/17460441.2016.1233964] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The crucial role of Toll-like Receptors (TLRs) in innate and adaptive immune systems is well discussed in the literature. In cancer, TLRs act as a double-edged sword that can promote or suppress tumor growth. Areas covered: In this article, the authors uncover the potential role of TLRs in lymphomas, which are cancers related to the lymphatic system and blood cells. TLRs are de facto inflammation-inducing receptors that can either worsen disease or ameliorate lymphoma treatment. From this perspective, the usage of TLRs to modulate the immune system toward lymphoma regression is desirable. Various strategies have been used so far, and novel ways are being sought out to cure lymphoma. Expert opinion: TLR ligands have successfully been used to improve patient health; however, these receptors must be finely tuned to further optimize therapy. For a better outcome, novel specific ligands, improved pharmacodynamics, and unique targets should be discerned. Ligands with conjugated molecules, nanoparticles, and targeted drug delivery can highly optimize the therapy for lymphoma with various etiologies.
Collapse
Affiliation(s)
- Maria Batool
- a Department of Molecular Science and Technology , Ajou University , Suwon , Korea
| | - Muhammad Ayaz Anwar
- a Department of Molecular Science and Technology , Ajou University , Suwon , Korea
| | - Sangdun Choi
- a Department of Molecular Science and Technology , Ajou University , Suwon , Korea
| |
Collapse
|
49
|
Krappmann D, Vincendeau M. Mechanisms of NF-κB deregulation in lymphoid malignancies. Semin Cancer Biol 2016; 39:3-14. [DOI: 10.1016/j.semcancer.2016.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/17/2022]
|
50
|
Inhibiting TLR9 and other UNC93B1-dependent TLRs paradoxically increases accumulation of MYD88L265P plasmablasts in vivo. Blood 2016; 128:1604-8. [PMID: 27458005 DOI: 10.1182/blood-2016-03-708065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/17/2016] [Indexed: 12/25/2022] Open
Abstract
The MYD88(L265P) mutation is found in 2% to 10% of chronic lymphocytic leukemia, 29% of activated B-cell type diffuse large B-cell lymphoma and 90% of Waldenström macroglobulinemia, making it conceptually attractive to treat these malignancies with inhibitors of endosomal Toll-like receptors (TLR9, TLR7) that activate MYD88. Here we show that genetic inhibition of endosomal TLRs has the opposite effect on accumulation of MYD88(L265P) B cells in vitro and in vivo. Activated mature B cells from wild-type, Unc93b1(3d/3d)-mutant, or Tlr9-deficient mice were transduced with retrovirus encoding MYD88(L265P) and analyzed either in vitro or after transplantation into Rag1(-/-) recipient mice. Unc93b1(3d/3d) mutation, which blocks TLR9 and TLR7 signaling, or Tlr9 deficiency suppressed MYD88(L265P) B-cell growth in vitro but paradoxically increased in vivo accumulation of MYD88(L265P) B cells as CD19(low) plasmablasts by 10- to 100-fold. These results reveal an unexpected, powerful inhibitory effect of TLR9 on MYD88(L265P) B-cell proliferation and differentiation that appears independent of TLR7, and they provide a preclinical indicator for caution in clinical trials of TLR7/9 inhibitors for MYD88(L265P) B-cell malignancies.
Collapse
|