1
|
Luo JF, Wang S, Fu J, Xu P, Shao N, Lu JH, Ming C. Integration of transcriptional and epigenetic regulation of TFEB reveals its dual functional roles in Pan-cancer. NAR Cancer 2024; 6:zcae043. [PMID: 39554489 PMCID: PMC11567160 DOI: 10.1093/narcan/zcae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/03/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
Transcription factor EB (TFEB) mainly regulates the autophagy-lysosomal pathway, associated with many diseases, including cancer. However, the role of TFEB in pan-cancer has not been investigated systematically. In this study, we comprehensively analyzed TFEB targets under three stresses in Hela cells by cross-validation of RNA-seq and ChIP-seq. 1712 novel TFEB targets have not been reported in the Gene Set Enrichment Analysis and ChIP Enrichment Analysis databases. We further investigated their distributions and roles among the pan-cancer co-expression networks across 32 cancers constructed by multiscale embedded gene co-expression network analysis (MEGENA) based on the Cancer Genome Atlas (TCGA) cohort. Specifically, TFEB might serve as a hidden player with multifaceted functions in regulating pan-cancer risk factors, e.g. CXCL2, PKMYT1 and BUB1, associated with cell cycle and immunosuppression. TFEB might also regulate protective factors, e.g. CD79A, related to immune promotion in the tumor microenvironment. We further developed a Shiny app website to present the comprehensive regulatory targets of TFEB under various stimuli, intending to support further research on TFEB functions. Summarily, we provided references for the TFEB downstream targets responding to three stresses and the dual roles of TFEB and its targets in pan-cancer, which are promising anticancer targets that warrant further exploration.
Collapse
Affiliation(s)
- Jing-Fang Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR999078, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR999078, China
- Ministry of Education Frontiers Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR999078, China
| | - Shijia Wang
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR999078, China
- Ministry of Education Frontiers Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR999078, China
| | - Jiajing Fu
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR999078, China
- Ministry of Education Frontiers Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR999078, China
| | - Peng Xu
- Centre of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Ningyi Shao
- Ministry of Education Frontiers Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR999078, China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR999078, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR999078, China
| | - Chen Ming
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR999078, China
- Ministry of Education Frontiers Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Macao SAR 999078, China
| |
Collapse
|
2
|
Kupcova K, Senavova J, Jura F, Herman V, Rajmonova A, Pacheco-Blanco M, Chrbolkova T, Hamova I, Davis RE, Havranek O. Vertical targeting of the PI3K/AKT pathway at multiple points is synergistic and effective for non-Hodgkin lymphoma. Exp Hematol Oncol 2024; 13:108. [PMID: 39487517 PMCID: PMC11529427 DOI: 10.1186/s40164-024-00568-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024] Open
Abstract
The phosphatidylinositol 3‑kinase/protein kinase B (PI3K/AKT) signaling pathway is critically active in many cell types, both normal and neoplastic. Many small-molecule inhibitors targeting different levels of the PI3K/AKT pathway have been developed for cancer therapy, but their efficacy is reduced by compensatory pathway re-activation mechanisms, and their tolerability by toxic side effects. We studied this problem using cell lines representing diffuse large B-cell lymphoma (SUDHL-4 and OCI-Ly7), a genetically-encoded live-cell reporter of AKT activity, and 3 small-molecule inhibitors targeting different levels of the pathway: idelalisib (PI3Kδ), GSK2334470 (PDPK1), and ipatasertib (AKT). Half-maximal (IC50) concentrations of these inhibitors for AKT activity inhibition at 1 h, when used individually, were much lower than their IC50 values for reduction of viable cell number after 4 days. Time-course studies explained this discrepancy: AKT activity in the continuous presence of the inhibitors returned to normal after 24 h, and was supranormal after inhibitor removal. Combining all 3 inhibitors produced sustained inhibition of AKT activity, was broadly synergistic at reducing viable cell number, enabled substantially lower doses of each inhibitor to be used, and was enhanced further by the mTOR inhibitor rapamycin. Moreover, combined PDPK1 and AKT inhibition showed synergy with multiple different PI3K inhibitors. In a syngeneic mouse cell line model of lymphoma (A20), the triple combination showed antitumor activity and no evidence of toxicity. Our findings provide proof of concept suggesting further study of the safety and efficacy of low-dose multilevel PI3K/AKT pathway inhibition, for lymphoma and perhaps other cancers.
Collapse
Affiliation(s)
- Kristyna Kupcova
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
- First Department of Medicine, Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jana Senavova
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
- First Department of Medicine, Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Filip Jura
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
| | - Vaclav Herman
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
- First Department of Medicine, Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Anezka Rajmonova
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
| | - Mariana Pacheco-Blanco
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
| | - Tereza Chrbolkova
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
| | - Iva Hamova
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic
- First Department of Medicine, Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - R Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ondrej Havranek
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Prague, 25250, Czech Republic.
- First Department of Medicine, Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
3
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
4
|
Ondrisova L, Seda V, Hlavac K, Pavelkova P, Hoferkova E, Chiodin G, Kostalova L, Mladonicka Pavlasova G, Filip D, Vecera J, Zeni PF, Oppelt J, Kahounova Z, Vichova R, Soucek K, Panovska A, Plevova K, Pospisilova S, Simkovic M, Vrbacky F, Lysak D, Fernandes SM, Davids MS, Maiques-Diaz A, Charalampopoulou S, Martin-Subero JI, Brown JR, Doubek M, Forconi F, Mayer J, Mraz M. FoxO1/Rictor axis induces a non-genetic adaptation to Ibrutinib via Akt activation in chronic lymphocytic leukemia. J Clin Invest 2024; 134:e173770. [PMID: 39436708 PMCID: PMC11601945 DOI: 10.1172/jci173770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
BTK inhibitor therapy induces peripheral blood lymphocytosis in chronic lymphocytic leukemia (CLL) lasting for several months. It remains unclear whether non-genetic adaptation mechanisms exist, allowing CLL cells' survival during BTK inhibitor-induced lymphocytosis and/or playing a role in therapy resistance. We show that in approximately 70 % of CLL cases, ibrutinib treatment in vivo increases Akt activity above pre-therapy levels within several weeks, leading to compensatory CLL cell survival and a more prominent lymphocytosis on therapy. Ibrutinib-induced Akt phosphorylation (pAktS473) is caused by the upregulation of FoxO1 transcription factor, which induces expression of Rictor, an assembly protein for mTORC2 protein complex that directly phosphorylates Akt at serine 473 (S473). Knock-out or inhibition of FoxO1 or Rictor led to a dramatic decrease in Akt phosphorylation and growth disadvantage for malignant B cells in the presence of ibrutinib (or PI3K inhibitor idelalisib) in vitro and in vivo. FoxO1/Rictor/pAktS473 axis represents an early non-genetic adaptation to BCR inhibitor therapy not requiring PI3Kδ or BTK kinase activity. We further demonstrate that FoxO1 can be targeted therapeutically, and its inhibition induces CLL cells' apoptosis alone or in combination with BTK inhibitors (ibrutinib, acalabrutinib, pirtobrutinib) and blocks their proliferation triggered by T-cell factors (CD40L, IL-4, and IL-21).
Collapse
Affiliation(s)
- Laura Ondrisova
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Vaclav Seda
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Krystof Hlavac
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petra Pavelkova
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Eva Hoferkova
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Giorgia Chiodin
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Lenka Kostalova
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | | | - Daniel Filip
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Josef Vecera
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | | | - Jan Oppelt
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
| | - Zuzana Kahounova
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Rachel Vichova
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Karel Soucek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Anna Panovska
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Karla Plevova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Sarka Pospisilova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Simkovic
- Fourth Department of Internal Medicine–Haematology, University Hospital Hradec Kralove and Faculty of Medicine Hradec Kralove, Charles University, Prague, Czechia
| | - Filip Vrbacky
- Fourth Department of Internal Medicine–Haematology, University Hospital Hradec Kralove and Faculty of Medicine Hradec Kralove, Charles University, Prague, Czechia
| | - Daniel Lysak
- Department of Haematology and Oncology, University Hospital Pilsen, Pilsen, Czechia
| | - Stacey M. Fernandes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alba Maiques-Diaz
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Stella Charalampopoulou
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Jose I. Martin-Subero
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Jennifer R. Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Doubek
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Francesco Forconi
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Haematology Department, Cancer Care Directorate, University Hospital Southampton NHS Trust, Southampton, United Kingdom
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marek Mraz
- Molecular Medicine, CEITEC Masaryk University, Brno, Czechia
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
5
|
Klinger B, Rausch I, Sieber A, Kutz H, Kruse V, Kirchner M, Mertins P, Kieser A, Blüthgen N, Kube D. Quantitative modeling of signaling in aggressive B cell lymphoma unveils conserved core network. PLoS Comput Biol 2024; 20:e1012488. [PMID: 39352924 PMCID: PMC11469524 DOI: 10.1371/journal.pcbi.1012488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/11/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
B cell receptor (BCR) signaling is required for the survival and maturation of B cells and is deregulated in B cell lymphomas. While proximal BCR signaling is well studied, little is known about the crosstalk of downstream effector pathways, and a comprehensive quantitative network analysis of BCR signaling is missing. Here, we semi-quantitatively modelled BCR signaling in Burkitt lymphoma (BL) cells using systematically perturbed phosphorylation data of BL-2 and BL-41 cells. The models unveiled feedback and crosstalk structures in the BCR signaling network, including a negative crosstalk from p38 to MEK/ERK. The relevance of the crosstalk was verified for BCR and CD40 signaling in different BL cells and confirmed by global phosphoproteomics on ERK itself and known ERK target sites. Compared to the starting network, the trained network for BL-2 cells was better transferable to BL-41 cells. Moreover, the BL-2 network was also suited to model BCR signaling in Diffuse large B cell lymphoma cells lines with aberrant BCR signaling (HBL-1, OCI-LY3), indicating that BCR aberration does not cause a major downstream rewiring.
Collapse
Affiliation(s)
- Bertram Klinger
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isabel Rausch
- Clinic of Hematology and Medical Oncology, University Medical Centre Goettingen, Göttingen, Germany
- ZytoVision GmbH, Bremerhaven, Germany
| | - Anja Sieber
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Helmut Kutz
- Research Unit Gene Vectors, Helmholtz Center Munich—German Research Center for Environmental Health, Munich, Germany
| | - Vanessa Kruse
- Clinic of Hematology and Medical Oncology, University Medical Centre Goettingen, Göttingen, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité—Universitaetsmedizin Berlin and Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité—Universitaetsmedizin Berlin and Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Arnd Kieser
- Research Unit Gene Vectors, Helmholtz Center Munich—German Research Center for Environmental Health, Munich, Germany
- Research Unit Signaling and Translation, Helmholtz Center Munich—German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Kube
- Clinic of Hematology and Medical Oncology, University Medical Centre Goettingen, Göttingen, Germany
| |
Collapse
|
6
|
Meriranta L, Sorri S, Huse K, Liu X, Spasevska I, Zafar S, Chowdhury I, Dufva O, Sahlberg E, Tandarić L, Karjalainen-Lindsberg ML, Hyytiäinen M, Varjosalo M, Myklebust JH, Leppä S. Disruption of KLHL6 Fuels Oncogenic Antigen Receptor Signaling in B-Cell Lymphoma. Blood Cancer Discov 2024; 5:331-352. [PMID: 38630892 PMCID: PMC11369598 DOI: 10.1158/2643-3230.bcd-23-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/31/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Pathomechanisms that activate oncogenic B-cell receptor (BCR) signaling in diffuse large B-cell lymphoma (DLBCL) are largely unknown. Kelch-like family member 6 (KLHL6) encoding a substrate-adapter for Cullin-3-RING E3 ubiquitin ligase with poorly established targets is recurrently mutated in DLBCL. By applying high-throughput protein interactome screens and functional characterization, we discovered that KLHL6 regulates BCR by targeting its signaling subunits CD79A and CD79B. Loss of physiologic KLHL6 expression pattern was frequent among the MCD/C5-like activated B-cell DLBCLs and was associated with higher CD79B levels and dismal outcome. Mutations in the bric-a-brac tramtrack broad domain of KLHL6 disrupted its localization and heterodimerization and increased surface BCR levels and signaling, whereas Kelch domain mutants had the opposite effect. Malfunctions of KLHL6 mutants extended beyond proximal BCR signaling with distinct phenotypes from KLHL6 silencing. Collectively, our findings uncover how recurrent mutations in KLHL6 alter BCR signaling and induce actionable phenotypic characteristics in DLBCL. Significance: Oncogenic BCR signaling sustains DLBCL cells. We discovered that Cullin-3-RING E3 ubiquitin ligase substrate-adapter KLHL6 targets BCR heterodimer (CD79A/CD79B) for ubiquitin-mediated degradation. Recurrent somatic mutations in the KLHL6 gene cause corrupt BCR signaling by disrupting surface BCR homeostasis. Loss of KLHL6 expression and mutant-induced phenotypes associate with targetable disease characteristics in B-cell lymphoma. See related commentary by Leveille et al. See related commentary by Corcoran et al.
Collapse
MESH Headings
- Humans
- Signal Transduction
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- CD79 Antigens/genetics
- CD79 Antigens/metabolism
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Mutation
- Cell Line, Tumor
- Carrier Proteins
Collapse
Affiliation(s)
- Leo Meriranta
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| | - Selma Sorri
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B-cell malignancies and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Xiaonan Liu
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Ivana Spasevska
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B-cell malignancies and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Sadia Zafar
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Iftekhar Chowdhury
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Olli Dufva
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| | - Eerika Sahlberg
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Luka Tandarić
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | | | - Marko Hyytiäinen
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - June H. Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B-cell malignancies and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Sirpa Leppä
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
7
|
Megha KB, Arathi A, Shikha S, Alka R, Ramya P, Mohanan PV. Significance of Melatonin in the Regulation of Circadian Rhythms and Disease Management. Mol Neurobiol 2024; 61:5541-5571. [PMID: 38206471 DOI: 10.1007/s12035-024-03915-0] [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: 10/09/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Melatonin, the 'hormone of darkness' is a neuronal hormone secreted by the pineal gland and other extra pineal sites. Responsible for the circadian rhythm and seasonal behaviour of vertebrates and mammals, melatonin is responsible for regulating various physiological conditions and the maintenance of sleep, body weight and the neuronal activities of the ocular sites. With its unique amphiphilic structure, melatonin can cross the cellular barriers and elucidate its activities in the subcellular components, including mitochondria. Melatonin is a potential scavenger of oxygen and nitrogen-reactive species and can directly obliterate the ROS and RNS by a receptor-independent mechanism. It can also regulate the pro- and anti-inflammatory cytokines in various pathological conditions and exhibit therapeutic activities against neurodegenerative, psychiatric disorders and cancer. Melatonin is also found to show its effects on major organs, particularly the brain, liver and heart, and also imparts a role in the modulation of the immune system. Thus, melatonin is a multifaceted candidate with immense therapeutic potential and is still considered an effective supplement on various therapies. This is primarily due to rectification of aberrant circadian rhythm by improvement of sleep quality associated with risk development of neurodegenerative, cognitive, cardiovascular and other metabolic disorders, thereby enhancing the quality of life.
Collapse
Affiliation(s)
- K B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, 695 012, Kerala, India
| | - A Arathi
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, 695 012, Kerala, India
| | - Saini Shikha
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Rao Alka
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Prabhu Ramya
- P.G. Department of Biotechnology, Government Arts College, Trivandrum, 695 014, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, 695 012, Kerala, India.
| |
Collapse
|
8
|
Melani C, Lakhotia R, Pittaluga S, Phelan JD, Huang DW, Wright G, Simard J, Muppidi J, Thomas CJ, Ceribelli M, Tosto FA, Yang Y, Xu W, Davies-Hill T, Pack SD, Peer CJ, Arisa O, Mena E, Lindenberg L, Bergvall E, Portell CA, Farah RJ, Lee ST, Pradhan A, Morrison C, Tadese A, Juanitez AM, Lu C, Jacob A, Simmons H, Figg WD, Steinberg SM, Jaffe ES, Roschewski M, Staudt LM, Wilson WH. Combination Targeted Therapy in Relapsed Diffuse Large B-Cell Lymphoma. N Engl J Med 2024; 390:2143-2155. [PMID: 38899693 PMCID: PMC11192235 DOI: 10.1056/nejmoa2401532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND The identification of oncogenic mutations in diffuse large B-cell lymphoma (DLBCL) has led to the development of drugs that target essential survival pathways, but whether targeting multiple survival pathways may be curative in DLBCL is unknown. METHODS We performed a single-center, phase 1b-2 study of a regimen of venetoclax, ibrutinib, prednisone, obinutuzumab, and lenalidomide (ViPOR) in relapsed or refractory DLBCL. In phase 1b, which included patients with DLBCL and indolent lymphomas, four dose levels of venetoclax were evaluated to identify the recommended phase 2 dose, with fixed doses of the other four drugs. A phase 2 expansion in patients with germinal-center B-cell (GCB) and non-GCB DLBCL was performed. ViPOR was administered every 21 days for six cycles. RESULTS In phase 1b of the study, involving 20 patients (10 with DLBCL), a single dose-limiting toxic effect of grade 3 intracranial hemorrhage occurred, a result that established venetoclax at a dose of 800 mg as the recommended phase 2 dose. Phase 2 included 40 patients with DLBCL. Toxic effects that were observed among all the patients included grade 3 or 4 neutropenia (in 24% of the cycles), thrombocytopenia (in 23%), anemia (in 7%), and febrile neutropenia (in 1%). Objective responses occurred in 54% of 48 evaluable patients with DLBCL, and complete responses occurred in 38%; complete responses were exclusively in patients with non-GCB DLBCL and high-grade B-cell lymphoma with rearrangements of MYC and BCL2 or BCL6 (or both). Circulating tumor DNA was undetectable in 33% of the patients at the end of ViPOR therapy. With a median follow-up of 40 months, 2-year progression-free survival and overall survival were 34% (95% confidence interval [CI], 21 to 47) and 36% (95% CI, 23 to 49), respectively. CONCLUSIONS Treatment with ViPOR was associated with durable remissions in patients with specific molecular DLBCL subtypes and was associated with mainly reversible adverse events. (Funded by the Intramural Research Program of the National Cancer Institute and the National Center for Advancing Translational Sciences of the National Institutes of Health and others; ClinicalTrials.gov number, NCT03223610.).
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Female
- Middle Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Sulfonamides/adverse effects
- Sulfonamides/administration & dosage
- Sulfonamides/therapeutic use
- Aged
- Male
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Lenalidomide/adverse effects
- Lenalidomide/administration & dosage
- Lenalidomide/therapeutic use
- Piperidines/adverse effects
- Piperidines/therapeutic use
- Piperidines/administration & dosage
- Adult
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Prednisone/adverse effects
- Prednisone/administration & dosage
- Prednisone/therapeutic use
- Adenine/analogs & derivatives
- Adenine/adverse effects
- Adenine/therapeutic use
- Adenine/administration & dosage
- Aged, 80 and over
- Recurrence
- Pyrazoles/adverse effects
- Pyrazoles/therapeutic use
- Pyrazoles/administration & dosage
- Pyrimidines/adverse effects
- Pyrimidines/therapeutic use
- Pyrimidines/administration & dosage
- Molecular Targeted Therapy
- Progression-Free Survival
Collapse
Affiliation(s)
- Christopher Melani
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rahul Lakhotia
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Stefania Pittaluga
- Laboratory of Pathology, Clinical Center, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James D. Phelan
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Da Wei Huang
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - George Wright
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jillian Simard
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jagan Muppidi
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Craig J. Thomas
- Division of Pre-Clinical Innovation Chemistry Technologies, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | - Michele Ceribelli
- Division of Pre-Clinical Innovation Chemistry Technologies, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | - Frances A. Tosto
- Division of Pre-Clinical Innovation Chemistry Technologies, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | - Yandan Yang
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Weihong Xu
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Theresa Davies-Hill
- Laboratory of Pathology, Clinical Center, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Svetlana D. Pack
- Laboratory of Pathology, Clinical Center, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Oluwatobi Arisa
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Esther Mena
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Liza Lindenberg
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ethan Bergvall
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Craig A. Portell
- Division of Hematology and Oncology, University of Virginia, Charlottesville, VA
| | - Rafic J. Farah
- Mario Lemieux Center for Blood Cancers, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Seung Tae Lee
- Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD
| | - Amynah Pradhan
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Candis Morrison
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Atekelt Tadese
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Anna Marie Juanitez
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Crystal Lu
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD
| | | | | | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Elaine S. Jaffe
- Laboratory of Pathology, Clinical Center, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, 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
| | - Wyndham H. Wilson
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
9
|
Negara I, Tomuleasa C, Buruiana S, Efremov DG. Molecular Subtypes and the Role of TP53 in Diffuse Large B-Cell Lymphoma and Richter Syndrome. Cancers (Basel) 2024; 16:2170. [PMID: 38927876 PMCID: PMC11201917 DOI: 10.3390/cancers16122170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid malignancy and a heterogeneous entity comprised of several biologically distinct subtypes. Recently, novel genetic classifications of DLBCL have been resolved based on common mutational patterns indicative of distinct pathways of transformation. However, the complicated and costly nature of the novel classifiers has precluded their inclusion into routine practice. In view of this, the status of the TP53 gene, which is mutated or deleted in 20-30% of the cases, has emerged as an important prognostic factor for DLBCL patients, setting itself apart from other predictors. TP53 genetic lesions are particularly enriched in a genetic subtype of DLBCL that shares genomic features with Richter Syndrome, highlighting the possibility of a subset of DLBCL arising from the transformation of an occult chronic lymphocytic leukemia-like malignancy, such as monoclonal B-cell lymphocytosis. Patients with TP53-mutated DLBCL, including those with Richter Syndrome, have a particularly poor prognosis and display inferior responses to standard chemoimmunotherapy regimens. The data presented in this manuscript argue for the need for improved and more practical risk-stratification models for patients with DLBCL and show the potential for the use of TP53 mutational status for prognostication and, in prospect, treatment stratification in DLBCL.
Collapse
Affiliation(s)
- Ivan Negara
- Molecular Hematology Unit, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
- Department of Internal Medicine, Hematology, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania;
| | - Sanda Buruiana
- Department of Internal Medicine, Hematology, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Dimitar G. Efremov
- Molecular Hematology Unit, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
- Macedonian Academy of Sciences and Arts, 1000 Skopje, North Macedonia
| |
Collapse
|
10
|
Choi J, Ceribelli M, Phelan JD, Häupl B, Huang DW, Wright GW, Hsiao T, Morris V, Ciccarese F, Wang B, Corcoran S, Scheich S, Yu X, Xu W, Yang Y, Zhao H, Zhou J, Zhang G, Muppidi J, Inghirami GG, Oellerich T, Wilson WH, Thomas CJ, Staudt LM. Molecular targets of glucocorticoids that elucidate their therapeutic efficacy in aggressive lymphomas. Cancer Cell 2024; 42:833-849.e12. [PMID: 38701792 PMCID: PMC11168741 DOI: 10.1016/j.ccell.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024]
Abstract
Glucocorticoids have been used for decades to treat lymphomas without an established mechanism of action. Using functional genomic, proteomic, and chemical screens, we discover that glucocorticoids inhibit oncogenic signaling by the B cell receptor (BCR), a recurrent feature of aggressive B cell malignancies, including diffuse large B cell lymphoma and Burkitt lymphoma. Glucocorticoids induce the glucocorticoid receptor (GR) to directly transactivate genes encoding negative regulators of BCR stability (LAPTM5; KLHL14) and the PI3 kinase pathway (INPP5D; DDIT4). GR directly represses transcription of CSK, a kinase that limits the activity of BCR-proximal Src-family kinases. CSK inhibition attenuates the constitutive BCR signaling of lymphomas by hyperactivating Src-family kinases, triggering their ubiquitination and degradation. With the knowledge that glucocorticoids disable oncogenic BCR signaling, they can now be deployed rationally to treat BCR-dependent aggressive lymphomas and used to construct mechanistically sound combination regimens with inhibitors of BTK, PI3 kinase, BCL2, and CSK.
Collapse
MESH Headings
- Humans
- Glucocorticoids/pharmacology
- Receptors, Antigen, B-Cell/metabolism
- Animals
- Signal Transduction/drug effects
- Receptors, Glucocorticoid/metabolism
- Mice
- Cell Line, Tumor
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Burkitt Lymphoma/drug therapy
- Burkitt Lymphoma/genetics
- Burkitt Lymphoma/metabolism
- Burkitt Lymphoma/pathology
- Molecular Targeted Therapy/methods
- Phosphatidylinositol 3-Kinases/metabolism
- src-Family Kinases/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Jaewoo Choi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - James D Phelan
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Björn Häupl
- Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - George W Wright
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tony Hsiao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivian Morris
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Ciccarese
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, 35128 Padova, Italy
| | - Boya Wang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sean Corcoran
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Scheich
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany; University Cancer Center (UCT) Frankfurt, University Hospital, Goethe University, 60590 Frankfurt am Main, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
| | - Xin Yu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Weihong Xu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yandan Yang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hong Zhao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joyce Zhou
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grace Zhang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jagan Muppidi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio G Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Thomas Oellerich
- Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany
| | - Wyndham H Wilson
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Craig J Thomas
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Wang X, Cottrell CA, Hu X, Ray R, Bottermann M, Villavicencio PM, Yan Y, Xie Z, Warner JE, Ellis-Pugh JR, Kalyuzhniy O, Liguori A, Willis JR, Menis S, Rämisch S, Eskandarzadeh S, Kubitz M, Tingle R, Phelps N, Groschel B, Himansu S, Carfi A, Kirsch KH, Weldon SR, Nair U, Schief WR, Batista FD. mRNA-LNP prime boost evolves precursors toward VRC01-like broadly neutralizing antibodies in preclinical humanized mouse models. Sci Immunol 2024; 9:eadn0622. [PMID: 38753808 PMCID: PMC11488661 DOI: 10.1126/sciimmunol.adn0622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Germline-targeting (GT) protein immunogens to induce VRC01-class broadly neutralizing antibodies (bnAbs) to the CD4-binding site of the HIV envelope (Env) have shown promise in clinical trials. Here, we preclinically validated a lipid nanoparticle-encapsulated nucleoside mRNA (mRNA-LNP) encoding eOD-GT8 60mer as a soluble self-assembling nanoparticle in mouse models. In a model with three humanized B cell lineages bearing distinct VRC01-precursor B cell receptors (BCRs) with similar affinities for eOD-GT8, all lineages could be simultaneously primed and undergo diversification and affinity maturation without exclusionary competition. Boosts drove precursor B cell participation in germinal centers; the accumulation of somatic hypermutations, including in key VRC01-class positions; and affinity maturation to boost and native-like antigens in two of the three precursor lineages. We have preclinically validated a prime-boost regimen of soluble self-assembling nanoparticles encoded by mRNA-LNP, demonstrating that multiple lineages can be primed, boosted, and diversified along the bnAb pathway.
Collapse
Affiliation(s)
- Xuesong Wang
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Christopher A. Cottrell
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Xiaozhen Hu
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
- Moderna Inc.; Cambridge, 02139, USA
| | - Rashmi Ray
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Maria Bottermann
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | | | - Yu Yan
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Zhenfei Xie
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - John E. Warner
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | | | - Oleksandr Kalyuzhniy
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Alessia Liguori
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Jordan R. Willis
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Sergey Menis
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Sebastian Rämisch
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Saman Eskandarzadeh
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Michael Kubitz
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Ryan Tingle
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Nicole Phelps
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | - Bettina Groschel
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
| | | | | | - Kathrin H. Kirsch
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Stephanie R. Weldon
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - Usha Nair
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
| | - William R. Schief
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
- Department of Immunology and Microbiology, The Scripps Research Institute; La Jolla, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute; La Jolla, 92037, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute; La Jolla, 92037, USA
- Moderna Inc.; Cambridge, 02139, USA
| | - Facundo D. Batista
- The Ragon Institute of Mass General, MIT, and Harvard; Cambridge, 02139, USA
- Department of Biology, Massachusetts Institute of Technology; Cambridge, 02139, USA
| |
Collapse
|
12
|
Eken JA, Koning MT, Kupcova K, Sepúlveda Yáñez JH, de Groen RA, Quinten E, Janssen J, van Bergen CA, Vermaat JS, Cleven A, Navarrete MA, Ylstra B, de Jong D, Havranek O, Jumaa H, Veelken H. Antigen-independent, autonomous B cell receptor signaling drives activated B cell DLBCL. J Exp Med 2024; 221:e20230941. [PMID: 38512136 PMCID: PMC10959178 DOI: 10.1084/jem.20230941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/29/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
Diffuse large B cell lymphoma of activated B cell type (ABC-DLBCL), a major cell-of-origin DLBCL subtype, is characterized by chronic active B cell receptor (BCR) signaling and NF-κB activation, which can be explained by activating mutations of the BCR signaling cascade in a minority of cases. We demonstrate that autonomous BCR signaling, akin to its essential pathogenetic role in chronic lymphocytic leukemia (CLL), can explain chronic active BCR signaling in ABC-DLBCL. 13 of 18 tested DLBCL-derived BCR, including 12 cases selected for expression of IgM, induced spontaneous calcium flux and increased phosphorylation of the BCR signaling cascade in murine triple knockout pre-B cells without antigenic stimulation or external BCR crosslinking. Autonomous BCR signaling was associated with IgM isotype, dependent on somatic BCR mutations and individual HCDR3 sequences, and largely restricted to non-GCB DLBCL. Autonomous BCR signaling represents a novel immunological oncogenic driver mechanism in DLBCL originating from individual BCR sequences and adds a new dimension to currently proposed genetics- and transcriptomics-based DLBCL classifications.
Collapse
Affiliation(s)
- Janneke A. Eken
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Marvyn T. Koning
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Kristyna Kupcova
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Internal Medicine—Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Julieta H. Sepúlveda Yáñez
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
- School of Medicine, Universidad de Magallanes, Punta Arenas, Chile
| | - Ruben A.L. de Groen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Edwin Quinten
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Jurriaan Janssen
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | | | - Joost S.P. Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Arjen Cleven
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Bauke Ylstra
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Daphne de Jong
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Internal Medicine—Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hassan Jumaa
- Institute of Immunology, University of Ulm, Ulm, Germany
| | - Hendrik Veelken
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
13
|
Lai Y, Guo S, Tang Q, Chang G, Zhang H, Li B, Feng Q, Hu K, Xu Z, Gao X, Zhang Q, Yi H, Song D, Zhang Y, Peng Y, Cai H, Zhu W, Shi J. Dihydrocelastrol induces cell death and suppresses angiogenesis through BCR/AP-1/junb signalling in diffuse large B cell lymphoma. Arch Biochem Biophys 2024; 754:109929. [PMID: 38367794 DOI: 10.1016/j.abb.2024.109929] [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: 12/07/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 02/19/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma. Although treatment options have improved, a large proportion of patients show low survival rates, highlighting an urgent need for novel therapeutic strategies. The aim of this study was to investigate the efficacy of the new small-molecule compound dihydrocelastrol (DHCE), acquired through the structural modification of celastrol (CE), in the treatment of DLBCL. DHCE showed potent anti-lymphoma efficacy and synergistic effects with doxorubicin. DHCE triggered DLBCL cell apoptosis and G0/G1-phase blockade, thereby hindering angiogenesis. DHCE inhibited B-cell receptor cascade signalling and Jun B and p65 nuclear translocation, thereby suppressing pro-tumourigenic signalling. Finally, DHCE exerted lower toxicity than CE, which showed severe hepatic, renal, and reproductive toxicity in vivo. Our findings support further investigation of the clinical efficacy of DHCE against DLBCL.
Collapse
Affiliation(s)
- Yue Lai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Shushan Guo
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qiongwei Tang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Gaomei Chang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qilin Feng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qikai Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hongfei Yi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Dongliang Song
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yifei Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Peng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Haiyan Cai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
14
|
Tkachenko A, Kupcova K, Havranek O. B-Cell Receptor Signaling and Beyond: The Role of Igα (CD79a)/Igβ (CD79b) in Normal and Malignant B Cells. Int J Mol Sci 2023; 25:10. [PMID: 38203179 PMCID: PMC10779339 DOI: 10.3390/ijms25010010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
B-cell receptor (BCR) is a B cell hallmark surface complex regulating multiple cellular processes in normal as well as malignant B cells. Igα (CD79a)/Igβ (CD79b) are essential components of BCR that are indispensable for its functionality, signal initiation, and signal transduction. CD79a/CD79b-mediated BCR signaling is required for the survival of normal as well as malignant B cells via a wide signaling network. Recent studies identified the great complexity of this signaling network and revealed the emerging role of CD79a/CD79b in signal integration. In this review, we have focused on functional features of CD79a/CD79b, summarized signaling consequences of CD79a/CD79b post-translational modifications, and highlighted specifics of CD79a/CD79b interactions within BCR and related signaling cascades. We have reviewed the complex role of CD79a/CD79b in multiple aspects of normal B cell biology and how is the normal BCR signaling affected by lymphoid neoplasms associated CD79A/CD79B mutations. We have also summarized important unresolved questions and highlighted issues that remain to be explored for better understanding of CD79a/CD79b-mediated signal transduction and the eventual identification of additional therapeutically targetable BCR signaling vulnerabilities.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Kristyna Kupcova
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
- First Department of Internal Medicine–Hematology, General University Hospital and First Faculty of Medicine, Charles University, 128 08 Prague, Czech Republic
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
- First Department of Internal Medicine–Hematology, General University Hospital and First Faculty of Medicine, Charles University, 128 08 Prague, Czech Republic
| |
Collapse
|
15
|
Yan C, He X, Qi R, Cao L, Zheng S, Huang C, Yang P, Wang J, Zhu M, Li S, Dong G, Jing H, Zhang W, Liu X. Prediction and prognostic potential of NR3C1 gene expression level in DLBCL patients. Hematology 2023; 28:2251199. [PMID: 37650932 DOI: 10.1080/16078454.2023.2251199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Objective: Diffuse Large B-Cell Lymphoma (DLBCL) is a common and frequently occurring subtype of Non-Hodgkin Lymphoma (NHL). The effective treatment and prognosis of DLBCL are still urgently needed to be explored. This article aims to shed light on the connection between DLBCL survival and NR3C1 expression levels. Methods: First, we divided the 952 DLBCL patients into an NR3C1 high-expression group and an NR3C1 low-expression group and compared the baseline characteristics of the two groups. Second, we used multivariate analysis to predict the dependent variable for age, pathology, ECOG score, lactate dehydrogenase (LDH) ratio, and NR3C1 expression level. Finally, we analyzed the progression-free survival (PFS) and overall survival rate (OS) of DLBCL patients with high or low NR3C1 expression. Results: DLBCL patients with high NR3C1 expression had a better prognosis than those with low NR3C1 expression (OS, P < 0.0001). In DLBCL patients of CHOP therapy, high NR3C1 expression was associated with a good survival prognosis in OS (OS, P = 0.028). Conclusion: In multivariate analysis, NR3C1 high expression was an independent prognostic factor that predicted a longer OS of DLBCL (OS, P = 0.0003). NR3C1 is considered an independent predictor of DLBCL patients and can be used as a biomarker for the prognosis of DLBCL.
Collapse
Affiliation(s)
- Changjian Yan
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| | - Xue He
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ruiying Qi
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Ling Cao
- Nanyang Second General Hospital, Nanyang, People's Republic of China
| | - Siping Zheng
- Gannan Medical University, Ganzhou, People's Republic of China
| | - Chunyuan Huang
- Gannan Medical University, Ganzhou, People's Republic of China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Jing Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Mingxia Zhu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Shaoxiang Li
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Xiaoni Liu
- Department of Respiratory Medicine, First Affiliated Hospital Gannan Medical University, Ganzhou, People's Republic of China
| |
Collapse
|
16
|
Mamidi MK, Huang J, Honjo K, Li R, Tabengwa EM, Neeli I, Randall NL, Ponnuchetty MV, Radic M, Leu CM, Davis RS. FCRL1 immunoregulation in B cell development and malignancy. Front Immunol 2023; 14:1251127. [PMID: 37822931 PMCID: PMC10562807 DOI: 10.3389/fimmu.2023.1251127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/01/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapeutic targeting of surface regulatory proteins and pharmacologic inhibition of critical signaling pathways has dramatically shifted our approach to the care of individuals with B cell malignancies. This evolution in therapy reflects the central role of the B cell receptor (BCR) signaling complex and its co-receptors in the pathogenesis of B lineage leukemias and lymphomas. Members of the Fc receptor-like gene family (FCRL1-6) encode cell surface receptors with complex tyrosine-based regulation that are preferentially expressed by B cells. Among them, FCRL1 expression peaks on naïve and memory B cells and is unique in terms of its intracellular co-activation potential. Recent studies in human and mouse models indicate that FCRL1 contributes to the formation of the BCR signalosome, modulates B cell signaling, and promotes humoral responses. Progress in understanding its regulatory properties, along with evidence for its over-expression by mature B cell leukemias and lymphomas, collectively imply important yet unmet opportunities for FCRL1 in B cell development and transformation. Here we review recent advances in FCRL1 biology and highlight its emerging significance as a promising biomarker and therapeutic target in B cell lymphoproliferative disorders.
Collapse
Affiliation(s)
- Murali K. Mamidi
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jifeng Huang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kazuhito Honjo
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ran Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Edlue M. Tabengwa
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nar’asha L. Randall
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Manasa V. Ponnuchetty
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chuen-Miin Leu
- Institute of Microbiology and Immunology, National Yang Ming ChiaoTung University, Taipei, Taiwan
| | - Randall S. Davis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Departments of Microbiology, and Biochemistry & Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
17
|
Karatrasoglou EA, Dimou M, Piperidou A, Lakiotaki E, Korkolopoulou P, Vassilakopoulos TP. The Role of mTOR in B Cell Lymphoid Malignancies: Biologic and Therapeutic Aspects. Int J Mol Sci 2023; 24:14110. [PMID: 37762410 PMCID: PMC10531792 DOI: 10.3390/ijms241814110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Non-Hodgkin lymphoma's (NHL) incidence is rising over time, and B cell lymphomas comprise the majority of lymphomas. The phosphoinositide 3-kinase (PI3K)/v-akt murine thymoma viral oncogene homologue 1 (Akt)/mammalian target of the rapamycin (mTOR) signaling pathway plays a critical role in a variety of cellular processes, such as cell proliferation and survival. Its role in lymphomagenesis is confirmed in many different types of B cell lymphomas. This review is mainly focused on the PI3K/v-akt/mTOR pathway-related oncogenic mechanisms in B cell NHLs with an emphasis on common B cell lymphoma types [diffuse large B cell lymphoma (DLBCL) and mantle cell lymphoma (MCL)]. Furthermore, it summarizes the literature regarding the clinical applications of the mTOR inhibitors temsirolimus and everolimus in B cell NHLs, which have been tested in a range of clinical trials enrolling patients with B cell malignancies, either as monotherapy or in combination with other agents or regimens.
Collapse
Affiliation(s)
- Eleni A. Karatrasoglou
- First Department of Pathology, National and Kapodistrian University of Athens, Laikon General Hospital, 15773 Athens, Greece; (E.L.); (P.K.)
| | - Maria Dimou
- Department of Hematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, 15773 Athens, Greece; (M.D.); (A.P.); (T.P.V.)
| | - Alexia Piperidou
- Department of Hematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, 15773 Athens, Greece; (M.D.); (A.P.); (T.P.V.)
| | - Eleftheria Lakiotaki
- First Department of Pathology, National and Kapodistrian University of Athens, Laikon General Hospital, 15773 Athens, Greece; (E.L.); (P.K.)
| | - Penelope Korkolopoulou
- First Department of Pathology, National and Kapodistrian University of Athens, Laikon General Hospital, 15773 Athens, Greece; (E.L.); (P.K.)
| | - Theodoros P. Vassilakopoulos
- Department of Hematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, 15773 Athens, Greece; (M.D.); (A.P.); (T.P.V.)
| |
Collapse
|
18
|
Cox MC, Esposito F, Postorino M, Venditti A, Di Napoli A. Serum Paraprotein Is Associated with Adverse Prognostic Factors and Outcome, across Different Subtypes of Mature B-Cell Malignancies-A Systematic Review. Cancers (Basel) 2023; 15:4440. [PMID: 37760410 PMCID: PMC10527377 DOI: 10.3390/cancers15184440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/20/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The presence of a serum paraprotein (PP) is usually associated with plasma-cell dyscrasias, Waldenstrom Macroglobulinemia/lymphoplasmacytic lymphoma, and cryoglobulinemia. However, PP is also often reported in other high- and low-grade B-cell malignancies. As these reports are sparse and heterogeneous, an overall view on this topic is lacking, Therefore, we carried out a complete literature review to detail the characteristics, and highlight differences and similarities among lymphoma entities associated with PP. In these settings, IgM and IgG are the prevalent PP subtypes, and their serum concentration is often low or even undetectable without immunofixation. The relevance of paraproteinemia and its prevalence, as well as the impact of IgG vs. IgM PP, seems to differ within B-NHL subtypes and CLL. Nonetheless, paraproteinemia is almost always associated with advanced disease, as well as with immunophenotypic, genetic, and clinical features, impacting prognosis. In fact, PP is reported as an independent prognostic marker of poor outcome. All the above call for implementing clinical practice, with the assessment of paraproteinemia, in patients' work-up. Indeed, more studies are needed to shed light on the biological mechanism causing more aggressive disease. Furthermore, the significance of paraproteinemia, in the era of targeted therapies, should be assessed in prospective trials.
Collapse
Affiliation(s)
- Maria Christina Cox
- UOC Malattie Linfoproliferative, Fondazione Policlinico Tor Vergata, 00133 Roma, Italy
| | - Fabiana Esposito
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, 00133 Roma, Italy; (F.E.)
| | - Massimiliano Postorino
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, 00133 Roma, Italy; (F.E.)
| | - Adriano Venditti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, 00133 Roma, Italy; (F.E.)
| | - Arianna Di Napoli
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University, 00189 Roma, Italy;
| |
Collapse
|
19
|
Qin Y, Qiu T, Xie Z, Chen X, Liu P, Yang J, He X, Gui L, Zhou S, Jiang H, Zhang C, Yang S, Tang L, Shi Y. MYD88 L265P and MYD88 other variants show different molecular characteristics and prognostic significance in diffuse large B-cell lymphoma. J Cancer Res Clin Oncol 2023; 149:8483-8494. [PMID: 37093346 PMCID: PMC10374827 DOI: 10.1007/s00432-023-04714-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/18/2023] [Indexed: 04/25/2023]
Abstract
PURPOSE This study aims to investigate the clinical and molecular differences between diffuse large B-cell lymphoma (DLBCL) patients with MYD88L265P and MYD88other. METHODS DLBCL patients with MYD88 variations were collected from the Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CHCAMS), and Suzhou Municipal Hospital from February 6th, 2007 to May 20th, 2022. Clinicopathological parameters and treatment outcomes between MYD88L265P and MYD88other were investigated. RESULTS A total of 132 patients with MYD88 variations from a cohort of 475 DLBCL patients were included, among which, 78 were MYD88L265P, while 54 were MYD88other. MYD88L265P was more common in non-GCB subtype than MYD88other (83% vs. 60%, P = 0.004). Besides, MYD88L265P was significantly related to higher proportion of testicle/ central nervous system involvement (31% vs. 6%, P < 0.001), PIM1 mutation (71% vs. 39%, P < 0.001), and PIM1 hypermutation (28% vs. 11%, P = 0.018), compared with MYD88other. Compared with MYD88L265P, MYD88other were more likely to have higher percentage of advanced stage (60% vs. 42%, P = 0.044), extranodal site ≥ 2 (45% vs. 28%, P = 0.044), elevated LDH (55% vs. 35%, P = 0.033), positive CD10 expression (36% vs. 16%, P = 0.009), BCL-6 translocation (20% vs. 8%, P = 0.033), and NOTCH pathway gene alteration (24% vs. 13%, P = 0.040). In non-GCB DLBCL subtype, patients with MYD88other were significantly associated with worse progression free survival (PFS) than those with MYD88L265P when treated initially with R-CHOP/R-CHOP-like regimen (P = 0.010). CONCLUSION The findings of this study indicate that DLBCL patients with MYD88L265P and MYD88other are likely to be two subgroups with different clinical and molecular characteristics. The survival of patients with MYD88other is not superior than those with MYD88L265P, even poorer when focusing on the non-GCB subtype.
Collapse
Affiliation(s)
- Yan Qin
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Zucheng Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Xinrui Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Peng Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Jianliang Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Xiaohui He
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Lin Gui
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Shengyu Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Hongxin Jiang
- Department of Medical Oncology, Suzhou Municipal Hospital, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, China
| | - Changgong Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Sheng Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, 100021, China.
| |
Collapse
|
20
|
Scheich S, Chen J, Liu J, Schnütgen F, Enssle JC, Ceribelli M, Thomas CJ, Choi J, Morris V, Hsiao T, Nguyen H, Wang B, Bolomsky A, Phelan JD, Corcoran S, Urlaub H, Young RM, Häupl B, Wright GW, Huang DW, Ji Y, Yu X, Xu W, Yang Y, Zhao H, Muppidi J, Pan KT, Oellerich T, Staudt LM. Targeting N-linked Glycosylation for the Therapy of Aggressive Lymphomas. Cancer Discov 2023; 13:1862-1883. [PMID: 37141112 PMCID: PMC10524254 DOI: 10.1158/2159-8290.cd-22-1401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 05/02/2023] [Indexed: 05/05/2023]
Abstract
Diffuse large B-cell lymphoma (DLBCL) can be subdivided into the activated B-cell (ABC) and germinal center B cell-like (GCB) subtypes. Self-antigen engagement of B-cell receptors (BCR) in ABC tumors induces their clustering, thereby initiating chronic active signaling and activation of NF-κB and PI3 kinase. Constitutive BCR signaling is essential in some GCB tumors but primarily activates PI3 kinase. We devised genome-wide CRISPR-Cas9 screens to identify regulators of IRF4, a direct transcriptional target of NF-κB and an indicator of proximal BCR signaling in ABC DLBCL. Unexpectedly, inactivation of N-linked protein glycosylation by the oligosaccharyltransferase-B (OST-B) complex reduced IRF4 expression. OST-B inhibition of BCR glycosylation reduced BCR clustering and internalization while promoting its association with CD22, which attenuated PI3 kinase and NF-κB activation. By directly interfering with proximal BCR signaling, OST-B inactivation killed models of ABC and GCB DLBCL, supporting the development of selective OST-B inhibitors for the treatment of these aggressive cancers. SIGNIFICANCE DLBCL depends on constitutive BCR activation and signaling. There are currently no therapeutics that target the BCR directly and attenuate its pathologic signaling. Here, we unraveled a therapeutically exploitable, OST-B-dependent glycosylation pathway that drives BCR organization and proximal BCR signaling. This article is highlighted in the In This Issue feature, p. 1749.
Collapse
Affiliation(s)
- Sebastian Scheich
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Jiamin Liu
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Frank Schnütgen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Julius C. Enssle
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Craig J. Thomas
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Jaewoo Choi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivian Morris
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Johns Hopkins University, Department of Biology, Baltimore, MD, 21218, USA
| | - Tony Hsiao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hang Nguyen
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Boya Wang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arnold Bolomsky
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James D. Phelan
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sean Corcoran
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
- Bioanalytics, Institute of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Ryan M. Young
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Björn Häupl
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - George W. Wright
- Biometric Research Branch, Division of Cancer Diagnosis and Treatment, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yanlong Ji
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Xin Yu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Weihong Xu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yandan Yang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hong Zhao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jagan Muppidi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kuan-Ting Pan
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Thomas Oellerich
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
21
|
Rovsing AB, Thomsen EA, Nielsen I, Skov TW, Luo Y, Dybkaer K, Mikkelsen JG. Resistance to vincristine in DLBCL by disruption of p53-induced cell cycle arrest and apoptosis mediated by KIF18B and USP28. Br J Haematol 2023; 202:825-839. [PMID: 37190875 DOI: 10.1111/bjh.18872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/21/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023]
Abstract
The frontline therapy R-CHOP for patients with diffuse large B-cell lymphoma (DLBCL) has remained unchanged for two decades despite numerous Phase III clinical trials investigating new alternatives. Multiple large studies have uncovered genetic subtypes of DLBCL enabling a targeted approach. To further pave the way for precision oncology, we perform genome-wide CRISPR screening to uncover the cellular response to one of the components of R-CHOP, vincristine, in the DLBCL cell line SU-DHL-5. We discover important pathways and subnetworks using gene-set enrichment analysis and protein-protein interaction networks and identify genes related to mitotic spindle organization that are essential during vincristine treatment. The inhibition of KIF18A, a mediator of chromosome alignment, using the small molecule inhibitor BTB-1 causes complete cell death in a synergistic manner when administered together with vincristine. We also identify the genes KIF18B and USP28 of which CRISPR/Cas9-directed knockout induces vincristine resistance across two DLBCL cell lines. Mechanistic studies show that lack of KIF18B or USP28 counteracts a vincristine-induced p53 response suggesting that resistance to vincristine has origin in the mitotic surveillance pathway (USP28-53BP1-p53). Collectively, our CRISPR screening data uncover potential drug targets and mechanisms behind vincristine resistance, which may support the development of future drug regimens.
Collapse
Affiliation(s)
| | | | - Ian Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI-Shenzhen, Shenzhen, China
| | - Karen Dybkaer
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | | |
Collapse
|
22
|
Melnik BC, Stadler R, Weiskirchen R, Leitzmann C, Schmitz G. Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma. Int J Mol Sci 2023; 24:ijms24076102. [PMID: 37047075 PMCID: PMC10094152 DOI: 10.3390/ijms24076102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Epidemiological evidence supports an association between cow’s milk consumption and the risk of diffuse large B-cell lymphoma (DLBCL), the most common non-Hodgkin lymphoma worldwide. This narrative review intends to elucidate the potential impact of milk-related agents, predominantly milk-derived exosomes (MDEs) and their microRNAs (miRs) in lymphomagenesis. Upregulation of PI3K-AKT-mTORC1 signaling is a common feature of DLBCL. Increased expression of B cell lymphoma 6 (BCL6) and suppression of B lymphocyte-induced maturation protein 1 (BLIMP1)/PR domain-containing protein 1 (PRDM1) are crucial pathological deviations in DLBCL. Translational evidence indicates that during the breastfeeding period, human MDE miRs support B cell proliferation via epigenetic upregulation of BCL6 (via miR-148a-3p-mediated suppression of DNA methyltransferase 1 (DNMT1) and miR-155-5p/miR-29b-5p-mediated suppression of activation-induced cytidine deaminase (AICDA) and suppression of BLIMP1 (via MDE let-7-5p/miR-125b-5p-targeting of PRDM1). After weaning with the physiological termination of MDE miR signaling, the infant’s BCL6 expression and B cell proliferation declines, whereas BLIMP1-mediated B cell maturation for adequate own antibody production rises. Because human and bovine MDE miRs share identical nucleotide sequences, the consumption of pasteurized cow’s milk in adults with the continued transfer of bioactive bovine MDE miRs may de-differentiate B cells back to the neonatal “proliferation-dominated” B cell phenotype maintaining an increased BLC6/BLIMP1 ratio. Persistent milk-induced epigenetic dysregulation of BCL6 and BLIMP1 expression may thus represent a novel driving mechanism in B cell lymphomagenesis. Bovine MDEs and their miR cargo have to be considered potential pathogens that should be removed from the human food chain.
Collapse
|
23
|
CHEK2 Alterations in Pediatric Malignancy: A Single-Institution Experience. Cancers (Basel) 2023; 15:cancers15061649. [PMID: 36980535 PMCID: PMC10046043 DOI: 10.3390/cancers15061649] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Background: Approximately 10% of pediatric malignancies are secondary to germline alterations in cancer-predisposing genes. Checkpoint kinase 2 (CHEK2) germline loss-of-function variants have been reported in pediatric cancer patients, but clinical phenotypes and outcomes are poorly described. We present our single-institution experience of pediatric oncology patients with CHEK2 germline alterations, including clinical presentations and outcomes. Methods: Pediatric oncology patients with CHEK2 germline alterations were identified among those assessed by clinical or translational research at the Institute for Genomic Medicine at Nationwide Children’s Hospital. A chart review of disease course was conducted on identified patients. Results: We identified 6 patients with germline CHEK2 variants from a cohort of 300 individuals, including 1 patient with concurrent presentation of Burkitt lymphoma and neuroblastoma, 3 patients with brain tumors, 1 patient with Ewing sarcoma, and 1 patient with myelodysplastic syndrome. Three patients had a family history of malignancies. Four patients were in remission; one was undergoing treatment; one patient had developed treatment-related meningiomas. We review prior data regarding CHEK2 variants in this population, challenges associated with variant interpretation, and genetic counseling for individuals with CHEK2 variants. Conclusions: CHEK2 germline loss-of-function alterations occur in patients with a variety of pediatric tumors. Larger multicenter studies will improve our understanding of the incidence, phenotype, and molecular biology of CHEK2 germline variants in pediatric cancers.
Collapse
|
24
|
Tuning charge density of chimeric antigen receptor optimizes tonic signaling and CAR-T cell fitness. Cell Res 2023; 33:341-354. [PMID: 36882513 PMCID: PMC10156745 DOI: 10.1038/s41422-023-00789-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/10/2023] [Indexed: 03/09/2023] Open
Abstract
Tonic signaling of chimeric antigen receptor (CAR), i.e., the spontaneous CAR activation in the absence of tumor antigen stimulation, is considered to be a pivotal event controlling CAR-T efficacy. However, the molecular mechanism underlying the spontaneous CAR signals remains elusive. Here, we unveil that positively charged patches (PCPs) on the surface of the CAR antigen-binding domain mediate CAR clustering and result in CAR tonic signaling. For CARs with high tonic signaling (e.g., GD2.CAR and CSPG4.CAR), reducing PCPs on CARs or boosting ionic strength in the culture medium during ex vivo CAR-T cell expansion minimizes spontaneous CAR activation and alleviates CAR-T cell exhaustion. In contrast, introducing PCPs into the CAR with weak tonic signaling, such as CD19.CAR, results in improved in vivo persistence and superior antitumor function. These results demonstrate that CAR tonic signaling is induced and maintained by PCP-mediated CAR clustering. Notably, the mutations we generated to alter the PCPs maintain the antigen-binding affinity and specificity of the CAR. Therefore, our findings suggest that the rational tuning of PCPs to optimize tonic signaling and in vivo fitness of CAR-T cells is a promising design strategy for the next-generation CAR.
Collapse
|
25
|
Heat-Shock Proteins in Leukemia and Lymphoma: Multitargets for Innovative Therapeutic Approaches. Cancers (Basel) 2023; 15:cancers15030984. [PMID: 36765939 PMCID: PMC9913431 DOI: 10.3390/cancers15030984] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Heat-shock proteins (HSPs) are powerful chaperones that provide support for cellular functions under stress conditions but also for the homeostasis of basic cellular machinery. All cancer cells strongly rely on HSPs, as they must continuously adapt to internal but also microenvironmental stresses to survive. In solid tumors, HSPs have been described as helping to correct the folding of misfolded proteins, sustain oncogenic pathways, and prevent apoptosis. Leukemias and lymphomas also overexpress HSPs, which are frequently associated with resistance to therapy. HSPs have therefore been proposed as new therapeutic targets. Given the specific biology of hematological malignancies, it is essential to revise their role in this field, providing a more adaptable and comprehensive picture that would help design future clinical trials. To that end, this review will describe the different pathways and functions regulated by HSP27, HSP70, HSP90, and, not least, HSP110 in leukemias and lymphomas.
Collapse
|
26
|
Shimkus G, Nonaka T. Molecular classification and therapeutics in diffuse large B-cell lymphoma. Front Mol Biosci 2023; 10:1124360. [PMID: 36818048 PMCID: PMC9936827 DOI: 10.3389/fmolb.2023.1124360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) encompasses a wide variety of disease states that have to date been subgrouped and characterized based on immunohistochemical methods, which provide limited prognostic value to clinicians and no alteration in treatment regimen. The addition of rituximab to CHOP therapy was the last leap forward in terms of treatment, but regimens currently follow a standardized course when disease becomes refractory with no individualization based on genotype. Research groups are tentatively proposing new strategies for categorizing DLBCL based on genetic abnormalities that are frequently found together to better predict disease course following dysregulation of specific pathways and to deliver targeted treatment. Novel algorithms in combination with next-generation sequencing techniques have identified between 4 and 7 subgroups of DLBCL, depending on the research team, with potentially significant and actionable genetic alterations. Various drugs aimed at pathways including BCR signaling, NF-κB dysfunction, and epigenetic regulation have shown promise in their respective groups and may show initial utility as second or third line therapies to patients with recurrent DLBCL. Implementation of subgroups will allow collection of necessary data to determine which groups are significant, which treatments may be indicated, and will provide better insight to clinicians and patients on specific disease course.
Collapse
Affiliation(s)
- Gaelen Shimkus
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Taichiro Nonaka
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States,Feist-Weiller Cancer Center, Louisiana State University Health Shreveport, Shreveport, LA, United States,*Correspondence: Taichiro Nonaka,
| |
Collapse
|
27
|
Huse K, Bai B, Hilden VI, Bollum LK, Våtsveen TK, Munthe LA, Smeland EB, Irish JM, Wälchli S, Myklebust JH. Mechanism of CD79A and CD79B Support for IgM+ B Cell Fitness through B Cell Receptor Surface Expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2042-2053. [PMID: 36426942 PMCID: PMC9643646 DOI: 10.4049/jimmunol.2200144] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 09/09/2022] [Indexed: 12/31/2022]
Abstract
The BCR consists of surface-bound Ig and a heterodimeric signaling unit comprised of CD79A and CD79B. Upon cognate Ag recognition, the receptor initiates important signals for B cell development and function. The receptor also conveys Ag-independent survival signals termed tonic signaling. Although the requirement of a CD79A/CD79B heterodimer for BCR complex assembly and surface expression is well established based on mice models, few studies have investigated this in human mature B cells. In this study, we found that human tonsillar B cells with high surface expression of IgM or IgG had potentiated BCR signaling compared with BCRlow cells, and high IgM expression in germinal center B cells was associated with reduced apoptosis. We explored the mechanism for IgM surface expression by CRISPR/Cas9-induced deletion of CD79A or CD79B in four B lymphoma cell lines. Deletion of either CD79 protein caused loss of surface IgM in all cell lines and reduced fitness in three. From two cell lines, we generated stable CD79A or CD79B knockout clones and demonstrated that loss of CD79A or CD79B caused a block in N-glycan maturation and accumulation of immature proteins, compatible with retention of BCR components in the endoplasmic reticulum. Rescue experiments with CD79B wild-type restored surface expression of CD79A and IgM with mature glycosylation, whereas a naturally occurring CD79B G137S mutant disrupting CD79A/CD79B heterodimerization did not. Our study highlights that CD79A and CD79B are required for surface IgM expression in human B cells and illuminates the importance of the IgM expression level for signaling and fitness.
Collapse
Affiliation(s)
- Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Baoyan Bai
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Norway
| | - Vera Irene Hilden
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lise K Bollum
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thea K Våtsveen
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Div. of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Ludvig A Munthe
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Div. of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Erlend B Smeland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jonathan Michael Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Cancer Treatment, Oslo University Hospital, Oslo, Norway
| | - June H. Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
28
|
Tang H, Liu Y, Ruan Y, Ge L, Zhang Q. Reconstructed Genome-Scale Metabolic Model Characterizes Adaptive Metabolic Flux Changes in Peripheral Blood Mononuclear Cells in Severe COVID-19 Patients. Int J Mol Sci 2022; 23:12400. [PMID: 36293257 PMCID: PMC9604493 DOI: 10.3390/ijms232012400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a mortal threat to human health. The elucidation of the relationship between peripheral immune cells and the development of inflammation is essential for revealing the pathogenic mechanism of COVID-19 and developing related antiviral drugs. The immune cell metabolism-targeting therapies exhibit a desirable anti-inflammatory effect in some treatment cases. In this study, based on differentially expressed gene (DEG) analysis, a genome-scale metabolic model (GSMM) was reconstructed by integrating transcriptome data to characterize the adaptive metabolic changes in peripheral blood mononuclear cells (PBMCs) in severe COVID-19 patients. Differential flux analysis revealed that metabolic changes such as enhanced aerobic glycolysis, impaired oxidative phosphorylation, fluctuating biogenesis of lipids, vitamins (folate and retinol), and nucleotides played important roles in the inflammation adaptation of PBMCs. Moreover, the main metabolic enzymes such as the solute carrier (SLC) family 2 member 3 (SLC2A3) and fatty acid synthase (FASN), responsible for the reactions with large differential fluxes, were identified as potential therapeutic targets. Our results revealed the inflammation regulation potentials of partial metabolic reactions with differential fluxes and their metabolites. This study provides a reference for developing potential PBMC metabolism-targeting therapy strategies against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Qingye Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
29
|
Henderson J, Havranek O, Ma MCJ, Herman V, Kupcova K, Chrbolkova T, Pacheco-Blanco M, Wang Z, Comer JM, Zal T, Davis RE. Detecting Förster resonance energy transfer in living cells by conventional and spectral flow cytometry. Cytometry A 2022; 101:818-834. [PMID: 34128311 DOI: 10.1002/cyto.a.24472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 01/27/2023]
Abstract
Assays based on Förster resonance energy transfer (FRET) can be used to study many processes in cell biology. Although this is most often done with microscopy for fluorescence detection, we report two ways to measure FRET in living cells by flow cytometry. Using a conventional flow cytometer and the "3-cube method" for intensity-based calculation of FRET efficiency, we measured the enzymatic activity of specific kinases in cells expressing a genetically-encoded reporter. For both AKT and protein kinase A, the method measured kinase activity in time-course, dose-response, and kinetic assays. Using the Cytek Aurora spectral flow cytometer, which applies linear unmixing to emission measured in multiple wavelength ranges, FRET from the same reporters was measured with greater single-cell precision, in real time and in the presence of other fluorophores. Results from gene-knockout studies suggested that spectral flow cytometry might enable the sorting of cells on the basis of FRET. The methods we present provide convenient and flexible options for using FRET with flow cytometry in studies of cell biology.
Collapse
Affiliation(s)
- Jared Henderson
- Department of Lymphoma and Myeloma, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA
| | - Ondrej Havranek
- Department of Lymphoma and Myeloma, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA.,BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.,Department of Hematology, Charles University and General University Hospital, Prague, Czech Republic
| | - Man Chun John Ma
- Department of Lymphoma and Myeloma, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA
| | - Vaclav Herman
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.,Department of Hematology, Charles University and General University Hospital, Prague, Czech Republic
| | - Kristyna Kupcova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Tereza Chrbolkova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | | | - Zhiqiang Wang
- Department of Lymphoma and Myeloma, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA
| | - Justin M Comer
- Department of Lymphoma and Myeloma, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA
| | - Tomasz Zal
- Department of Leukemia, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA.,Department of Translational Molecular Pathology, The University of Texas-MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Arthur R, Wathen A, Lemm EA, Stevenson FK, Forconi F, Linley AJ, Steele AJ, Packham G, Valle-Argos B. BTK-independent regulation of calcium signalling downstream of the B-cell receptor in malignant B-cells. Cell Signal 2022; 96:110358. [PMID: 35597428 DOI: 10.1016/j.cellsig.2022.110358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 11/20/2022]
Abstract
BTK inhibitors (BTKi) have dramatically improved outcomes for patients with chronic lymphocytic leukaemia (CLL) and some forms of B-cell lymphoma. However, new strategies are needed to enhance responses. Here we have performed a detailed analysis of the effects of BTKi on B-cell receptor (BCR)-induced signalling using primary malignant cells from CLL patients and B-lymphoma cell lines. Although BTK is considered as a key activator of PLCγ2, BTKi (ibrutinib and acalabrutinib) failed to fully inhibit calcium responses in CLL samples with strong BCR signalling capacity. This BTKi-resistant calcium signalling was sufficient to engage downstream calcium-dependent transcription and suppress CLL cell apoptosis and was entirely independent of BTK and not just its kinase activity as similar results were obtained using a BTK-degrading PROTAC. BTK-independent calcium signalling was also observed in two B-lymphoma cell lines where BTKi had little effect on the initial phase of the calcium response but did accelerate the subsequent decline in intracellular calcium. In contrast to BTKi, calcium responses were completely blocked by inhibition of SYK in CLL and lymphoma cells. Engagement of BTK-independent calcium responses was associated with BTK-independent phosphorylation of PLCγ2 on Y753 and Y759 in both CLL and lymphoma cells. Moreover, in CLL samples, inhibition of RAC, which can mediate BTK-independent activation of PLCγ2, cooperated with ibrutinib to suppress calcium responses. BTK-independent calcium signalling may limit the effectiveness of BTKi to suppress BCR signalling responses and our results suggest inhibition of SYK or dual inhibition of BTK and RAC as alternative strategies to strengthen pathway blockade.
Collapse
Affiliation(s)
- Rachael Arthur
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Alexander Wathen
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Elizabeth A Lemm
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Freda K Stevenson
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Francesco Forconi
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Adam J Linley
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Institute of Systems, Molecular and Integrative Biology, 5(th) Floor Nuffield Building, Crown Street, Liverpool L69 3BX, United Kingdom
| | - Andrew J Steele
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Graham Packham
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom.
| | - Beatriz Valle-Argos
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| |
Collapse
|
31
|
Hodgkin Lymphoma: Biology and Differential Diagnostic Problem. Diagnostics (Basel) 2022; 12:diagnostics12061507. [PMID: 35741318 PMCID: PMC9221773 DOI: 10.3390/diagnostics12061507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 11/17/2022] Open
Abstract
Hodgkin lymphomas (HLs) are lymphoid neoplasms that are morphologically defined as being composed of dysplastic cells, namely, Hodgkin and Reed–Sternberg cells, in a reactive inflammatory background. The biological nature of HLs has long been unclear; however, our understanding of HL-related genetics and tumor microenvironment interactions is rapidly expanding. For example, cell surface overexpression of programmed cell death 1 ligand 1 (CD274/PD-L1) is now considered a defining feature of an HL subset, and targeting such immune checkpoint molecules is a promising therapeutic option. Still, HLs comprise multiple disease subtypes, and some HL features may overlap with its morphological mimics, posing challenging diagnostic and therapeutic problems. In this review, we summarize the recent advances in understanding the biology of HLs, and discuss approaches to differentiating HL and its mimics.
Collapse
|
32
|
Taylor J, Wilmore S, Marriot S, Rogers-Broadway KR, Fell R, Minton AR, Branch T, Ashton-Key M, Coldwell M, Stevenson FK, Forconi F, Steele AJ, Packham G, Yeomans A. B-cell receptor signaling induces proteasomal degradation of PDCD4 via MEK1/2 and mTORC1 in malignant B cells. Cell Signal 2022; 94:110311. [PMID: 35306137 PMCID: PMC9077442 DOI: 10.1016/j.cellsig.2022.110311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 12/12/2022]
Abstract
B-cell receptor (BCR) signaling plays a major role in the pathogenesis of B-cell malignancies and is an established target for therapy, including in chronic lymphocytic leukemia cells (CLL), the most common B-cell malignancy. We previously demonstrated that activation of BCR signaling in primary CLL cells downregulated expression of PDCD4, an inhibitor of the translational initiation factor eIF4A and a potential tumor suppressor in lymphoma. Regulation of the PDCD4/eIF4A axis appeared to be important for expression of the MYC oncoprotein as MYC mRNA translation was increased following BCR stimulation and MYC protein induction was repressed by pharmacological inhibition of eIF4A. Here we show that MYC expression is also associated with PDCD4 down-regulation in CLL cells in vivo and characterize the signaling pathways that mediate BCR-induced PDCD4 down-regulation in CLL and lymphoma cells. PDCD4 downregulation was mediated by proteasomal degradation as it was inhibited by proteasome inhibitors in both primary CLL cells and B-lymphoma cell lines. In lymphoma cells, PDCD4 degradation was predominantly dependent on signaling via the AKT pathway. By contrast, in CLL cells, both ERK and AKT pathways contributed to PDCD4 down-regulation and dual inhibition using ibrutinib with either MEK1/2 or mTORC1 inhibition was required to fully reverse PDCD4 down-regulation. Consistent with this, dual inhibition of BTK with MEK1/2 or mTORC1 resulted in the strongest inhibition of BCR-induced MYC expression. This study provides important new insight into the regulation of mRNA translation in B-cell malignancies and a rationale for combinations of kinase inhibitors to target translation control and MYC expression.
Collapse
Affiliation(s)
- Joe Taylor
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Sarah Wilmore
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Sophie Marriot
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Karly-Rai Rogers-Broadway
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rachel Fell
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Annabel R Minton
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tom Branch
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Meg Ashton-Key
- Department of Cellular Pathology, Southampton General Hospital, Southampton, United Kingdom
| | - Mark Coldwell
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, United Kingdom
| | - Freda K Stevenson
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Francesco Forconi
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew J Steele
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.
| | - Alison Yeomans
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
33
|
Zhuang Y, Che J, Wu M, Guo Y, Xu Y, Dong X, Yang H. Altered pathways and targeted therapy in double hit lymphoma. J Hematol Oncol 2022; 15:26. [PMID: 35303910 PMCID: PMC8932183 DOI: 10.1186/s13045-022-01249-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/07/2022] [Indexed: 12/20/2022] Open
Abstract
High-grade B-cell lymphoma with translocations involving MYC and BCL2 or BCL6, usually referred to as double hit lymphoma (DHL), is an aggressive hematological malignance with distinct genetic features and poor clinical prognosis. Current standard chemoimmunotherapy fails to confer satisfying outcomes and few targeted therapeutics are available for the treatment against DHL. Recently, the delineating of the genetic landscape in tumors has provided insight into both biology and targeted therapies. Therefore, it is essential to understand the altered signaling pathways of DHL to develop treatment strategies with better clinical benefits. Herein, we summarized the genetic alterations in the two DHL subtypes (DHL-BCL2 and DHL-BCL6). We further elucidate their implications on cellular processes, including anti-apoptosis, epigenetic regulations, B-cell receptor signaling, and immune escape. Ongoing and potential therapeutic strategies and targeted drugs steered by these alterations were reviewed accordingly. Based on these findings, we also discuss the therapeutic vulnerabilities that coincide with these genetic changes. We believe that the understanding of the DHL studies will provide insight into this disease and capacitate the finding of more effective treatment strategies.
Collapse
Affiliation(s)
- Yuxin Zhuang
- Department of Lymphoma, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, People’s Republic of China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People’s Republic of China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, People’s Republic of China
| | - Meijuan Wu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, People’s Republic of China
| | - Yu Guo
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, People’s Republic of China
| | - Yongjin Xu
- Department of Lymphoma, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, People’s Republic of China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People’s Republic of China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, People’s Republic of China
- Cancer Center, Zhejiang University, Hangzhou, People’s Republic of China
| | - Haiyan Yang
- Department of Lymphoma, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, People’s Republic of China
| |
Collapse
|
34
|
Profitós-Pelejà N, Santos JC, Marín-Niebla A, Roué G, Ribeiro ML. Regulation of B-Cell Receptor Signaling and Its Therapeutic Relevance in Aggressive B-Cell Lymphomas. Cancers (Basel) 2022; 14:860. [PMID: 35205606 PMCID: PMC8870007 DOI: 10.3390/cancers14040860] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/27/2023] Open
Abstract
The proliferation and survival signals emanating from the B-cell receptor (BCR) constitute a crucial aspect of mature lymphocyte's life. Dysregulated BCR signaling is considered a potent contributor to tumor survival in different subtypes of B-cell non-Hodgkin lymphomas (B-NHLs). In the last decade, the emergence of BCR-associated kinases as rational therapeutic targets has led to the development and approval of several small molecule inhibitors targeting either Bruton's tyrosine kinase (BTK), spleen tyrosine kinase (SYK), or phosphatidylinositol 3 kinase (PI3K), offering alternative treatment options to standard chemoimmunotherapy, and making some of these drugs valuable assets in the anti-lymphoma armamentarium. Despite their initial effectiveness, these precision medicine strategies are limited by primary resistance in aggressive B-cell lymphoma such as diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL), especially in the case of first generation BTK inhibitors. In these patients, BCR-targeting drugs often fail to produce durable responses, and nearly all cases eventually progress with a dismal outcome, due to secondary resistance. This review will discuss our current understanding of the role of antigen-dependent and antigen-independent BCR signaling in DLBCL and MCL and will cover both approved inhibitors and investigational molecules being evaluated in early preclinical studies. We will discuss how the mechanisms of action of these molecules, and their off/on-target effects can influence their effectiveness and lead to toxicity, and how our actual knowledge supports the development of more specific inhibitors and new, rationally based, combination therapies, for the management of MCL and DLBCL patients.
Collapse
Affiliation(s)
- Núria Profitós-Pelejà
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Juliana Carvalho Santos
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Ana Marín-Niebla
- Department of Hematology, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron University Hospital, 08035 Barcelona, Spain
| | - Gaël Roué
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Marcelo Lima Ribeiro
- Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista 12916-900, Brazil
| |
Collapse
|
35
|
Alfaifi A, Bahashwan S, Alsaadi M, Malhan H, Aqeel A, Al-Kahiry W, Almehdar H, Qadri I. Metabolic Biomarkers in B-Cell Lymphomas for Early Diagnosis and Prediction, as Well as Their Influence on Prognosis and Treatment. Diagnostics (Basel) 2022; 12:394. [PMID: 35204484 PMCID: PMC8871334 DOI: 10.3390/diagnostics12020394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 11/22/2022] Open
Abstract
B-cell lymphomas exhibit a vast variety of clinical and histological characteristics that might complicate the diagnosis. Timely diagnosis is crucial, as treatments for aggressive subtypes are considered successful and frequently curative, whereas indolent B-cell lymphomas are incurable and often need several therapies. The purpose of this review is to explore the current advancements achieved in B-cell lymphomas metabolism and how these indicators help to early detect metabolic changes in B-cell lymphomas and the use of predictive biological markers in refractory or relapsed disease. Since the year 1920, the Warburg effect has been known as an integral part of metabolic reprogramming. Compared to normal cells, cancerous cells require more glucose. These cancer cells undergo aerobic glycolysis instead of oxidative phosphorylation to metabolize glucose and form lactate as an end product. With the help of these metabolic alterations, a novel biomass is generated by the formation of various precursors. An aggressive metabolic phenotype is an aerobic glycolysis that has the advantage of producing high-rate ATP and preparing the biomass for the amino acid, as well as fatty acid, synthesis needed for a rapid proliferation of cells, while aerobic glycolysis is commonly thought to be the dominant metabolism in cancer cells. Later on, many metabolic biomarkers, such as increased levels of lactate dehydrogenase (LDH), plasma lactate, and deficiency of thiamine in B-cell lymphoma patients, were discovered. Various kinds of molecules can be used as biomarkers, such as genes, proteins, or hormones, because they all refer to body health. Here, we focus only on significant metabolic biomarkers in B-cell lymphomas. In conclusion, many metabolic biomarkers have been shown to have clinical validity, but many others have not been subjected to extensive testing to demonstrate their clinical usefulness in B-cell lymphoma. Furthermore, they play an essential role in the discovery of new therapeutic targets.
Collapse
Affiliation(s)
- Abdullah Alfaifi
- Department of Biological Science, Faculty of Science, King AbdulAziz University, Jeddah 21589, Saudi Arabia; (A.A.); (M.A.); (H.A.)
- Fayfa General Hospital, Ministry of Health, Jazan 83581, Saudi Arabia
| | - Salem Bahashwan
- Hematology Research Unit, King Fahad Medical Research Center, King AbdulAziz University, Jeddah 21589, Saudi Arabia;
- Department of Hematology, Faculty of Medicine, King AbdulAziz University, Jeddah 21589, Saudi Arabia
- King AbdulAziz University Hospital, King AbdulAziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed Alsaadi
- Department of Biological Science, Faculty of Science, King AbdulAziz University, Jeddah 21589, Saudi Arabia; (A.A.); (M.A.); (H.A.)
- Hematology Research Unit, King Fahad Medical Research Center, King AbdulAziz University, Jeddah 21589, Saudi Arabia;
| | - Hafiz Malhan
- Prince Mohammed Bin Nasser Hospital, Ministry of Health, Jazan 82943, Saudi Arabia; (H.M.); (A.A.); (W.A.-K.)
| | - Aqeel Aqeel
- Prince Mohammed Bin Nasser Hospital, Ministry of Health, Jazan 82943, Saudi Arabia; (H.M.); (A.A.); (W.A.-K.)
| | - Waiel Al-Kahiry
- Prince Mohammed Bin Nasser Hospital, Ministry of Health, Jazan 82943, Saudi Arabia; (H.M.); (A.A.); (W.A.-K.)
| | - Hussein Almehdar
- Department of Biological Science, Faculty of Science, King AbdulAziz University, Jeddah 21589, Saudi Arabia; (A.A.); (M.A.); (H.A.)
| | - Ishtiaq Qadri
- Department of Biological Science, Faculty of Science, King AbdulAziz University, Jeddah 21589, Saudi Arabia; (A.A.); (M.A.); (H.A.)
| |
Collapse
|
36
|
Zhang X, Duan YT, Wang Y, Zhao XD, Sun YM, Lin DZ, Chen Y, Wang YX, Zhou ZW, Liu YX, Jiang LH, Geng MY, Ding J, Meng LH. SAF-248, a novel PI3Kδ-selective inhibitor, potently suppresses the growth of diffuse large B-cell lymphoma. Acta Pharmacol Sin 2022; 43:209-219. [PMID: 33782541 PMCID: PMC8724319 DOI: 10.1038/s41401-021-00644-1] [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: 12/22/2020] [Accepted: 03/09/2021] [Indexed: 01/03/2023] Open
Abstract
PI3Kδ is expressed predominately in leukocytes and overexpressed in B-cell-related malignances. PI3Kδ has been validated as a promising target for cancer therapy, and specific PI3Kδ inhibitors were approved for clinical practice. However, the substantial toxicity and relatively low efficacy as a monotherapy in diffuse large B-cell lymphoma (DLBCL) limit their clinical use. In this study, we described a novel PI3Kδ inhibitor SAF-248, which exhibited high selectivity for PI3Kδ (IC50 = 30.6 nM) over other PI3K isoforms at both molecular and cellular levels, while sparing most of the other human protein kinases in the kinome profiling. SAF-248 exhibited superior antiproliferative activity against 27 human lymphoma and leukemia cell lines compared with the approved PI3Kδ inhibitor idelalisib. In particular, SAF-248 potently inhibited the proliferation of a panel of seven DLBCL cell lines (with GI50 values < 1 μM in 5 DLBCL cell lines). We demonstrated that SAF-248 concentration-dependently blocked PI3K signaling followed by inducing G1 phase arrest and apoptosis in DLBCL KARPAS-422, Pfeiffer and TMD8 cells. Its activity against the DLBCL cells was negatively correlated to the protein level of PI3Kα. Oral administration of SAF-248 dose-dependently inhibited the growth of xenografts derived from Pfeiffer and TMD8 cells. Activation of mTORC1, MYC and JAK/STAT signaling was observed upon prolonged treatment and co-targeting these pathways would potentiate the activity of SAF-248. Taken together, SAF-248 is a promising selective PI3Kδ inhibitor for the treatment of DLBCL and rational drug combination would further improve its efficacy.
Collapse
Affiliation(s)
- Xi Zhang
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Yu-ting Duan
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yi Wang
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | | | - Yi-ming Sun
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Dong-ze Lin
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Yi Chen
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yu-xiang Wang
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Zu-wen Zhou
- Fochon Pharmaceuticals, Ltd., Chongqing, 404100 China
| | - Yan-xin Liu
- Fochon Pharmaceuticals, Ltd., Chongqing, 404100 China
| | - Li-hua Jiang
- Fochon Pharmaceuticals, Ltd., Chongqing, 404100 China
| | - Mei-yu Geng
- grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.9227.e0000000119573309Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jian Ding
- grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.9227.e0000000119573309Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Ling-hua Meng
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
37
|
Ren A, Sun J, Yin W, Westerberg LS, Miller H, Lee P, Candotti F, Guan F, Lei J, Gong Q, Chen Y, Liu C. Signaling networks in B cell development and related therapeutic strategies. J Leukoc Biol 2021; 111:877-891. [PMID: 34528729 DOI: 10.1002/jlb.2ru0221-088rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
B cells are essential for Ab production during humoral immune responses. From decades of B cell research, there is now a detailed understanding of B cell subsets, development, functions, and most importantly, signaling pathways. The complicated pathways in B cells and their interactions with each other are stage-dependent, varying with surface marker expression during B cell development. With the increasing understanding of B cell development and signaling pathways, the mechanisms underlying B cell related diseases are being unraveled as well, making it possible to provide more precise and effective treatments. In this review, we describe several essential and recently discovered signaling pathways in B cell development and take a look at newly developed therapeutic strategies targeted at B cell signaling.
Collapse
Affiliation(s)
- Anwen Ren
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Heather Miller
- The Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
38
|
Heward J, Konali L, D'Avola A, Close K, Yeomans A, Philpott M, Dunford J, Rahim T, Al Seraihi AF, Wang J, Korfi K, Araf S, Iqbal S, Bewicke-Copley F, Kumar E, Barisic D, Calaminici M, Clear A, Gribben J, Johnson P, Neve R, Cutillas P, Okosun J, Oppermann U, Melnick A, Packham G, Fitzgibbon J. KDM5 inhibition offers a novel therapeutic strategy for the treatment of KMT2D mutant lymphomas. Blood 2021; 138:370-381. [PMID: 33786580 PMCID: PMC8351530 DOI: 10.1182/blood.2020008743] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
Loss-of-function mutations in KMT2D are a striking feature of germinal center (GC) lymphomas, resulting in decreased histone 3 lysine 4 (H3K4) methylation and altered gene expression. We hypothesized that inhibition of the KDM5 family, which demethylates H3K4me3/me2, would reestablish H3K4 methylation and restore the expression of genes repressed on loss of KMT2D. KDM5 inhibition increased H3K4me3 levels and caused an antiproliferative response in vitro, which was markedly greater in both endogenous and gene-edited KMT2D mutant diffuse large B-cell lymphoma cell lines, whereas tumor growth was inhibited in KMT2D mutant xenografts in vivo. KDM5 inhibition reactivated both KMT2D-dependent and -independent genes, resulting in diminished B-cell signaling and altered expression of B-cell lymphoma 2 (BCL2) family members, including BCL2 itself. KDM5 inhibition may offer an effective therapeutic strategy for ameliorating KMT2D loss-of-function mutations in GC lymphomas.
Collapse
Affiliation(s)
- James Heward
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lola Konali
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Annalisa D'Avola
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Karina Close
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Alison Yeomans
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Martin Philpott
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - James Dunford
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Tahrima Rahim
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Ahad F Al Seraihi
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jun Wang
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Koorosh Korfi
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Shamzah Araf
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Sameena Iqbal
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Findlay Bewicke-Copley
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Emil Kumar
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Darko Barisic
- Department of Medicine, Weill Cornell Medicine, New York, NY; and
| | - Maria Calaminici
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Andrew Clear
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - John Gribben
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Peter Johnson
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | | | - Pedro Cutillas
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jessica Okosun
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Udo Oppermann
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Ari Melnick
- Department of Medicine, Weill Cornell Medicine, New York, NY; and
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Jude Fitzgibbon
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
39
|
Hu N, Wang F, Sun T, Xu Z, Zhang J, Bernard D, Xu S, Wang S, Kaminski M, Devata S, Phillips T, Malek SN. Follicular Lymphoma-associated BTK Mutations are Inactivating Resulting in Augmented AKT Activation. Clin Cancer Res 2021; 27:2301-2313. [PMID: 33419778 DOI: 10.1158/1078-0432.ccr-20-3741] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 09/29/2020] [Accepted: 01/05/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE On the basis of the recent discovery of mutations in Bruton tyrosine kinase (BTK) in follicular lymphoma, we studied their functional properties. EXPERIMENTAL DESIGN We identified novel somatic BTK mutations in 7% of a combined total of 139 follicular lymphoma and 11 transformed follicular lymphoma cases, none of which had received prior treatment with B-cell receptor (BCR) targeted drugs. We reconstituted wild-type (WT) and mutant BTK into various engineered lymphoma cell lines. We measured BCR-induced signal transduction events in engineered cell lines and primary human follicular lymphoma B cells. RESULTS We uncovered that all BTK mutants destabilized the BTK protein and some created BTK kinase-dead mutants. The phospholipase C gamma 2 (PLCγ2) is a substrate of BTK but the BTK mutants did not alter PLCγ2 phosphorylation. Instead, we discovered that BTK mutants induced an exaggerated AKT phosphorylation phenotype in anti-Ig-treated recombinant lymphoma cell lines. The short hairpin RNA-mediated knockdown of BTK expression in primary human nonmalignant lymph node-derived B cells resulted in strong anti-Ig-induced AKT activation, as did the degradation of BTK protein in cell lines using ibrutinib-based proteolysis targeting chimera. Finally, through analyses of primary human follicular lymphoma B cells carrying WT or mutant BTK, we detected elevated AKT phosphorylation following surface Ig crosslinking in all follicular lymphoma B cells, including all BTK-mutant follicular lymphoma. The augmented AKT phosphorylation following BCR crosslinking could be abrogated by pretreatment with a PI3Kδ inhibitor. CONCLUSIONS Altogether, our data uncover novel unexpected properties of follicular lymphoma-associated BTK mutations with direct implications for targeted therapy development in follicular lymphoma.See related commentary by Afaghani and Taylor, p. 2123.
Collapse
Affiliation(s)
- Nan Hu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Fangyang Wang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Tianyu Sun
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Zhengfan Xu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jing Zhang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Denzil Bernard
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Shilin Xu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Shaomeng Wang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Mark Kaminski
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Suma Devata
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Tycel Phillips
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Sami N Malek
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
40
|
Berditchevski F, Fennell E, Murray PG. Calcium-dependent signalling in B-cell lymphomas. Oncogene 2021; 40:6321-6328. [PMID: 34625709 PMCID: PMC8585665 DOI: 10.1038/s41388-021-02025-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/24/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022]
Abstract
Induced waves of calcium fluxes initiate multiple signalling pathways that play an important role in the differentiation and maturation of B-cells. Finely tuned transient Ca+2 fluxes from the endoplasmic reticulum in response to B-cell receptor (BCR) or chemokine receptor activation are followed by more sustained calcium influxes from the extracellular environment and contribute to the mechanisms responsible for the proliferation of B-cells, their migration within lymphoid organs and their differentiation. Dysregulation of these well-balanced mechanisms in B-cell lymphomas results in uncontrolled cell proliferation and resistance to apoptosis. Consequently, several cytotoxic drugs (and anti-proliferative compounds) used in standard chemotherapy regimens for the treatment of people with lymphoma target calcium-dependent pathways. Furthermore, ~10% of lymphoma associated mutations are found in genes with functions in calcium-dependent signalling, including those affecting B-cell receptor signalling pathways. In this review, we provide an overview of the Ca2+-dependent signalling network and outline the contribution of its key components to B cell lymphomagenesis. We also consider how the oncogenic Epstein-Barr virus, which is causally linked to the pathogenesis of a number of B-cell lymphomas, can modify Ca2+-dependent signalling.
Collapse
Affiliation(s)
- Fedor Berditchevski
- grid.6572.60000 0004 1936 7486Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT UK
| | - Eanna Fennell
- grid.10049.3c0000 0004 1936 9692Health Research Institute, University of Limerick, Castletroy, Limerick, V94 T9PX Ireland
| | - Paul G. Murray
- grid.10049.3c0000 0004 1936 9692Health Research Institute, University of Limerick, Castletroy, Limerick, V94 T9PX Ireland ,grid.6572.60000 0004 1936 7486Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT UK
| |
Collapse
|
41
|
Kirsch BJ, Chang SJ, Betenbaugh MJ, Le A. Non-Hodgkin Lymphoma Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1311:103-116. [PMID: 34014537 DOI: 10.1007/978-3-030-65768-0_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-Hodgkin lymphomas (NHLs) are a heterogeneous group of lymphoid neoplasms with different biological characteristics. About 90% of all lymphomas in the United States originate from B lymphocytes, while the remaining originate from T cells [1]. The treatment of NHLs depends on the neoplastic histology and stage of the tumor, which will indicate whether radiotherapy, chemotherapy, or a combination is the best suitable treatment [2]. The American Cancer Society describes the staging of lymphoma as follows: Stage I is lymphoma in a single node or area. Stage II is when that lymphoma has spread to another node or organ tissue. Stage III is when it has spread to lymph nodes on two sides of the diaphragm. Stage IV is when cancer has significantly spread to organs outside the lymph system. Radiation therapy is the traditional therapeutic route for localized follicular and mucosa-associated lymphomas. Chemotherapy is utilized for the treatment of large-cell lymphomas and high-grade lymphomas [2]. However, the treatment of indolent lymphomas remains problematic as the patients often have metastasis, for which no standard approach exists [2].
Collapse
Affiliation(s)
- Brian James Kirsch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA
| | - Shu-Jyuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Michael James Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA
| | - Anne Le
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA. .,Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
42
|
Mehta A, Verma A, Gupta G, Tripathi R, Sharma A. Double Hit and Double Expresser Diffuse Large B Cell Lymphoma Subtypes: Discrete Subtypes and Major Predictors of Overall Survival. Indian J Hematol Blood Transfus 2020; 36:627-634. [PMID: 33100703 PMCID: PMC7572989 DOI: 10.1007/s12288-019-01248-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/28/2019] [Indexed: 01/18/2023] Open
Abstract
Double hit lymphomas (DHL) and double expresser lymphomas (DEL) are subsets of diffuse large B cell lymphomas (DLBCL) which are being increasingly recognised as cause of treatment failure. This emphasizes the need for their separation from other DLBCL cases in order to prognosticate and administer more aggressive treatment to this set of patients. The present study was conducted with the aim to identify the DHL/DEL patients and study their distinctive clinicopathological profile and overall survival. This retrospective analysis involved 172 cases of DLBCL sub-classified on the basis of cell of origin. Immunohistochemical (IHC) analysis for MYC, BCL2, BCL6, MUM1 and CD10 was performed. Rearrangement studies were performed using break apart Fluorescent in situ hybridization. Overall survival (OS) was also evaluated. Distinctive clinical and pathological features of DHL and DEL were identified. Rearrangement study by FISH revealed seven cases of DHL (MYC + BCL2 &/or BCL6 rearrangement). Also, 20 patients (11.6%) showed a concurrent expression of BCL2 and MYC oncoproteins (DEL) on IHC. Most (6/7) DHL patients were double expressors also. The DHL patients demonstrated a significant association with female gender, high serum LDH levels (> 750 U/L) and GCB phenotype. DEL patients contrarily predominated amongst males, had intermediate LDH levels (251-500 U/L) and non GCB phenotype. The OS of the patients was 63.8% at 4 years. The OS of the DLBCL, DEL and DHL patients was 71.9%, 46.9%, and 0%, respectively at 4 years (p value 0.010). In case of DEL subtype, factors such as age < 60 years (66.7%), male sex (60.8%), nodal localization (52.5%), early disease stage (84.6%), low IPI score (60%), absence of B symptoms (50%), LDH < 250 U/L (80%) and GCB phenotype (53.3%) were associated with better OS. Further, the OS of DHL cases was 0% at 4 years. Double hit and double expresser lymphomas have poor prognostic outcomes and should be separated from DLBCL. All DELs should be tested for DHLs and especially those with immunoblastic morphology. DHL and DEL subtypes delineate the subtypes with inferior OS and reinstate the need for aggressive interventions.
Collapse
Affiliation(s)
- Anurag Mehta
- Department of Laboratory and Transfusion Services, Rajiv Gandhi Cancer Institute and Research Centre, Rohini, Delhi India
- Molecular Diagnostic Services, Rajiv Gandhi Cancer Institute and Research Centre, Sector-5, Rohini, Delhi 110085 India
| | - Ajita Verma
- Department of Laboratory and Transfusion Services, Rajiv Gandhi Cancer Institute and Research Centre, Rohini, Delhi India
| | - Garima Gupta
- Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, Rohini, Delhi India
| | - Rupal Tripathi
- Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, Rohini, Delhi India
| | - Anurag Sharma
- Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, Rohini, Delhi India
| |
Collapse
|
43
|
Thomsen EA, Rovsing AB, Anderson MV, Due H, Huang J, Luo Y, Dybkaer K, Mikkelsen JG. Identification of BLNK and BTK as mediators of rituximab-induced programmed cell death by CRISPR screens in GCB-subtype diffuse large B-cell lymphoma. Mol Oncol 2020; 14:1978-1997. [PMID: 32585766 PMCID: PMC7463323 DOI: 10.1002/1878-0261.12753] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/15/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Diffuse large B‐cell lymphoma (DLBCL) is characterized by extensive genetic heterogeneity, and this results in unpredictable responses to the current treatment, R‐CHOP, which consists of a cancer drug combination supplemented with the humanized CD20‐targeting monoclonal antibody rituximab. Despite improvements in the patient response rate through rituximab addition to the treatment plan, up to 40% of DLBCL patients end in a relapsed or refractory state due to inherent or acquired resistance to the regimen. Here, we employ a lentiviral genome‐wide clustered regularly interspaced short palindromic repeats library screening approach to identify genes involved in facilitating the rituximab response in cancerous B cells. Along with the CD20‐encoding MS4A1 gene, we identify genes related to B‐cell receptor (BCR) signaling as mediators of the intracellular signaling response to rituximab. More specifically, the B‐cell linker protein (BLNK) and Bruton's tyrosine kinase (BTK) genes stand out as pivotal genes in facilitating direct rituximab‐induced apoptosis through mechanisms that occur alongside complement‐dependent cytotoxicity (CDC). Our findings demonstrate that rituximab triggers BCR signaling in a BLNK‐ and BTK‐dependent manner and support the existing notion that intertwined CD20 and BCR signaling pathways in germinal center B‐cell‐like‐subtype DLBCL lead to programmed cell death.
Collapse
Affiliation(s)
| | | | | | - Hanne Due
- Department of Hematology, Aalborg University Hospital, Denmark
| | - Jinrong Huang
- Department of Biomedicine, Aarhus University, Denmark.,Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI-Shenzhen, China.,Department of Biology, University of Copenhagen, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Denmark.,Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI-Shenzhen, China
| | - Karen Dybkaer
- Department of Hematology, Aalborg University Hospital, Denmark
| | | |
Collapse
|
44
|
Jain N, Hartert K, Tadros S, Fiskus W, Havranek O, Ma MCJ, Bouska A, Heavican T, Kumar D, Deng Q, Moore D, Pak C, Liu CL, Gentles AJ, Hartmann E, Kridel R, Smedby KE, Juliusson G, Rosenquist R, Gascoyne RD, Rosenwald A, Giancotti F, Neelapu SS, Westin J, Vose JM, Lunning MA, Greiner T, Rodig S, Iqbal J, Alizadeh AA, Davis RE, Bhalla K, Green MR. Targetable genetic alterations of TCF4 ( E2-2) drive immunoglobulin expression in diffuse large B cell lymphoma. Sci Transl Med 2020; 11:11/497/eaav5599. [PMID: 31217338 DOI: 10.1126/scitranslmed.aav5599] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/31/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022]
Abstract
The activated B cell (ABC-like) subtype of diffuse large B cell lymphoma (DLBCL) is characterized by chronic activation of signaling initiated by immunoglobulin μ (IgM). By analyzing the DNA copy number profiles of 1000 DLBCL tumors, we identified gains of 18q21.2 as the most frequent genetic alteration in ABC-like DLBCL. Using integrative analysis of matched gene expression profiling data, we found that the TCF4 (E2-2) transcription factor gene was the target of these alterations. Overexpression of TCF4 in ABC-like DLBCL cell lines led to its occupancy on immunoglobulin (IGHM) and MYC gene enhancers and increased expression of these genes at the transcript and protein levels. Inhibition of TCF4 activity with dominant-negative constructs was synthetically lethal to ABC-like DLBCL cell lines harboring TCF4 DNA copy gains, highlighting these gains as an attractive potential therapeutic target. Furthermore, the TCF4 gene was one of the top BRD4-regulated genes in DLBCL cell lines. BET proteolysis-targeting chimera (PROTAC) ARV771 extinguished TCF4, MYC, and IgM expression and killed ABC-like DLBCL cells in vitro. In DLBCL xenograft models, ARV771 treatment reduced tumor growth and prolonged survival. This work highlights a genetic mechanism for promoting immunoglobulin signaling in ABC-like DLBCL and provides a functional rationale for the use of BET inhibitors in this disease.
Collapse
Affiliation(s)
- Neeraj Jain
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keenan Hartert
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Saber Tadros
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Warren Fiskus
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ondrej Havranek
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Man Chun John Ma
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alyssa Bouska
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tayla Heavican
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dhiraj Kumar
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qing Deng
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dalia Moore
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christine Pak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Andrew J Gentles
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Elena Hartmann
- Institute of Pathology, University of Würzburg, Würzburg 97080, Germany.,Comprehensive Cancer Center Mainfranken, Wurzburg 97080, Germany
| | - Robert Kridel
- Princess Margaret Cancer Center, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Karin Ekstrom Smedby
- Department of Medicine, Solna, Clinical Epidemiology Unit, Karolinska Institutet, and Hematology Center, Karolinska University Hospital, Stockholm SE-171 76, Sweden
| | - Gunnar Juliusson
- Department of Laboratory Medicine, Stem Cell Center, Lund University, Lund SE-221 00, Sweden
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Universitetssjukhuset, Stockholm SE-171 76, Sweden
| | - Randy D Gascoyne
- Center for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC V5Z 4E6, Canada
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Würzburg 97080, Germany.,Comprehensive Cancer Center Mainfranken, Wurzburg 97080, Germany
| | - Filippo Giancotti
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jason Westin
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Julie M Vose
- Division of Hematology and Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew A Lunning
- Division of Hematology and Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Timothy Greiner
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Javeed Iqbal
- Department of Pathology and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - R Eric Davis
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kapil Bhalla
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael R Green
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. .,Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
45
|
Mechanisms of B Cell Receptor Activation and Responses to B Cell Receptor Inhibitors in B Cell Malignancies. Cancers (Basel) 2020; 12:cancers12061396. [PMID: 32481736 PMCID: PMC7352865 DOI: 10.3390/cancers12061396] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/27/2022] Open
Abstract
The B cell receptor (BCR) pathway has been identified as a potential therapeutic target in a number of common B cell malignancies, including chronic lymphocytic leukemia, diffuse large B cell lymphoma, Burkitt lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B cell lymphoma, and Waldenstrom's macroglobulinemia. This finding has resulted in the development of numerous drugs that target this pathway, including various inhibitors of the kinases BTK, PI3K, and SYK. Several of these drugs have been approved in recent years for clinical use, resulting in a profound change in the way these diseases are currently being treated. However, the response rates and durability of responses vary largely across the different disease entities, suggesting a different proportion of patients with an activated BCR pathway and different mechanisms of BCR pathway activation. Indeed, several antigen-dependent and antigen-independent mechanisms have recently been described and shown to result in the activation of distinct downstream signaling pathways. The purpose of this review is to provide an overview of the mechanisms responsible for the activation of the BCR pathway in different B cell malignancies and to correlate these mechanisms with clinical responses to treatment with BCR inhibitors.
Collapse
|
46
|
Wright GW, Huang DW, Phelan JD, Coulibaly ZA, Roulland S, Young RM, Wang JQ, Schmitz R, Morin RD, Tang J, Jiang A, Bagaev A, Plotnikova O, Kotlov N, Johnson CA, Wilson WH, Scott DW, Staudt LM. A Probabilistic Classification Tool for Genetic Subtypes of Diffuse Large B Cell Lymphoma with Therapeutic Implications. Cancer Cell 2020; 37:551-568.e14. [PMID: 32289277 PMCID: PMC8459709 DOI: 10.1016/j.ccell.2020.03.015] [Citation(s) in RCA: 632] [Impact Index Per Article: 126.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/03/2020] [Accepted: 03/16/2020] [Indexed: 12/22/2022]
Abstract
The development of precision medicine approaches for diffuse large B cell lymphoma (DLBCL) is confounded by its pronounced genetic, phenotypic, and clinical heterogeneity. Recent multiplatform genomic studies revealed the existence of genetic subtypes of DLBCL using clustering methodologies. Here, we describe an algorithm that determines the probability that a patient's lymphoma belongs to one of seven genetic subtypes based on its genetic features. This classification reveals genetic similarities between these DLBCL subtypes and various indolent and extranodal lymphoma types, suggesting a shared pathogenesis. These genetic subtypes also have distinct gene expression profiles, immune microenvironments, and outcomes following immunochemotherapy. Functional analysis of genetic subtype models highlights distinct vulnerabilities to targeted therapy, supporting the use of this classification in precision medicine trials.
Collapse
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Cell Proliferation
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genetic Heterogeneity
- Humans
- Lymphoma, Large B-Cell, Diffuse/classification
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Targeted Therapy
- Precision Medicine
- Tumor Cells, Cultured
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- George W Wright
- Biometric Research Branch, Division of Cancer Diagnosis and Treatment, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James D Phelan
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zana A Coulibaly
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandrine Roulland
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan M Young
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James Q Wang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Roland Schmitz
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan D Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Jeffrey Tang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Aixiang Jiang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | | | | | | | - Calvin A Johnson
- Office of Intramural Research, Center for Information Technology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wyndham H Wilson
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David W Scott
- British Columbia Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Lemm EA, Valle-Argos B, Smith LD, Richter J, Gebreselassie Y, Carter MJ, Karolova J, Svaton M, Helman K, Weston-Bell NJ, Karydis L, Williamson CT, Lenz G, Pettigrew J, Harwig C, Stevenson FK, Cragg M, Forconi F, Steele AJ, Cross J, Mackenzie L, Klener P, Packham G. Preclinical Evaluation of a Novel SHIP1 Phosphatase Activator for Inhibition of PI3K Signaling in Malignant B Cells. Clin Cancer Res 2020; 26:1700-1711. [PMID: 31831562 PMCID: PMC7124891 DOI: 10.1158/1078-0432.ccr-19-2202] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/23/2019] [Accepted: 12/09/2019] [Indexed: 01/09/2023]
Abstract
PURPOSE PI3K signaling is a common feature of B-cell neoplasms, including chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL), and PI3K inhibitors have been introduced into the clinic. However, there remains a clear need to develop new strategies to target PI3K signaling. PI3K activity is countered by Src homology domain 2-containing inositol-5'-phosphatase 1 (SHIP1) and, here, we have characterized the activity of a novel SHIP1 activator, AQX-435, in preclinical models of B-cell malignancies. EXPERIMENTAL DESIGN In vitro activity of AQX-435 was evaluated using primary CLL cells and DLBCL-derived cell lines. In vivo activity of AQX-435, alone or in combination with the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib, was assessed using DLBCL cell line and patient-derived xenograft models. RESULTS Pharmacologic activation of SHIP1 using AQX-435 was sufficient to inhibit anti-IgM-induced PI3K-mediated signaling, including induction of AKT phosphorylation and MYC expression, without effects on upstream SYK phosphorylation. AQX-435 also cooperated with the BTK inhibitor ibrutinib to enhance inhibition of anti-IgM-induced AKT phosphorylation. AQX-435 induced caspase-dependent apoptosis of CLL cells preferentially as compared with normal B cells, and overcame in vitro survival-promoting effects of microenvironmental stimuli. Finally, AQX-435 reduced AKT phosphorylation and growth of DLBCL in vivo and cooperated with ibrutinib for tumor growth inhibition. CONCLUSIONS Our results using AQX-435 demonstrate that SHIP1 activation may be an effective novel therapeutic strategy for treatment of B-cell neoplasms, alone or in combination with ibrutinib.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Cell Line, Tumor
- Enzyme Activators/pharmacology
- Female
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mice
- Mice, Inbred NOD
- Phosphatidylinositol 3-Kinases/chemistry
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
- Sesquiterpenes/pharmacology
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Elizabeth A Lemm
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Beatriz Valle-Argos
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Lindsay D Smith
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Johanna Richter
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Yohannes Gebreselassie
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Matthew J Carter
- Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jana Karolova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- CLIP - Childhood Leukaemia Investigation Prague, Second Faculty of Medicine and Charles University Hospital in Motol, Prague, Czech Republic
| | - Michael Svaton
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- CLIP - Childhood Leukaemia Investigation Prague, Second Faculty of Medicine and Charles University Hospital in Motol, Prague, Czech Republic
| | - Karel Helman
- Faculty of Informatics and Statistics, University of Economics, Prague, Czech Republic
| | - Nicola J Weston-Bell
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Laura Karydis
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Chris T Williamson
- Aquinox Pharmaceuticals (Canada) Inc., Vancouver, British Columbia, Canada
| | - Georg Lenz
- Department of Medicine A for Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Jeremy Pettigrew
- Aquinox Pharmaceuticals (Canada) Inc., Vancouver, British Columbia, Canada
| | - Curtis Harwig
- Aquinox Pharmaceuticals (Canada) Inc., Vancouver, British Columbia, Canada
| | - Freda K Stevenson
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mark Cragg
- Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Francesco Forconi
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew J Steele
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jennifer Cross
- Aquinox Pharmaceuticals (Canada) Inc., Vancouver, British Columbia, Canada
| | - Lloyd Mackenzie
- Aquinox Pharmaceuticals (Canada) Inc., Vancouver, British Columbia, Canada
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- CLIP - Childhood Leukaemia Investigation Prague, Second Faculty of Medicine and Charles University Hospital in Motol, Prague, Czech Republic
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.
| |
Collapse
|
48
|
Sun F, Fang X, Wang X. Signal Pathways and Therapeutic Prospects of Diffuse Large B Cell Lymphoma. Anticancer Agents Med Chem 2020; 19:2047-2059. [PMID: 32009599 DOI: 10.2174/1871520619666190925143216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/18/2019] [Accepted: 07/18/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Diffuse Large B Cell Lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma which is heterogeneous both clinically and morphologically. Over the past decades, significant advances have been made in the understanding of the molecular genesis, leading to the identification of multiple pathways and molecules that can be targeted for clinical benefit. OBJECTIVE The current review aims to present a brief overview of signal pathways of DLBCL, which mainly focus on B-cell antigen Receptor (BCR), Nuclear Factor-κB (NF-κB), Phosphatidylinositol-3-Kinase (PI3K) - protein kinase B (Akt) - mammalian Target of Rapamycin (mTOR), Janus Kinase (JAK) - Signal Transducer and Activator (STAT), Wnt/β-catenin, and P53 pathways. METHODS Activation of signal pathways may contribute to the generation, development, chemotherapy sensitivity of DLBCL, and expression of pathway molecules is associated with the prognosis of DLBCL. Some agents targeting these pathways have been proved effective and relevant clinical trials are in progress. These agents used single or combined with chemotherapy/each other might raise the possibility of improving clinical outcomes in DLBCL. CONCLUSION This review presents several signal pathways of DLBCL and targeted agents had a tendency to improve the curative effect, especially in high-risk or relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Feifei Sun
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No.324, Jingwu Road, Jinan, Shandong 250021, China.,Shandong University School of Medicine, Jinan, Shandong 250012, China
| |
Collapse
|
49
|
Tarantelli C, Lupia A, Stathis A, Bertoni F. Is There a Role for Dual PI3K/mTOR Inhibitors for Patients Affected with Lymphoma? Int J Mol Sci 2020; 21:E1060. [PMID: 32033478 PMCID: PMC7037719 DOI: 10.3390/ijms21031060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
The activation of the PI3K/AKT/mTOR pathway is a main driver of cell growth, proliferation, survival, and chemoresistance of cancer cells, and, for this reason, represents an attractive target for developing targeted anti-cancer drugs. There are plenty of preclinical data sustaining the anti-tumor activity of dual PI3K/mTOR inhibitors as single agents and in combination in lymphomas. Clinical responses, including complete remissions (especially in follicular lymphoma patients), are also observed in the very few clinical studies performed in patients that are affected by relapsed/refractory lymphomas or chronic lymphocytic leukemia. In this review, we summarize the literature on dual PI3K/mTOR inhibitors focusing on the lymphoma setting, presenting both the three compounds still in clinical development and those with a clinical program stopped or put on hold.
Collapse
Affiliation(s)
- Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland;
| | - Antonio Lupia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, USI, 6900 Lugano, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland;
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
| |
Collapse
|
50
|
Targeting chronic NFAT activation with calcineurin inhibitors in diffuse large B-cell lymphoma. Blood 2020; 135:121-132. [DOI: 10.1182/blood.2019001866] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/07/2019] [Indexed: 12/31/2022] Open
Abstract
Abstract
Diffuse large B-cell lymphoma (DLBCL) represents the most common adult lymphoma and can be divided into 2 major molecular subtypes: the germinal center B-cell-like and the aggressive activated B-cell-like (ABC) DLBCL. Previous studies suggested that chronic B-cell receptor signaling and increased NF-κB activation contribute to ABC DLBCL survival. Here we show that the activity of the transcription factor NFAT is chronically elevated in both DLBCL subtypes. Surprisingly, NFAT activation is independent of B-cell receptor signaling, but mediated by an increased calcium flux and calcineurin-mediated dephosphorylation of NFAT. Intriguingly, although NFAT is activated in both DLBCL subtypes, long-term calcineurin inhibition with cyclosporin A or FK506, both clinically approved drugs, triggers potent cytotoxicity specifically in ABC DLBCL cells. The antitumor effects of calcineurin inhibitors are associated with the reduced expression of c-Jun, interleukin-6, and interleukin-10, which were identified as NFAT target genes that are particularly important for the survival of ABC DLBCL. Furthermore, calcineurin blockade synergized with BCL-2 and MCL-1 inhibitors in killing ABC DLBCL cells. Collectively, these findings identify constitutive NFAT signaling as a crucial functional driver of ABC DLBCL and highlight calcineurin inhibition as a novel strategy for the treatment of this aggressive lymphoma subtype.
Collapse
|