1
|
Schmitz E, Ridout A, Smith AL, Eiken AP, Skupa SA, Drengler EM, Singh S, Rana S, Natarajan A, El-Gamal D. Immunogenic Cell Death Traits Emitted from Chronic Lymphocytic Leukemia Cells Following Treatment with a Novel Anti-Cancer Agent, SpiD3. Biomedicines 2024; 12:2857. [PMID: 39767763 PMCID: PMC11673838 DOI: 10.3390/biomedicines12122857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Targeted therapies (e.g., ibrutinib) have markedly improved chronic lymphocytic leukemia (CLL) management; however, ~20% of patients experience disease relapse, suggesting the inadequate depth and durability of these front-line strategies. Moreover, immunotherapeutic success in CLL has been stifled by its pro-tumor microenvironment milieu and low mutational burden, cultivating poor antigenicity and limited ability to generate anti-tumor immunity through adaptive immune cell engagement. Previously, we have demonstrated how a three-carbon-linker spirocyclic dimer (SpiD3) promotes futile activation of the unfolded protein response (UPR) in CLL cells through immense misfolded-protein mimicry, culminating in insurmountable ER stress and programmed CLL cell death. Method: Herein, we used flow cytometry and cell-based assays to capture the kinetics and magnitude of SpiD3-induced damage-associated molecular patterns (DAMPs) in CLL cell lines and primary samples. Result: SpiD3 treatment, in vitro and in vivo, demonstrated the capacity to propagate immunogenic cell death through emissions of classically immunogenic DAMPs (CALR, ATP, HMGB1) and establish a chemotactic gradient for bone marrow-derived dendritic cells. Conclusions: Thus, this study supports future investigation into the relationship between novel therapeutics, manners of cancer cell death, and their contributions to adaptive immune cell engagement as a means for improving anti-cancer therapy in CLL.
Collapse
Affiliation(s)
- Elizabeth Schmitz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Abigail Ridout
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Audrey L. Smith
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Alexandria P. Eiken
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Sydney A. Skupa
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Erin M. Drengler
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Sarbjit Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.S.); (A.L.S.); (A.P.E.); (S.A.S.); (E.M.D.); (S.S.); (S.R.); (A.N.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Camerini E, Amsen D, Kater AP, Peters FS. The complexities of T-cell dysfunction in chronic lymphocytic leukemia. Semin Hematol 2024; 61:163-171. [PMID: 38782635 DOI: 10.1053/j.seminhematol.2024.04.001] [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: 11/14/2023] [Revised: 03/13/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by profound alterations and defects in the T-cell compartment. This observation has gained renewed interest as T-cell treatment strategies, which are successfully applied in more aggressive B-cell malignancies, have yielded disappointing results in CLL. Despite ongoing efforts to understand and address the observed T-cell defects, the exact mechanisms and nature underlying this dysfunction remain largely unknown. In this review, we examine the supporting signals from T cells to CLL cells in the lymph node niche, summarize key findings on T-cell functional defects, delve into potential underlying causes, and explore novel strategies for reversing these deficiencies. Our goal is to identify strategies aimed at resolving CLL-induced T-cell dysfunction which, in the future, will enhance the efficacy of autologous T-cell-based therapies for CLL patients.
Collapse
Affiliation(s)
- Elena Camerini
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Derk Amsen
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Landsteiner Laboratory for Blood Cell Research at Sanquin, Amsterdam, The Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Fleur S Peters
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Vom Stein AF, Hallek M, Nguyen PH. Role of the tumor microenvironment in CLL pathogenesis. Semin Hematol 2024; 61:142-154. [PMID: 38220499 DOI: 10.1053/j.seminhematol.2023.12.004] [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: 11/07/2023] [Revised: 12/02/2023] [Accepted: 12/23/2023] [Indexed: 01/16/2024]
Abstract
Chronic lymphocytic leukemia (CLL) cells extensively interact with and depend on their surrounding tumor microenvironment (TME). The TME encompasses a heterogeneous array of cell types, soluble signals, and extracellular vesicles, which contribute significantly to CLL pathogenesis. CLL cells and the TME cooperatively generate a chronic inflammatory milieu, which reciprocally reprograms the TME and activates a signaling network within CLL cells, promoting their survival and proliferation. Additionally, the inflammatory milieu exerts chemotactic effects, attracting CLL cells and other immune cells to the lymphoid tissues. The intricate CLL-TME interactions also facilitate immune evasion and compromise leukemic cell surveillance. We also review recent advances that have shed light on additional aspects that are substantially influenced by the CLL-TME interplay.
Collapse
Affiliation(s)
- Alexander F Vom Stein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf; Center for Molecular Medicine Cologne; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - Michael Hallek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf; Center for Molecular Medicine Cologne; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - Phuong-Hien Nguyen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf; Center for Molecular Medicine Cologne; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany.
| |
Collapse
|
4
|
Smith AL, Skupa SA, Eiken AP, Reznicek TE, Schmitz E, Williams N, Moore DY, D’Angelo CR, Kallam A, Lunning MA, Bociek RG, Vose JM, Mohamed E, Mahr AR, Denton PW, Powell B, Bollag G, Rowley MJ, El-Gamal D. BET inhibition reforms the immune microenvironment and alleviates T cell dysfunction in chronic lymphocytic leukemia. JCI Insight 2024; 9:e177054. [PMID: 38775157 PMCID: PMC11141939 DOI: 10.1172/jci.insight.177054] [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: 11/01/2023] [Accepted: 04/12/2024] [Indexed: 06/02/2024] Open
Abstract
Redundant tumor microenvironment (TME) immunosuppressive mechanisms and epigenetic maintenance of terminal T cell exhaustion greatly hinder functional antitumor immune responses in chronic lymphocytic leukemia (CLL). Bromodomain and extraterminal (BET) proteins regulate key pathways contributing to CLL pathogenesis and TME interactions, including T cell function and differentiation. Herein, we report that blocking BET protein function alleviates immunosuppressive networks in the CLL TME and repairs inherent CLL T cell defects. The pan-BET inhibitor OPN-51107 reduced exhaustion-associated cell signatures resulting in improved T cell proliferation and effector function in the Eμ-TCL1 splenic TME. Following BET inhibition (BET-i), TME T cells coexpressed significantly fewer inhibitory receptors (IRs) (e.g., PD-1, CD160, CD244, LAG3, VISTA). Complementary results were witnessed in primary CLL cultures, wherein OPN-51107 exerted proinflammatory effects on T cells, regardless of leukemic cell burden. BET-i additionally promotes a progenitor T cell phenotype through reduced expression of transcription factors that maintain terminal differentiation and increased expression of TCF-1, at least in part through altered chromatin accessibility. Moreover, direct T cell effects of BET-i were unmatched by common targeted therapies in CLL. This study demonstrates the immunomodulatory action of BET-i on CLL T cells and supports the inclusion of BET inhibitors in the management of CLL to alleviate terminal T cell dysfunction and potentially enhance tumoricidal T cell activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Nolan Williams
- Eppley Institute for Research in Cancer and Allied Diseases
| | - Dalia Y. Moore
- Eppley Institute for Research in Cancer and Allied Diseases
| | - Christopher R. D’Angelo
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Avyakta Kallam
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Matthew A. Lunning
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - R. Gregory Bociek
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Julie M. Vose
- Division of Hematology and Oncology, Department of Internal Medicine, and
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Eslam Mohamed
- College of Medicine and College of Graduate Studies, California Northstate University, Elk Grove, California, USA
| | - Anna R. Mahr
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Paul W. Denton
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Ben Powell
- Plexxikon Inc., South San Francisco, California, USA
| | | | | | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases
- Fred & Pamela Buffett Cancer Center (FPBCC), University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| |
Collapse
|
5
|
Giannoni P, Marini C, Cutrona G, Sambuceti GM, Fais F, de Totero D. Unraveling the Bone Tissue Microenvironment in Chronic Lymphocytic Leukemia. Cancers (Basel) 2023; 15:5058. [PMID: 37894425 PMCID: PMC10605026 DOI: 10.3390/cancers15205058] [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: 09/14/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most frequent leukemia in Western countries. Although characterized by the progressive expansion and accumulation of leukemic B cells in peripheral blood, CLL cells develop in protective niches mainly located within lymph nodes and bone marrow. Multiple interactions between CLL and microenvironmental cells may favor the expansion of a B cell clone, further driving immune cells toward an immunosuppressive phenotype. Here, we summarize the current understanding of bone tissue alterations in CLL patients, further addressing and suggesting how the multiple interactions between CLL cells and osteoblasts/osteoclasts can be involved in these processes. Recent findings proposing the disruption of the endosteal niche by the expansion of a leukemic B cell clone appear to be a novel field of research to be deeply investigated and potentially relevant to provide new therapeutic approaches.
Collapse
Affiliation(s)
- Paolo Giannoni
- Department of Experimental Medicine, Biology Section, University of Genova, 16132 Genova, Italy;
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (G.M.S.)
- CNR Institute of Bioimages and Molecular Physiology, 20054 Milano, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (G.C.); (F.F.)
| | - Gian Mario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (C.M.); (G.M.S.)
- Department of Health Sciences, University of Genova, 16132 Genova, Italy
| | - Franco Fais
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (G.C.); (F.F.)
- Department of Experimental Medicine, Anatomy Section, University of Genova, 16132 Genova, Italy
| | - Daniela de Totero
- Department of Health Sciences, University of Genova, 16132 Genova, Italy
| |
Collapse
|
6
|
Ding Z, Shi R, Hu W, Tian L, Sun R, Wu Y, Zhang X. Cancer-associated fibroblasts in hematologic malignancies: elucidating roles and spotlighting therapeutic targets. Front Oncol 2023; 13:1193978. [PMID: 37746306 PMCID: PMC10511871 DOI: 10.3389/fonc.2023.1193978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Hematologic malignancies comprise a diverse range of blood, bone marrow, and organ-related disorders that present significant challenges due to drug resistance, relapse, and treatment failure. Cancer-associated fibroblasts (CAFs) represent a critical component of the tumor microenvironment (TME) and have recently emerged as potential therapeutic targets. In this comprehensive review, we summarize the latest findings on the roles of CAFs in various hematologic malignancies, including acute leukemia, multiple myeloma, chronic lymphocytic leukemia, myeloproliferative neoplasms, and lymphoma. We also explore their involvement in tumor progression, drug resistance, and the various signaling pathways implicated in their activation and function. While the underlying mechanisms and the existence of multiple CAF subtypes pose challenges, targeting CAFs and their associated pathways offers a promising avenue for the development of innovative treatments to improve patient outcomes in hematologic malignancies.
Collapse
Affiliation(s)
- Ziyang Ding
- The Second Clinical School of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weikang Hu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Tian
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Sun
- Department of Radiation Oncology, Jinling Hospital, Nanjing, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Zhang
- Department of Hematology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Haselager MV, van Driel BF, Perelaer E, de Rooij D, Lashgari D, Loos R, Kater AP, Moerland PD, Eldering E. In Vitro 3D Spheroid Culture System Displays Sustained T Cell-dependent CLL Proliferation and Survival. Hemasphere 2023; 7:e938. [PMID: 37637994 PMCID: PMC10448932 DOI: 10.1097/hs9.0000000000000938] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/26/2023] [Indexed: 08/29/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells are highly dependent on microenvironmental cells and signals. The lymph node (LN) is the critical site of in vivo CLL proliferation and development of resistance to both chemotherapy and targeted agents. We present a new model that incorporates key aspects of the CLL LN, which enables investigation of CLL cells in the context of a protective niche. We describe a three-dimensional (3D) in vitro culture system using ultra-low attachment plates to create spheroids of CLL cells derived from peripheral blood. Starting from CLL:T cell ratios as observed in LN samples, CLL activation was induced by either direct stimulation and/or indirectly via T cells. Compared with two-dimensional cultures, 3D cultures promoted CLL proliferation in a T cell-dependent manner, and enabled expansion for up to 7 weeks, including the formation of follicle-like structures after several weeks of culture. This model enables high-throughput drug screening, of which we describe response to Btk inhibition, venetoclax resistance, and T cell-mediated cytotoxicity as examples. In summary, we present the first LN-mimicking in vitro 3D culture for primary CLL, which enables readouts such as real-time drug screens, kinetic growth assays, and spatial localization. This is the first in vitro CLL system that allows testing of response and resistance to venetoclax and Bruton's tyrosine kinase inhibitors in the context of the tumor microenvironment, thereby opening up new possibilities for clinically useful applications.
Collapse
Affiliation(s)
- Marco V. Haselager
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Bianca F. van Driel
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
| | - Eduard Perelaer
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
| | - Dennis de Rooij
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
| | - Danial Lashgari
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
| | - Remco Loos
- Center for Innovation and Translational Research Europe, Bristol Myers Squibb, Sevilla, Spain
| | - Arnon P. Kater
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
| | - Perry D. Moerland
- Department of Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology Amsterdam, The Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Dubois K, Tannoury M, Bauvois B, Susin SA, Garnier D. Extracellular Vesicles in Chronic Lymphocytic Leukemia: Tumor Microenvironment Messengers as a Basis for New Targeted Therapies? Cancers (Basel) 2023; 15:cancers15082307. [PMID: 37190234 DOI: 10.3390/cancers15082307] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
In addition to intrinsic genomic and nongenomic alterations, tumor progression is also dependent on the tumor microenvironment (TME, mainly composed of the extracellular matrix (ECM), secreted factors, and bystander immune and stromal cells). In chronic lymphocytic leukemia (CLL), B cells have a defect in cell death; contact with the TME in secondary lymphoid organs dramatically increases the B cells' survival via the activation of various molecular pathways, including the B cell receptor and CD40 signaling. Conversely, CLL cells increase the permissiveness of the TME by inducing changes in the ECM, secreted factors, and bystander cells. Recently, the extracellular vesicles (EVs) released into the TME have emerged as key arbiters of cross-talk with tumor cells. The EVs' cargo can contain various bioactive substances (including metabolites, proteins, RNA, and DNA); upon delivery to target cells, these substances can induce intracellular signaling and drive tumor progression. Here, we review recent research on the biology of EVs in CLL. EVs have diagnostic/prognostic significance and clearly influence the clinical outcome of CLL; hence, from the perspective of blocking CLL-TME interactions, EVs are therapeutic targets. The identification of novel EV inhibitors might pave the way to the development of novel combination treatments for CLL and the optimization of currently available treatments (including immunotherapy).
Collapse
Affiliation(s)
- Kenza Dubois
- Sorbonne Université, Université Paris Cité, Inserm, Centre de Recherche des Cordeliers, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| | - Mariana Tannoury
- Sorbonne Université, Université Paris Cité, Inserm, Centre de Recherche des Cordeliers, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| | - Brigitte Bauvois
- Sorbonne Université, Université Paris Cité, Inserm, Centre de Recherche des Cordeliers, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| | - Santos A Susin
- Sorbonne Université, Université Paris Cité, Inserm, Centre de Recherche des Cordeliers, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| | - Delphine Garnier
- Sorbonne Université, Université Paris Cité, Inserm, Centre de Recherche des Cordeliers, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| |
Collapse
|
9
|
Affiliation(s)
- Eric Eldering
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam,Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Cancer Center Amsterdam (CCA) and Amsterdam Infection and Immunity Institute (AIII), Amsterdam, the Netherlands,
| |
Collapse
|
10
|
Avsec D, Škrlj Miklavčič M, Burnik T, Kandušer M, Bizjak M, Podgornik H, Mlinarič-Raščan I. Inhibition of p38 MAPK or immunoproteasome overcomes resistance of chronic lymphocytic leukemia cells to Bcl-2 antagonist venetoclax. Cell Death Dis 2022; 13:860. [PMID: 36209148 PMCID: PMC9547871 DOI: 10.1038/s41419-022-05287-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 01/23/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a hematological neoplasm of CD19-positive mature-appearing B lymphocytes. Despite the clinical success of targeted therapies in CLL, the development of resistance diminishes their therapeutic activity. This is also true for the Bcl-2 antagonist venetoclax. We investigated the molecular mechanisms that drive venetoclax resistance in CLL, with a clear focus to provide new strategies to successfully combat it. Activation of CLL cells with IFNγ, PMA/ionomycin, and sCD40L diminished the cytotoxicity of venetoclax. We demonstrated that the metabolic activity of cells treated with 1 nM venetoclax alone was 48% of untreated cells, and was higher for cells co-treated with IFNγ (110%), PMA/ionomycin (78%), and sCD40L (62%). As of molecular mechanism, we showed that PMA/ionomycin and sCD40L triggered translocation of NFκB in primary CLL cells, while IFNγ activated p38 MAPK, suppressed spontaneous and venetoclax-induced apoptosis and induced formation of the immunoproteasome. Inhibition of immunoproteasome with ONX-0914 suppressed activity of immunoproteasome and synergized with venetoclax against primary CLL cells. On the other hand, inhibition of p38 MAPK abolished cytoprotective effects of IFNγ. We demonstrated that venetoclax-resistant (MEC-1 VER) cells overexpressed p38 MAPK and p-Bcl-2 (Ser70), and underexpressed Mcl-1, Bax, and Bak. Inhibition of p38 MAPK or immunoproteasome triggered apoptosis in CLL cells and overcame the resistance to venetoclax of MEC-1 VER cells and venetoclax-insensitive primary CLL cells. In conclusion, the p38 MAPK pathway and immunoproteasome represent novel targets to combat venetoclax resistance in CLL.
Collapse
Affiliation(s)
- Damjan Avsec
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Marja Škrlj Miklavčič
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Tilen Burnik
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Maša Kandušer
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Maruša Bizjak
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| | - Helena Podgornik
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia ,grid.29524.380000 0004 0571 7705University Medical Centre Ljubljana, Department of Haematology, SI-1000 Ljubljana, Slovenia
| | - Irena Mlinarič-Raščan
- grid.8954.00000 0001 0721 6013University of Ljubljana, Faculty of Pharmacy, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
Liang T, Wang X, Liu Y, Ai H, Wang Q, Wang X, Wei X, Song Y, Yin Q. Decreased TCF1 and BCL11B expression predicts poor prognosis for patients with chronic lymphocytic leukemia. Front Immunol 2022; 13:985280. [PMID: 36211334 PMCID: PMC9539190 DOI: 10.3389/fimmu.2022.985280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
T cell immune dysfunction is a prominent characteristic of chronic lymphocytic leukemia (CLL) and the main cause of failure for immunotherapy and multi-drug resistance. There remains a lack of specific biomarkers for evaluating T cell immune status with outcome for CLL patients. T cell factor 1 (TCF1, encoded by the TCF7 gene) can be used as a critical determinant of successful anti-tumor immunotherapy and a prognostic indicator in some solid tumors; however, the effects of TCF1 in CLL remain unclear. Here, we first analyzed the biological processes and functions of TCF1 and co-expressing genes using the GEO and STRING databases with the online tools Venny, Circos, and Database for Annotation, Visualization, and Integrated Discovery (DAVID). Then the expression and prognostic values of TCF1 and its partner gene B cell leukemia/lymphoma 11B (BCL11B) were explored for 505 CLL patients from 6 datasets and validated with 50 CLL patients from Henan cancer hospital (HNCH). TCF1 was downregulated in CLL patients, particularly in CD8+ T cells, which was significantly correlated with poor time-to-first treatment (TTFT) and overall survival (OS) as well as short restricted mean survival time (RMST). Function and pathway enrichment analysis revealed that TCF1 was positively correlated with BCL11B, which is involved in regulating the activation and differentiation of T cells in CLL patients. Intriguingly, BCL11B was highly consistent with TCF1 in its decreased expression and prediction of poor prognosis. More importantly, the combination of TCF1 and BCL11B could more accurately assess prognosis than either alone. Additionally, decreased TCF1 and BCL11B expression serves as an independent risk factor for rapid disease progression, coinciding with high-risk indicators, including unmutated IGHV, TP53 alteration, and advanced disease. Altogether, this study demonstrates that decreased TCF1 and BCL11B expression is significantly correlated with poor prognosis, which may be due to decreased TCF1+CD8+ T cells, impairing the effector CD8+ T cell differentiation regulated by TCF1/BCL11B.
Collapse
|
12
|
Hou P, Dai W, Jin Y, Zhao F, Liu J, Liu H. Maternal exposure to di-2-ethylhexyl phthalate (DEHP) depresses lactation capacity in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155813. [PMID: 35550907 DOI: 10.1016/j.scitotenv.2022.155813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/20/2022] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
Increasing evidence shows that di-2-ethylhexyl phthalate (DEHP), mostly commonly used phthalate for the production of flexible polyvinyl chloride (PVC), has the potential to induce serious health risks in humans. However, the understanding of DEHP-induced lactation performance remains largely unknown. We sought to investigate the adverse effects of DEHP on lactation and examine the underlying mechanism linking DEHP exposure with the lactation alterations. We successfully adapted a maternal DEHP exposure model in female pregnant/lactating mice. Then we determined effects of DEHP exposure on food intake, body weight and milk production as well as the alterations in endocrine factors in lactating mice. The integrated metabonomic and transcriptomic analyses of the mammary gland were performed to measure the changed metabolites and genes related to DEHP exposure-induced lactation alterations. We observed the reduced food intake with elevated blood leptin and the decreased milk yield as well as the reduced levels of serum prolactin, growth hormone, insulin-like growth factor 1 and insulin after exposed to DEHP. Furthermore, 208 metabolites and 3452 genes were separately identified as differentially expressed features associated with DEHP exposure. Integrated metabonomic and transcriptomic analyses demonstrated that DEHP caused lactation depression mainly through impairing energy generation, inducing stress responses along with the hypoactivation of inflammation, reducing the production of antioxidants, disrupting hormone homeostasis and repressing the synthesis of milk constituents (the lower glucose availability for lactose synthesis; the disruption of milk fat globule membrane for lipid droplet formation; the ribosomal dysfunction and disruption of post-modifications for milk protein synthesis). We demonstrated that DEHP disrupted several lactation-related hormone homeostasis and multiple processes like energy insufficiency, inflammation activation, oxidative stress aggravation and disturbance of milk production in the mammary gland of female lactating mice. Our results provide valuable information for the health risk of plastic additive (DEHP) on female lactation dysfunction.
Collapse
Affiliation(s)
- Pengfei Hou
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenting Dai
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanshan Jin
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fengqi Zhao
- Laboratory of Lactation and Metabolic Physiology, Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, USA
| | - Jianxin Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Liu Y, Song Y, Yin Q. Effects of ibrutinib on T-cell immunity in patients with chronic lymphocytic leukemia. Front Immunol 2022; 13:962552. [PMID: 36059445 PMCID: PMC9437578 DOI: 10.3389/fimmu.2022.962552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL), a highly heterogeneous B-cell malignancy, is characterized by tumor microenvironment disorder and T-cell immune dysfunction, which play a major role in the proliferation and survival of CLL cells. Ibrutinib is the first irreversible inhibitor of Bruton’s tyrosine kinase (BTK). In addition to targeting B-cell receptor (BCR) signaling to kill tumor cells, increasing evidence has suggested that ibrutinib regulates the tumor microenvironment and T-cell immunity in a direct and indirect manner. For example, ibrutinib not only reverses the tumor microenvironment by blocking cytokine networks and toll-like receptor signaling but also regulates T cells in number, subset distribution, T-cell receptor (TCR) repertoire and immune function by inhibiting interleukin-2 inducible T-cell kinase (ITK) and reducing the expression of inhibitory receptors, and so on. In this review, we summarize the current evidence for the effects of ibrutinib on the tumor microenvironment and cellular immunity of patients with CLL, particularly for the behavior and function of T cells, explore its potential mechanisms, and provide a basis for the clinical benefits of long-term ibrutinib treatment and combined therapy based on T-cell-based immunotherapies.
Collapse
|
14
|
Targeting metabolic reprogramming in chronic lymphocytic leukemia. Exp Hematol Oncol 2022; 11:39. [PMID: 35761419 PMCID: PMC9235173 DOI: 10.1186/s40164-022-00292-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/05/2022] [Indexed: 11/28/2022] Open
Abstract
Metabolic reprogramming, fundamentally pivotal in carcinogenesis and progression of cancer, is considered as a promising therapeutic target against tumors. In chronic lymphocytic leukemia (CLL) cells, metabolic abnormalities mediate alternations in proliferation and survival compared with normal B cells. However, the role of metabolic reprogramming is still under investigation in CLL. In this review, the critical metabolic processes of CLL were summarized, particularly glycolysis, lipid metabolism and oxidative phosphorylation. The effects of T cells and stromal cells in the microenvironment on metabolism of CLL were also elucidated. Besides, the metabolic alternation is regulated by some oncogenes and tumor suppressor regulators, especially TP53, MYC and ATM. Thus, the agents targeting metabolic enzymes or signal pathways may impede the progression of CLL. Both the inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) statins and the lipoprotein lipase inhibitor orlistat induce the apoptosis of CLL cells. In addition, a series of oxidative phosphorylation inhibitors play important roles in decreasing the proliferation of CLL cells. We epitomized recent advancements in metabolic reprogramming in CLL and discussed their clinical potentiality for innovative therapy options. Metabolic reprogramming plays a vital role in the initiation and progression of CLL. Therapeutic approaches targeting metabolism have their advantages in improving the survival of CLL patients. This review may shed novel light on the metabolism of CLL, leading to the development of targeted agents based on the reshaping metabolism of CLL cells.
Collapse
|
15
|
In Vitro and In Vivo Models of CLL–T Cell Interactions: Implications for Drug Testing. Cancers (Basel) 2022; 14:cancers14133087. [PMID: 35804862 PMCID: PMC9264798 DOI: 10.3390/cancers14133087] [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: 06/05/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Chronic lymphocytic leukemia (CLL) cells in the peripheral blood and lymphoid microenvironment display substantially different gene expression profiles and proliferative capaci-ty. It has been suggested that CLL–T-cell interactions are key pro-proliferative stimuli in immune niches. We review in vitro and in vivo model systems that mimic CLL-T-cell interactions to trigger CLL proliferation and study therapy resistance. We focus on studies describing the co-culture of leukemic cells with T cells, or supportive cell lines expressing T-cell factors, and simplified models of CLL cells’ stimulation with recombinant factors. In the second part, we summarize mouse models revealing the role of T cells in CLL biology and implications for generating patient-derived xenografts by co-transplanting leukemic cells with T cells. Abstract T cells are key components in environments that support chronic lymphocytic leukemia (CLL), activating CLL-cell proliferation and survival. Here, we review in vitro and in vivo model systems that mimic CLL–T-cell interactions, since these are critical for CLL-cell division and resistance to some types of therapy (such as DNA-damaging drugs or BH3-mimetic venetoclax). We discuss approaches for direct CLL-cell co-culture with autologous T cells, models utilizing supportive cell lines engineered to express T-cell factors (such as CD40L) or stimulating CLL cells with combinations of recombinant factors (CD40L, interleukins IL4 or IL21, INFγ) and additional B-cell receptor (BCR) activation with anti-IgM antibody. We also summarize strategies for CLL co-transplantation with autologous T cells into immunodeficient mice (NOD/SCID, NSG, NOG) to generate patient-derived xenografts (PDX) and the role of T cells in transgenic CLL mouse models based on TCL1 overexpression (Eµ-TCL1). We further discuss how these in vitro and in vivo models could be used to test drugs to uncover the effects of targeted therapies (such as inhibitors of BTK, PI3K, SYK, AKT, MEK, CDKs, BCL2, and proteasome) or chemotherapy (fludarabine and bendamustine) on CLL–T-cell interactions and CLL proliferation.
Collapse
|
16
|
Nurse-like cells sequester B cells in chronic lymphocytic leukemia disorganized lymph nodes via an alternative production of CCL21. Blood Adv 2022; 6:4691-4704. [PMID: 35679464 PMCID: PMC9631672 DOI: 10.1182/bloodadvances.2021006169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 05/31/2022] [Indexed: 12/02/2022] Open
Abstract
Stromal cell architecture is deeply altered in CLL lymph nodes. CCL21, produced by leukemia-induced macrophages, improves retention and niching of malignant CCR7+ B cells in CLL lymph nodes.
Tumor microenvironment exerts a critical role in sustaining homing, retention, and survival of chronic lymphocytic leukemia (CLL) cells in secondary lymphoid organs. Such conditions foster immune surveillance escape and resistance to therapies. The physiological microenvironment is rendered tumor permissive by an interplay of chemokines, chemokine receptors, and adhesion molecules as well as by direct interactions between malignant lymphocytes and stromal cells, T cells, and specialized macrophages referred to as nurselike cells (NLCs). To characterize this complex interplay, we investigated the altered architecture on CLL lymph nodes biopsies and observed a dramatic loss of tissue subcompartments and stromal cell networks as compared with nonmalignant lymph nodes. A supplemental high density of CD68+ cells expressing the homeostatic chemokine CCL21 was randomly distributed. Using an imaging flow cytometry approach, CCL21 mRNA and the corresponding protein were observed in single CD68+ NLCs differentiated in vitro from CLL peripheral blood mononuclear cells. The chemokine was sequestered at the NLC membrane, helping capture of CCR7-high-expressing CLL B cells. Inhibiting the CCL21/CCR7 interaction by blocking antibodies or using therapeutic ibrutinib altered the adhesion of leukemic cells. Our results indicate NLCs as providers of an alternative source of CCL21, taking over the physiological task of follicular reticular cells, whose network is deeply altered in CLL lymph nodes. By retaining malignant B cells, CCL21 provides a protective environment for their niching and survival, thus allowing tumor evasion and resistance to treatment. These findings argue for a specific targeting or reeducation of NLCs as a new immunotherapy strategy for this disease.
Collapse
|
17
|
Mimmi S, Maisano D, Dattilo V, Gentile M, Chiurazzi F, D’Ambrosio A, Zimbo A, Nisticò N, Aloisio A, Vecchio E, Fiume G, Iaccino E, Quinto I. Unmutated IGHV1-69 CLL Clone Displays a Distinct Gene Expression Profile by a Comparative qRT-PCR Assay. Biomedicines 2022; 10:biomedicines10030604. [PMID: 35327406 PMCID: PMC8945665 DOI: 10.3390/biomedicines10030604] [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: 01/11/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 11/29/2022] Open
Abstract
Chronic Lymphocytic Leukemia (CLL) is a heterogeneous disease characterized by variable clinical courses among different patients. This notion was supported by the possible coexistence of two or more independent CLL clones within the same patients, identified by the characterization of the B cell receptor immunoglobulin (BcR IG) idiotypic sequence. By using the antigen-binding site of the BcR IG as bait, the identification and isolation of aggressive and drug-resistance leukemic B-cell clones could allow a deeper biological and molecular investigation. Indeed, by the screening of phage display libraries, we previously selected a peptide binder of the idiotypic region of CLL BCR IGs expressing the unmutated rearrangement IGHV1-69 and used it as a probe to perform a peptide-based cell sorting by flow cytometry in peripheral blood samples from patients with CLL. Since the IGHV1-69 clones persisted during the follow-up time in both patients, we explored the possibility of these clones having acquired an evolutive advantage compared to the other coexisting clones in terms of a higher expression of genes involved in the survival and apoptosis escape processes. To this end, we studied the expression patterns of a panel of genes involved in apoptosis regulation and in NF-kB-dependent pro-survival signals by comparative qRT-PCR assays. According to the results, IGHV1-69 clones showed a higher expression of pro-survival and anti-apoptotic genes as compared to the other CLL clones with different immunogenetic characteristics. Moreover, these IGHV1-69 clones did not carry any characteristic genetic lesions, indicating the relevance of our approach in performing a comprehensive molecular characterization of single tumor clones, as well as for designing new personalized therapeutic approaches for the most aggressive and persistent tumor clones.
Collapse
Affiliation(s)
- Selena Mimmi
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
- Correspondence: (S.M.); (V.D.); Tel.: +39-0961-369-4057 (S.M. & V.D.)
| | - Domenico Maisano
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Vincenzo Dattilo
- Laboratory Genetics Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Correspondence: (S.M.); (V.D.); Tel.: +39-0961-369-4057 (S.M. & V.D.)
| | - Massimo Gentile
- Hematology Unit, Department of Onco-Hematology, A.O of Cosenza, 87100 Cosenza, Italy;
| | - Federico Chiurazzi
- Hematological Clinic, Department of Clinical Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.C.); (A.D.)
| | - Alessandro D’Ambrosio
- Hematological Clinic, Department of Clinical Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.C.); (A.D.)
| | - Annamaria Zimbo
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Nancy Nisticò
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Annamaria Aloisio
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Eleonora Vecchio
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Giuseppe Fiume
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Enrico Iaccino
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| | - Ileana Quinto
- Laboratory of Immunology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.M.); (A.Z.); (N.N.); (A.A.); (E.V.); (G.F.); (E.I.); (I.Q.)
| |
Collapse
|
18
|
Overcoming resistance hurdles. Blood 2022; 139:804-805. [PMID: 35142854 DOI: 10.1182/blood.2021014430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 11/20/2022] Open
|
19
|
Gauthier M. La leucémie lymphoïde chronique. Rev Med Interne 2022; 43:356-364. [DOI: 10.1016/j.revmed.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/29/2021] [Accepted: 01/29/2022] [Indexed: 12/01/2022]
|
20
|
Rodríguez-Caballero A, Fuentes Herrero B, Oliva Ariza G, Criado I, Alcoceba M, Prieto C, Pérez Caro M, García-Montero AC, González Díaz M, Forconi F, Sarmento-Ribeiro AB, Almeida J, Orfao A. The Hydropathy Index of the HCDR3 Region of the B-Cell Receptor Identifies Two Subgroups of IGHV-Mutated Chronic Lymphocytic Leukemia Patients With Distinct Outcome. Front Oncol 2021; 11:723722. [PMID: 34765543 PMCID: PMC8577851 DOI: 10.3389/fonc.2021.723722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
The HCDR3 sequences of the B-cell receptor (BCR) undergo constraints in length, amino acid use, and charge during maturation of B-cell precursors and after antigen encounter, leading to BCR and antibodies with high affinity to specific antigens. Chronic lymphocytic leukemia consists of an expansion of B-cells with a mixed immature and "antigen-experienced" phenotype, with either a mutated (M-CLL) or unmutated (U-CLL) tumor BCR, associated with distinct patient outcomes. Here, we investigated the hydropathy index of the BCR of 138 CLL patients and its association with the IGHV mutational status and patient outcome. Overall, two clearly distinct subgroups of M-CLL patients emerged, based on a neutral (mean hydropathy index of -0.1) vs. negatively charged BCR (mean hydropathy index of -1.1) with molecular features closer to those of B-cell precursors and peripheral/mature B-cells, respectively. Despite that M-CLL with neutral HCDR3 did not show traits associated with a mature B-cell repertoire, important differences in IGHV gene usage of tumor cells and patient outcome were observed in this subgroup of patients once compared to both U-CLL and M-CLL with negatively charged HCDR3 sequences. Compared to M-CLL with negatively charged HCDR3 sequences, M-CLL with neutral HCDR3 sequences showed predominance of men, more advanced stages of the disease, and a greater frequency of genetic alterations-e.g., del(17p)-together with a higher rate of disease progression and shorter time to therapy (TTT), independently of other prognostic factors. Our data suggest that the hydropathy index of the HCDR3 sequences of CLL cells allows the identification of a subgroup of M-CLL with intermediate prognostic features between U-CLL and the more favorable subgroup of M-CLL with a negatively charged BCR.
Collapse
Affiliation(s)
- Arancha Rodríguez-Caballero
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Blanca Fuentes Herrero
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Guillermo Oliva Ariza
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Ignacio Criado
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Alcoceba
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca/Biomedical Research Institute of Salamanca (HUS/IBSAL), Salamanca, Spain
| | - Carlos Prieto
- Bioinformatics Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
| | - María Pérez Caro
- Spanish National DNA Bank Carlos III, University of Salamanca, Salamanca, Spain
| | - Andrés C. García-Montero
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Marcos González Díaz
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca/Biomedical Research Institute of Salamanca (HUS/IBSAL), Salamanca, Spain
| | - Francesco Forconi
- Haematology Oncology Group, School of Cancer Sciences, Cancer Research UK Centre and National Institute for Health Research Experimental Cancer Medicine, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| | - Ana Bela Sarmento-Ribeiro
- Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center Institute of Cancer Molecular and Cellular Biology (IBMCC), University of Salamanca-The Spanish National Research Council (USAL-CSIC), Department of Medicine and Cytometry Service, Nucleus Research Support Platform from University of Salamanca (NUCLEUS), University of Salamanca, Salamanca, Spain
- CIBERONC Program of Liquid Biopsy, Hematologic Tumors, Centro de Investigación Biomédica en Red de Cáncer CB16/12/00400 and CB16/12/00233 (CIBERONC), Madrid, Spain
- Molecular and Cellular Biology of Hematologic Tumors, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
21
|
Mantle cell lymphoma polarizes tumor-associated macrophages into M2-like macrophages, which in turn promote tumorigenesis. Blood Adv 2021; 5:2863-2878. [PMID: 34297045 DOI: 10.1182/bloodadvances.2020003871] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are recognized as a hallmark of certain solid cancers and predictors of poor prognosis; however, the functional role of TAMs in lymphoid malignancies, including B-cell lymphoma, has not been well defined. We identified infiltration of F4/80+ TAMs in a syngeneic mouse model using the recently generated murine mantle cell lymphoma (MCL) cell line FC-muMCL1. Multicolor flow cytometric analysis of syngeneic lymphoma tumors showed distinct polarization of F4/80+ TAMs into CD206+ M2 and CD80+ M1 phenotypes. Using human MCL cell lines (Mino, Granta, and JVM2), we further showed that MCL cells polarized monocyte-derived macrophages toward an M2-like phenotype, as assessed by CD163+ expression and increased interleukin-10 (IL-10) level; however, levels of the M1 markers CD80 and IL-12 remained unaffected. To show that macrophages contribute to MCL tumorigenesis, we xenografted the human MCL cell line Mino along with CD14+ monocytes and compared tumor growth between these 2 groups. Results showed that xenografted Mino along with CD14+ monocytes significantly increased the tumor growth in vivo compared with MCL cells alone (P < .001), whereas treatment with liposomal clodronate (to deplete the macrophages) reversed the effect of CD14+ monocytes on growth of MCL xenografts (P < .001). Mechanistically, IL-10 secreted by MCL-polarized M2-like macrophages was found to be responsible for increasing MCL growth by activating STAT1 signaling, whereas IL-10 neutralizing antibody or STAT1 inhibition by fludarabine or STAT1 short hairpin RNA significantly abolished MCL growth (P < .01). Collectively, our data show the existence of a tumor microenvironmental network of macrophages and MCL tumor and suggest the importance of macrophages in interventional therapeutic strategies against MCL and other lymphoid malignancies.
Collapse
|
22
|
Mavridou D, Psatha K, Aivaliotis M. Proteomics and Drug Repurposing in CLL towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13143391. [PMID: 34298607 PMCID: PMC8303629 DOI: 10.3390/cancers13143391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Despite continued efforts, the current status of knowledge in CLL molecular pathobiology, diagnosis, prognosis and treatment remains elusive and imprecise. Proteomics approaches combined with advanced bioinformatics and drug repurposing promise to shed light on the complex proteome heterogeneity of CLL patients and mitigate, improve, or even eliminate the knowledge stagnation. In relation to this concept, this review presents a brief overview of all the available proteomics and drug repurposing studies in CLL and suggests the way such studies can be exploited to find effective therapeutic options combined with drug repurposing strategies to adopt and accost a more “precision medicine” spectrum. Abstract CLL is a hematological malignancy considered as the most frequent lymphoproliferative disease in the western world. It is characterized by high molecular heterogeneity and despite the available therapeutic options, there are many patient subgroups showing the insufficient effectiveness of disease treatment. The challenge is to investigate the individual molecular characteristics and heterogeneity of these patients. Proteomics analysis is a powerful approach that monitors the constant state of flux operators of genetic information and can unravel the proteome heterogeneity and rewiring into protein pathways in CLL patients. This review essences all the available proteomics studies in CLL and suggests the way these studies can be exploited to find effective therapeutic options combined with drug repurposing approaches. Drug repurposing utilizes all the existing knowledge of the safety and efficacy of FDA-approved or investigational drugs and anticipates drug alignment to crucial CLL therapeutic targets, leading to a better disease outcome. The drug repurposing studies in CLL are also discussed in this review. The next goal involves the integration of proteomics-based drug repurposing in precision medicine, as well as the application of this procedure into clinical practice to predict the most appropriate drugs combination that could ensure therapy and the long-term survival of each CLL patient.
Collapse
Affiliation(s)
- Dimitra Mavridou
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantina Psatha
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Greece
- Correspondence: (K.P.); (M.A.)
| | - Michalis Aivaliotis
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Greece
- Correspondence: (K.P.); (M.A.)
| |
Collapse
|
23
|
Overcoming of Microenvironment Protection on Primary Chronic Lymphocytic Leukemia Cells after Treatment with BTK and MDM2 Pharmacological Inhibitors. ACTA ACUST UNITED AC 2021; 28:2439-2451. [PMID: 34287267 PMCID: PMC8293193 DOI: 10.3390/curroncol28040223] [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: 06/08/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 11/26/2022]
Abstract
In B-chronic lymphocytic leukemia (B-CLL), the interaction between leukemic cells and the microenvironment promotes tumor cell survival. The Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib is one of the first-in-class molecules for the treatment of B-CLL patients; however, the emerging mechanisms of resistance to ibrutinib call for new therapeutic strategies. The purpose of the current study was to investigate the ability of ibrutinib plus the MDM2-inhibitor nutlin-3 to counteract the tumor microenvironment protective effect. We observed that primary B-CLL cells cultivated in microenvironment mimicking conditions were protected from apoptosis by the up-regulation of c-MYC and of p53. In the same setting, combined treatments with ibrutinib plus nutlin-3 led to significantly higher levels of apoptosis compared to the single treatments, counteracting the c-MYC up-regulation. Moreover, the combination induced high p53 levels and a significant dissipation of the mitochondrial membrane potential, together with BAX cleavage in the more active p18 form and phospho-BAD down-regulation, that are key components of the mitochondrial apoptotic pathway, enhancing the apoptosis level. Our findings propose a new therapeutic strategy to overcome the tumor microenvironment protection involved in B-CLL resistance to drugs, with possible clinical implications also for other hematologic and solid tumors for which ibrutinib is considered a therapeutic option.
Collapse
|
24
|
Vlachonikola E, Stamatopoulos K, Chatzidimitriou A. T Cell Defects and Immunotherapy in Chronic Lymphocytic Leukemia. Cancers (Basel) 2021; 13:3255. [PMID: 34209724 PMCID: PMC8268526 DOI: 10.3390/cancers13133255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 12/31/2022] Open
Abstract
In the past few years, independent studies have highlighted the relevance of the tumor microenvironment (TME) in cancer, revealing a great variety of TME-related predictive markers, as well as identifying novel therapeutic targets in the TME. Cancer immunotherapy targets different components of the immune system and the TME at large in order to reinforce effector mechanisms or relieve inhibitory and suppressive signaling. Currently, it constitutes a clinically validated treatment for many cancers, including chronic lymphocytic leukemia (CLL), an incurable malignancy of mature B lymphocytes with great dependency on microenvironmental signals. Although immunotherapy represents a promising therapeutic option with encouraging results in CLL, the dysfunctional T cell compartment remains a major obstacle in such approaches. In the scope of this review, we outline the current immunotherapeutic treatment options in CLL in the light of recent immunogenetic and functional evidence of T cell impairment. We also highlight possible approaches for overcoming T cell defects and invigorating potent anti-tumor immune responses that would enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Elisavet Vlachonikola
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, 57001 Thessaloniki, Greece; (E.V.); (K.S.)
- Department of Genetics and Molecular Biology, Faculty of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Kostas Stamatopoulos
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, 57001 Thessaloniki, Greece; (E.V.); (K.S.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Anastasia Chatzidimitriou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, 57001 Thessaloniki, Greece; (E.V.); (K.S.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
25
|
The Protein Landscape of Chronic Lymphocytic Leukemia (CLL). Blood 2021; 138:2514-2525. [PMID: 34189564 DOI: 10.1182/blood.2020009741] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 06/09/2021] [Indexed: 11/20/2022] Open
Abstract
Many functional consequences of mutations on tumor phenotypes in chronic lymphocytic leukemia (CLL) are unknown. This may be in part due to a scarcity of information on the proteome of CLL. We profiled the proteome of 117 CLL patient samples with data-independent acquisition mass spectrometry (DIA-MS) and integrated the results with genomic, transcriptomic, ex vivo drug response and clinical outcome data. We found trisomy 12, IGHV mutational status, mutated SF3B1, trisomy 19, del(17)(p13), del(11)(q22.3), mutated DDX3X, and MED12 to influence protein expression (FDR < 5%). Trisomy 12 and IGHV status were the major determinants of protein expression variation in CLL as shown by principal component analysis (1055 and 542 differentially expressed proteins, FDR=5%). Gene set enrichment analyses of CLL with trisomy 12 implicated BCR/PI3K/AKT signaling as a tumor driver. These findings were supported by analyses of protein abundance buffering and protein complex formation, which identified limited protein abundance buffering and an upregulated protein complex involved in BCR, AKT, MAPK and PI3K signaling in trisomy 12 CLL. A survey of proteins associated with trisomy 12/IGHV-independent drug response linked STAT2 protein expression with response to kinase inhibitors including BTK and MEK inhibitors. STAT2 was upregulated in U-CLL, trisomy 12 CLL and required for chemokine/cytokine signaling (interferon response). This study highlights the importance of protein abundance data as a non-redundant layer of information in tumor biology, and provides a protein expression reference map for CLL.
Collapse
|
26
|
Prognostic Value of Tie2-Expressing Monocytes in Chronic Lymphocytic Leukemia Patients. Cancers (Basel) 2021; 13:cancers13112817. [PMID: 34198760 PMCID: PMC8200999 DOI: 10.3390/cancers13112817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 06/02/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Tie2-expressing monocytes (TEM) characterized by the phenotype of CD14+CD16+Tie2+ are seen as the new immunosuppressive force in tumors. However, little is known about the role of circulating TEM in chronic lymphocytic leukemia (CLL) as opposed to their role in solid tumors. In the current study, we observed an increased percentage of TEMs in CLL patients. A greater than 14.82% proportion of TEM foretells an unfavorable prognosis. This threshold has predicted a shorter time from diagnosis to therapy, and worse overall survival. Despite these results, a multivariable Cox regression model performed in 104 CLL patients did not identify TEM as an independent predictor of survival. However, TEM, as an important element of the tumor-microenvironment, can be an important complement to other prognostic indicators. Abstract Tie2-expressing monocytes (TEMs) are associated with tumor progression and metastasis. This unique subset of monocytes has been identified as a potential prognostic marker in several solid tumors. However, TEMs remain poorly characterized in hematological cancers, including chronic lymphocytic leukemia (CLL). This study analyzed, for the first time, the clinical significance of TEM population in CLL patients. Flow cytometry analysis of TEMs (defined as CD14+CD16+Tie2+ cells) was performed at the time of diagnosis on peripheral blood mononuclear cells from 104 untreated CLL patients. Our results revealed an expansion of circulating TEM in CLL patients. These monocytes express high levels of VEGF and suppressive IL-10. A high percentage of TEM was associated closely with unfavorable prognostic markers (ZAP-70, CD38, 17p and 11q deletion, and IGHV mutational status). Moreover, increased percentages of circulating TEMs were significantly higher in patients not responding to the first-line therapy as compared to responding patients, suggesting its potential predictive value. High TEM percentage was also correlated with shorter overall survival (OS) and shorter time to treatment (TTT). Importantly, based on multivariate Cox regression analysis, TEM percentage was an independent predictor for TTT. Thus, we can suggest the adverse role of TEMs in CLL.
Collapse
|
27
|
Arruga F, Serra S, Vitale N, Guerra G, Papait A, Baffour Gyau B, Tito F, Efremov D, Vaisitti T, Deaglio S. Targeting of the A2A adenosine receptor counteracts immunosuppression in vivo in a mouse model of chronic lymphocytic leukemia. Haematologica 2021; 106:1343-1353. [PMID: 32299906 PMCID: PMC8094100 DOI: 10.3324/haematol.2019.242016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Indexed: 11/09/2022] Open
Abstract
Tumor immunosuppression is a major cause for treatment failure and disease relapse, both in solid tumors and leukemia. Local hypoxia is among the conditions that cause immunosuppression, acting at least in part through the upregulation of extracellular adenosine levels, which potently suppress T cell responses and skew macrophages towards an M2 phenotype. Hence, there is intense investigation to identify drugs that target this axis. By using the TCL1 adoptive transfer CLL mouse model, we show that adenosine production and signaling are upregulated in the hypoxic lymphoid niches, where intense colonization of leukemic cells occurs. This leads to a progressive remodeling of the immune system towards tolerance, with expansion of T regulatory cells (Tregs), loss of CD8+ T cell cytotoxicity and differentiation of murine macrophages towards the patrolling (M2-like) subset. In vivo administration of SCH58261, an inhibitor the A2A adenosine receptor, re-awakens T cell responses, while limiting Tregs expansion, and re-polarizes monocytes towards the inflammatory (M1-like) phenotype. These results show for the first time the in vivo contribution of adenosine signaling to immune tolerance in CLL, and the translational implication of drugs interrupting this pathway. Although the effects of SCH58261 on leukemic cells are limited, interfering with adenosine signaling may represent an appealing strategy for combination-based therapeutic approaches.
Collapse
Affiliation(s)
- Francesca Arruga
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Sara Serra
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Nicoletta Vitale
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulia Guerra
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Andrea Papait
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Benjamin Baffour Gyau
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Francesco Tito
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Tiziana Vaisitti
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
28
|
Peters FS, Strefford JC, Eldering E, Kater AP. T-cell dysfunction in chronic lymphocytic leukemia from an epigenetic perspective. Haematologica 2021; 106:1234-1243. [PMID: 33691381 PMCID: PMC8586819 DOI: 10.3324/haematol.2020.267914] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/18/2021] [Indexed: 11/09/2022] Open
Abstract
Cellular immunotherapeutic approaches such as chimeric antigen receptor (CAR) T-cell therapy in chronic lymphocytic leukemia (CLL) thus far have not met the high expectations. Therefore it is essential to better understand the molecular mechanisms of CLLinduced T-cell dysfunction. Even though a significant number of studies are available on T-cell function and dysfunction in CLL patients, none examine dysfunction at the epigenomic level. In non-malignant T-cell research, epigenomics is widely employed to define the differentiation pathway into T-cell exhaustion. Additionally, metabolic restrictions in the tumor microenvironment that cause T-cell dysfunction are often mediated by epigenetic changes. With this review paper we argue that understanding the epigenetic (dys)regulation in T cells of CLL patients should be leveled to the knowledge we currently have of the neoplastic B cells themselves. This will permit a complete understanding of how these immune cell interactions regulate T- and B-cell function. Here we relate the cellular and phenotypic characteristics of CLL-induced T-cell dysfunction to epigenetic studies of T-cell regulation emerging from chronic viral infection and tumor models. This paper proposes a framework for future studies into the epigenetic regulation of CLL-induced Tcell dysfunction, knowledge that will help to guide improvements in the utility of autologous T-cell based therapies in CLL.
Collapse
Affiliation(s)
- Fleur S Peters
- Experimental Immunology; Departments of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Departments of Cancer Center Amsterdam, Amsterdam, the Netherlands; Departments of Amsterdam Institute of Infection and Immunity, Amsterdam, the Netherlands; Departments of Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, the Netherlands and.
| | - Jonathan C Strefford
- Departments of Academic Unit of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Eric Eldering
- Experimental Immunology; Departments of Cancer Center Amsterdam, Amsterdam, the Netherlands; Departments of Amsterdam Institute of Infection and Immunity, Amsterdam, the Netherlands; Departments of Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, the Netherlands
| | - Arnon P Kater
- Departments of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Departments of Cancer Center Amsterdam, Amsterdam, the Netherlands; Departments of Amsterdam Institute of Infection and Immunity, Amsterdam, the Netherlands; Departments of Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, the Netherlands and
| |
Collapse
|
29
|
Vlachonikola E, Stamatopoulos K, Chatzidimitriou A. T Cells in Chronic Lymphocytic Leukemia: A Two-Edged Sword. Front Immunol 2021; 11:612244. [PMID: 33552073 PMCID: PMC7857025 DOI: 10.3389/fimmu.2020.612244] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy of mature, antigen-experienced B lymphocytes. Despite great progress recently achieved in the management of CLL, the disease remains incurable, underscoring the need for further investigation into the underlying pathophysiology. Microenvironmental crosstalk has an established role in CLL pathogenesis and progression. Indeed, the malignant CLL cells are strongly dependent on interactions with other immune and non-immune cell populations that shape a highly orchestrated network, the tumor microenvironment (TME). The composition of the TME, as well as the bidirectional interactions between the malignant clone and the microenvironmental elements have been linked to disease heterogeneity. Mounting evidence implicates T cells present in the TME in the natural history of the CLL as well as in the establishment of certain CLL hallmarks e.g. tumor evasion and immune suppression. CLL is characterized by restrictions in the T cell receptor gene repertoire, T cell oligoclonal expansions, as well as shared T cell receptor clonotypes amongst patients, strongly alluding to selection by restricted antigenic elements of as yet undisclosed identity. Further, the T cells in CLL exhibit a distinctive phenotype with features of “exhaustion” likely as a result of chronic antigenic stimulation. This might be relevant to the fact that, despite increased numbers of oligoclonal T cells in the periphery, these cells are incapable of mounting effective anti-tumor immune responses, a feature perhaps also linked with the elevated numbers of T regulatory subpopulations. Alterations of T cell gene expression profile are associated with defects in both the cytoskeleton and immune synapse formation, and are generally induced by direct contact with the malignant clone. That said, these abnormalities appear to be reversible, which is why therapies targeting the T cell compartment represent a reasonable therapeutic option in CLL. Indeed, novel strategies, including CAR T cell immunotherapy, immune checkpoint blockade and immunomodulation, have come to the spotlight in an attempt to restore the functionality of T cells and enhance targeted cytotoxic activity against the malignant clone.
Collapse
Affiliation(s)
- Elisavet Vlachonikola
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece.,Department of Genetics and Molecular Biology, Faculty of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kostas Stamatopoulos
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Chatzidimitriou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Deciphering the complex circulating immune cell microenvironment in chronic lymphocytic leukaemia using patient similarity networks. Sci Rep 2021; 11:322. [PMID: 33431934 PMCID: PMC7801466 DOI: 10.1038/s41598-020-79121-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/26/2020] [Indexed: 11/08/2022] Open
Abstract
The tissue microenvironment in chronic lymphocytic leukaemia (CLL) plays a key role in the pathogenesis of CLL, but the complex blood microenvironment in CLL has not yet been fully characterised. Therefore, immunophenotyping of circulating immune cells in 244 CLL patients and 52 healthy controls was performed using flow cytometry and analysed by multivariate Patient Similarity Networks (PSNs). Our study revealed high inter-individual heterogeneity in the distribution and activation of bystander immune cells in CLL, depending on the bulk of the CLL cells. High CLL counts were associated with low activation on circulating monocytes and T cells and vice versa. The highest activation of immune cells, particularly of intermediate and non-classical monocytes, was evident in patients treated with novel agents. PSNs revealed a low activation of immune cells in CLL progression, irrespective of IgHV status, Binet stage and TP53 disruption. Patients with high intermediate monocytes (> 5.4%) with low activation were 2.5 times more likely (95% confidence interval 1.421–4.403, P = 0.002) to had shorter time-to-treatment than those with low monocyte counts. Our study demonstrated the association between the activation of circulating immune cells and the bulk of CLL cells. The highest activation of bystander immune cells was detected in patients with slow disease course and in those treated with novel agents. The subset of intermediate monocytes showed predictive value for time-to-treatment in CLL.
Collapse
|
31
|
Chronic lymphocytic leukemia B-cell-derived TNFα impairs bone marrow myelopoiesis. iScience 2020; 24:101994. [PMID: 33458625 PMCID: PMC7797930 DOI: 10.1016/j.isci.2020.101994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/15/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022] Open
Abstract
TNFα is implicated in chronic lymphocytic leukemia (CLL) immunosuppression and disease progression. TNFα is constitutively produced by CLL B cells and is a negative regulator of bone marrow (BM) myelopoiesis. Here, we show that co-culture of CLL B cells with purified normal human hematopoietic stem and progenitor cells (HSPCs) directly altered protein levels of the myeloid and erythroid cell fate determinants PU.1 and GATA-2 at the single-cell level within transitional HSPC subsets, mimicking ex vivo expression patterns. Physical separation of CLL cells from control HSPCs or neutralizing TNFα abrogated upregulation of PU.1, yet restoration of GATA-2 required TNFα neutralization, suggesting both cell contact and soluble-factor-mediated regulation. We further show that CLL patient BM myeloid progenitors are diminished in frequency and function, an effect recapitulated by chronic exposure of control HSPCs to low-dose TNFα. These findings implicate CLL B-cell-derived TNFα in impaired BM myelopoiesis. CLL patient BM HSPCs exhibit aberrant molecular and functional characteristics CLL B-cell-derived TNFα upregulates PU.1 and GATA-2 in BM HSPCs The effects of CLL B-cell-derived TNFα are reversible upon TNFα neutralization Chronic TNFα exposure in vitro recapitulates ex vivo HSPC functional deficiencies
Collapse
|
32
|
Griggio V, Perutelli F, Salvetti C, Boccellato E, Boccadoro M, Vitale C, Coscia M. Immune Dysfunctions and Immune-Based Therapeutic Interventions in Chronic Lymphocytic Leukemia. Front Immunol 2020; 11:594556. [PMID: 33312177 PMCID: PMC7708380 DOI: 10.3389/fimmu.2020.594556] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by a wide range of tumor-induced alterations, which affect both the innate and adaptive arms of the immune response, and accumulate during disease progression. In recent years, the development of targeted therapies, such as the B-cell receptor signaling inhibitors and the Bcl-2 protein inhibitor venetoclax, has dramatically changed the treatment landscape of CLL. Despite their remarkable anti-tumor activity, targeted agents have some limitations, which include the development of drug resistance mechanisms and the inferior efficacy observed in high-risk patients. Therefore, additional treatments are necessary to obtain deeper responses and overcome drug resistance. Allogeneic hematopoietic stem cell transplantation (HSCT), which exploits immune-mediated graft-versus-leukemia effect to eradicate tumor cells, currently represents the only potentially curative therapeutic option for CLL patients. However, due to its potential toxicities, HSCT can be offered only to a restricted number of younger and fit patients. The growing understanding of the complex interplay between tumor cells and the immune system, which is responsible for immune escape mechanisms and tumor progression, has paved the way for the development of novel immune-based strategies. Despite promising preclinical observations, results from pilot clinical studies exploring the safety and efficacy of novel immune-based therapies have been sometimes suboptimal in terms of long-term tumor control. Therefore, further advances to improve their efficacy are needed. In this context, possible approaches include an earlier timing of immunotherapy within the treatment sequencing, as well as the possibility to improve the efficacy of immunotherapeutic agents by administering them in combination with other anti-tumor drugs. In this review, we will provide a comprehensive overview of main immune defects affecting patients with CLL, also describing the complex networks leading to immune evasion and tumor progression. From the therapeutic standpoint, we will go through the evolution of immune-based therapeutic approaches over time, including i) agents with broad immunomodulatory effects, such as immunomodulatory drugs, ii) currently approved and next-generation monoclonal antibodies, and iii) immunotherapeutic strategies aiming at activating or administering immune effector cells specifically targeting leukemic cells (e.g. bi-or tri-specific antibodies, tumor vaccines, chimeric antigen receptor T cells, and checkpoint inhibitors).
Collapse
Affiliation(s)
- Valentina Griggio
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Perutelli
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Chiara Salvetti
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elia Boccellato
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Mario Boccadoro
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Candida Vitale
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Marta Coscia
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
33
|
de Weerdt I, Lameris R, Scheffer GL, Vree J, de Boer R, Stam AG, van de Ven R, Levin MD, Pals ST, Roovers RC, Parren PWHI, de Gruijl TD, Kater AP, van der Vliet HJ. A Bispecific Antibody Antagonizes Prosurvival CD40 Signaling and Promotes Vγ9Vδ2 T cell-Mediated Antitumor Responses in Human B-cell Malignancies. Cancer Immunol Res 2020; 9:50-61. [PMID: 33177109 DOI: 10.1158/2326-6066.cir-20-0138] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/05/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022]
Abstract
Novel T cell-based therapies for the treatment of B-cell malignancies, such as chronic lymphocytic leukemia (CLL) and multiple myeloma (MM), are thought to have strong potential. Progress, however, has been hampered by low efficacy and high toxicity. Tumor targeting by Vγ9Vδ2 T cells, a conserved T-cell subset with potent intrinsic antitumor properties, mediated by a bispecific antibody represents a novel approach promising high efficacy with limited toxicity. Here, we describe the generation of a bispecific Vγ9Vδ2 T-cell engager directed against CD40, which, due to its overexpression and biological footprint in malignant B cells, represents an attractive target. The CD40-targeting moiety of the bispecific antibody was selected because it can prevent CD40L-induced prosurvival signaling and reduce CD40-mediated resistance of CLL cells to venetoclax. Selective activation of Vγ9Vδ2 T cells in the presence of CD40+ tumor cells induced potent Vγ9Vδ2 T-cell degranulation, cytotoxicity against CLL and MM cells in vitro, and in vivo control of MM in a xenograft model. The CD40-bispecific γδ T-cell engager demonstrated lysis of leukemic cells by autologous Vγ9Vδ2 T cells present in patient-derived samples. Taken together, our CD40 bispecific γδ T-cell engager increased the sensitivity of leukemic cells to apoptosis and induced a potent Vγ9Vδ2 T cell-dependent antileukemic response. It may, therefore, represent a potential candidate for the development of novel treatments for B-cell malignancies.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- CD40 Antigens/immunology
- Cell Line, Tumor
- Female
- HEK293 Cells
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphocyte Activation/drug effects
- Male
- Mice
- Mice, Inbred NOD
- Middle Aged
- Signal Transduction/drug effects
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Iris de Weerdt
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Roeland Lameris
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - George L Scheffer
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Jana Vree
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Renate de Boer
- Department of Hematology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anita G Stam
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rieneke van de Ven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, the Netherlands
| | - Steven T Pals
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, the Netherlands
| | | | - Paul W H I Parren
- Lava Therapeutics, Utrecht, the Netherlands
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Arnon P Kater
- Department of Hematology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, the Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- Lava Therapeutics, Utrecht, the Netherlands
| |
Collapse
|
34
|
Micera A, Bruno L, Cacciamani A, Rongioletti M, Squitti R. Alzheimer's Disease and Retinal Degeneration: A Glimpse at Essential Trace Metals in Ocular Fluids and Tissues. Curr Alzheimer Res 2020; 16:1073-1083. [PMID: 31642780 DOI: 10.2174/1567205016666191023114015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/28/2019] [Accepted: 10/21/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Life expectancy is increasing all over the world, although neurodegenerative disorders might drastically affect the individual activity of aged people. Of those, Alzheimer's Disease (AD) is one of the most social-cost age-linked diseases of industrialized countries. To date, retinal diseases seem to be more common in the developing world and characterize principally aged people. Agerelated Macular Degeneration (AMD) is a late-onset, neurodegenerative retinal disease that shares several clinical and pathological features with AD, including stress stimuli such as oxidative stress, inflammation and amyloid formations. METHODS In both diseases, the detrimental intra/extra-cellular deposits have many similarities. Aging, hypercholesterolemia, hypertension, obesity, arteriosclerosis and smoking are risk factors to develop both diseases. Cellular aging routes have similar organelle and signaling patterns in retina and brain. The possibility to find out new research strategies represent a step forward to disclose potential treatment for both of them. Essential trace metals play critical roles in both physiological and pathological condition of retina, optic nerve and brain, by influencing metabolic processes chiefly upon complex multifactorial pathogenesis. CONCLUSION Hence, this review addresses current knowledge about some up-to-date investigated essential trace metals associated with AD and AMD. Changes in the levels of systemic and ocular fluid essential metals might reflect the early stages of AMD, possibly disclosing neurodegeneration pathways shared with AD, which might open to potential early detection.
Collapse
Affiliation(s)
- Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Rome, Italy
| | - Luca Bruno
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Rome, Italy
| | - Andrea Cacciamani
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Rome, Italy
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, San Giovanni Calibita, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Rosanna Squitti
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, BS, Italy
| |
Collapse
|
35
|
Pagliaro L, Sorrentino C, Roti G. Targeting Notch Trafficking and Processing in Cancers. Cells 2020; 9:E2212. [PMID: 33003595 PMCID: PMC7600097 DOI: 10.3390/cells9102212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
The Notch family comprises a group of four ligand-dependent receptors that control evolutionarily conserved developmental and homeostatic processes and transmit signals to the microenvironment. NOTCH undergoes remodeling, maturation, and trafficking in a series of post-translational events, including glycosylation, ubiquitination, and endocytosis. The regulatory modifications occurring in the endoplasmic reticulum/Golgi precede the intramembrane γ-secretase proteolysis and the transfer of active NOTCH to the nucleus. Hence, NOTCH proteins coexist in different subcellular compartments and undergo continuous relocation. Various factors, including ion concentration, enzymatic activity, and co-regulatory elements control Notch trafficking. Interfering with these regulatory mechanisms represents an innovative therapeutic way to bar oncogenic Notch signaling. In this review, we briefly summarize the role of Notch signaling in cancer and describe the protein modifications required for NOTCH to relocate across different subcellular compartments. We focus on the functional relationship between these modifications and the corresponding therapeutic options, and our findings could support the development of trafficking modulators as a potential alternative to the well-known γ-secretase inhibitors.
Collapse
Affiliation(s)
| | | | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (L.P.); (C.S.)
| |
Collapse
|
36
|
Relevant Cytokines in the B Cell Lymphoma Micro-Environment. Cancers (Basel) 2020; 12:cancers12092525. [PMID: 32899476 PMCID: PMC7564074 DOI: 10.3390/cancers12092525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/22/2020] [Accepted: 09/02/2020] [Indexed: 12/30/2022] Open
Abstract
Cytokines are soluble protein factors with importance in intercellular communication and, as such, play pivotal roles in the pathogenesis of B cell malignancies. Evidence from in vitro cultures permitted us to choose example cytokines that bind to different biochemical receptor types. Activated malignant B cells or stromal fibroblasts and macrophages prominently secrete the chemokines CCL3 or CXCL12 and CXCL13, respectively. Apart from helper T cells, various cell types of the B cell lymphoma microenvironment are capable of producing the cytokines IL-4, IL-6, IL-10 and TNFα. Owing to its impact on the development of myeloid cells, CSF-1 is among important soluble factors in the B cell lymphoma microenvironment. Inhibitors of B cell receptor-associated kinases often act via the blockade of cytokine production, but also prevent cytokine effects, e.g., chemotaxis. Increments in blood levels in chronic lymphocytic leukemia patients compared to healthy donors and normalization upon treatment with ibrutinib can be explained by producing cell types and modulation of cytokine production observed in vitro.
Collapse
|
37
|
Distinct immune composition in lymph node and peripheral blood of CLL patients is reshaped during venetoclax treatment. Blood Adv 2020; 3:2642-2652. [PMID: 31506282 DOI: 10.1182/bloodadvances.2019000360] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Morbidity and mortality due to immunosuppression remain among the foremost clinical challenges in chronic lymphocytic leukemia (CLL). Although immunosuppression is considered to originate within the lymph node (LN) microenvironment, alterations in T and natural killer (NK) cells have almost exclusively been studied in peripheral blood (PB). Whereas chemoimmunotherapy further deteriorates immune function, novel targeted agents like the B-cell lymphoma 2 inhibitor venetoclax potentially spare nonmalignant lymphocytes; however, the effects of venetoclax on nonleukemic cells have not been explored. We address these unresolved issues using a comprehensive analysis of nonmalignant lymphocytes in paired LN and PB samples from untreated CLL patients, and by analyzing the effects of venetoclax-based treatment regimens on the immune system in PB samples from previously untreated and relapsed/refractory patients. CLL-derived LNs contained twice the amount of suppressive regulatory T cells (Tregs) and CLL supportive follicular T helper (Tfh) cells compared with PB. This was accompanied by a low frequency of cytotoxic lymphocytes. The expression of PD-1 by CD8+ T cells was significantly higher in LN compared with PB. Venetoclax-based treatment led to deep responses in the majority of patients, but also to decreased absolute numbers of B, T, and NK cells. Tfh cell, Treg, and PD-1+ CD8+ T cell numbers were reduced more than fivefold after venetoclax-based therapy, and overproduction of inflammatory cytokines was reduced. Furthermore, we observed restoration of NK cell function. These data support the notion that the immunosuppressive state in CLL is more prominent within the LN. Venetoclax-based regimens reduced the immunosuppressive footprint of CLL, suggesting immune recovery after the elimination of leukemic cells.
Collapse
|
38
|
Relation of Neutrophil Gelatinase-Associated Lipocalin Overexpression to the Resistance to Apoptosis of Tumor B Cells in Chronic Lymphocytic Leukemia. Cancers (Basel) 2020; 12:cancers12082124. [PMID: 32751884 PMCID: PMC7465759 DOI: 10.3390/cancers12082124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
The resistance to apoptosis of chronic lymphocytic leukemia (CLL) cells partly results from the deregulated production of survival signals from leukemic cells. Despite the development of new therapies in CLL, drug resistance and disease relapse still occur. Recently, neutrophil gelatinase-associated lipocalin (NGAL), a secreted glycoprotein, has been suggested to have a critical role in the biology of tumors. Thus, we investigated the relevance of NGAL in CLL pathogenesis, analyzed the expression of its cellular receptor (NGAL-R) on malignant B cells and tested whether CLL cells are resistant to apoptosis through an autocrine process involving NGAL and NGAL-R. We observed that NGAL concentrations were elevated in the serum of CLL patients at diagnosis. After treatment (and regardless of the therapeutic regimen), serum NGAL levels normalized in CLL patients in remission but not in relapsed patients. In parallel, NGAL and NGAL-R were upregulated in leukemic cells from untreated CLL patients when compared to normal peripheral blood mononuclear cells (PBMCs), and returned to basal levels in PBMCs from patients in remission. Cultured CLL cells released endogenous NGAL. Anti-NGAL-R antibodies enhanced NGAL-R+ leukemia cell death. Conversely, recombinant NGAL protected NGAL-R+ CLL cells against apoptosis by activating a STAT3/Mcl-1 signaling pathway. Our results suggest that NGAL and NGAL-R, overexpressed in untreated CLL, participate in the deregulation of the apoptotic machinery in CLL cells, and may be potential therapeutic clues for CLL treatment.
Collapse
|
39
|
Zubair H, Khan MA, Anand S, Srivastava SK, Singh S, Singh AP. Modulation of the tumor microenvironment by natural agents: implications for cancer prevention and therapy. Semin Cancer Biol 2020; 80:237-255. [PMID: 32470379 PMCID: PMC7688484 DOI: 10.1016/j.semcancer.2020.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 05/10/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
The development of cancer is not just the growth and proliferation of a single transformed cell, but its surrounding environment also coevolves with it. Indeed, successful cancer progression depends on the ability of the tumor cells to develop a supportive tumor microenvironment consisting of various types of stromal cells. The interactions between the tumor and stromal cells are bidirectional and mediated through a variety of growth factors, cytokines, metabolites, and other biomolecules secreted by these cells. Tumor-stromal crosstalk creates optimal conditions for the tumor growth, metastasis, evasion of immune surveillance, and therapy resistance, and its targeting is being explored for clinical management of cancer. Natural agents from plants and marine life have been at the forefront of traditional medicine. Numerous epidemiological studies have reported the health benefits imparted on the consumption of certain fruits, vegetables, and their derived products. Indeed, a significant majority of anti-cancer drugs in clinical use are either naturally occurring compounds or their derivatives. In this review, we describe fundamental cellular and non-cellular components of the tumor microenvironment and discuss the significance of natural compounds in their targeting. Existing literature provides hope that novel prevention and therapeutic approaches will emerge from ongoing scientific efforts leading to the reduced tumor burden and improve clinical outcomes in cancer patients.
Collapse
Affiliation(s)
- Haseeb Zubair
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Mohammad Aslam Khan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Shashi Anand
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA; Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.
| |
Collapse
|
40
|
Allegra A, Musolino C, Tonacci A, Pioggia G, Casciaro M, Gangemi S. Clinico-Biological Implications of Modified Levels of Cytokines in Chronic Lymphocytic Leukemia: A Possible Therapeutic Role. Cancers (Basel) 2020; 12:cancers12020524. [PMID: 32102441 PMCID: PMC7072434 DOI: 10.3390/cancers12020524] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/08/2020] [Accepted: 02/22/2020] [Indexed: 12/19/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is the main cause of mortality among hematologic diseases in Western nations. B-CLL is correlated with an intense alteration of the immune system. The altered functions of innate immune elements and adaptive immune factors are interconnected in B-CLL and are decisive for its onset, evolution, and therapeutic response. Modifications in the cytokine balance could support the growth of the leukemic clone via a modulation of cellular proliferation and apoptosis, as some cytokines have been reported to be able to affect the life of B-CLL cells in vivo. In this review, we will examine the role played by cytokines in the cellular dynamics of B-CLL patients, interpret the contradictions sometimes present in the literature regarding their action, and evaluate the possibility of manipulating their production in order to intervene in the natural history of the disease.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Caterina Musolino
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy;
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Marco Casciaro
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
- Correspondence:
| |
Collapse
|
41
|
Rodriguez CM, Gilardoni MB, Remedi MM, Sastre D, Heller V, Pellizas CG, Donadio AC. Tumor-stroma interaction increases CD147 expression in neoplastic B lymphocytes in chronic lymphocytic leukemia. Blood Cells Mol Dis 2020; 82:102405. [PMID: 32007924 DOI: 10.1016/j.bcmd.2020.102405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) microenvironment plays a critical role in disease pathogenesis. Matrix metalloproteinases (MMPs) are involved in CLL-B cell migration and survival. CD147 is associated with MMPs production by tumor and stromal cells. AIM To analyze CD147, MMP2 and MMP9 expression in CLL-B cells and its modulation by fibroblasts (Fb)-CLL-B cell interaction. METHODS CLL-B cells were co-cultured with Fb, as a simulation of CLL microenvironment. CD147 was evaluated in healthy donor (HD)-B cells and CLL-B cells by flow cytometry. MMP2 and MMP9 activity in CLL-plasma samples and conditioned media (CMs) was studied by zymography. RESULTS MMP9/MMP2 plasma levels were significantly higher in CLL patients than in HD. CD147 expression (median fluorescence intensity) in CLL patients characterized 3 groups: low- (19.1 ± 3.2; n=3), middle- (42.7 ± 12.8; n=18) and high- (76.5 ± 9.6; n=5) related to CD147 expression in HD-B cells. CD147 expression significantly increased in CLL-B cells after Fb-CLL-B cell co-culture. A significant increase in proMMP2 activity was observed in CMs obtained from Fb-CLL-B cell co-cultures in comparison with isolated CLL-B cells. CONCLUSIONS CD147 expression in CLL-B cells and MMPs secretion was induced by Fb-CLL-B cell contact, suggesting CD147 participation in the CLL pathophysiology.
Collapse
Affiliation(s)
- Cecilia M Rodriguez
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina; Universidad Nacional de Córdoba, Hospital Nacional de Clínicas, Laboratorio de Oncohematología, Argentina
| | - Mónica B Gilardoni
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina.
| | - María M Remedi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| | - Darío Sastre
- Universidad Nacional de Córdoba, Hospital Nacional de Clínicas, Laboratorio de Oncohematología, Argentina
| | - Viviana Heller
- Universidad Nacional de Córdoba, Hospital Nacional de Clínicas, Laboratorio de Oncohematología, Argentina
| | - Claudia G Pellizas
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| | - Ana C Donadio
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Córdoba, Argentina
| |
Collapse
|
42
|
Matrix metalloproteinase-9 induces a pro-angiogenic profile in chronic lymphocytic leukemia cells. Biochem Biophys Res Commun 2019; 520:198-204. [DOI: 10.1016/j.bbrc.2019.09.127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/27/2019] [Indexed: 12/25/2022]
|
43
|
Abstract
AbstractPurposeThis article summarizes current targeted therapies that have received regulatory approval for the treatment of B- and T-cell lymphomas.SummaryOver the last 20 years, new drug therapies for lymphomas of B cells and T cells have expanded considerably. Targeted therapies for B-cell lymphomas include: (1) monoclonal antibodies directed at the CD20 lymphocyte antigen, examples of which are rituximab, ofatumumab, and obinutuzumab; (2) gene transfer therapy, an example of which is chimeric antigen receptor–modified T-cell (CAR-T) therapy directed at the CD19 antigen expressed on the cell surface of both immature and mature B cells; and (3) small-molecule inhibitors (ibrutinib, acalabrutinib, copanlisib, duvelisib, and idelalisib) that target the B-cell receptor signaling pathway. Of note, brentuximab vedotin is an antibody–drug conjugate that targets CD30, another lymphocyte antigen expressed on the cell surface of both Hodgkin lymphoma (a variant of B-cell lymphoma) and some T-cell lymphomas. Although aberrant epigenetic signaling pathways are present in both B- and T-cell lymphomas, epigenetic inhibitors (examples include belinostat, vorinostat, and romidepsin) are currently approved by the Food and Drug Administration for T-cell lymphomas only. In addition, therapies that target the tumor microenvironment have been developed. Examples include mogamulizumab, bortezomib, lenalidomide, nivolumab, and pembrolizumab. In summary, the efficacy of these agents has led to the development of supportive care to mitigate adverse effects, due to the presence of on- or off-target toxicities.ConclusionThe therapeutic landscape of lymphomas has continued to evolve. In turn, the efficacy of these agents has led to the development of supportive care to mitigate adverse effects, due to the presence of on- or off-target toxicities. Further opportunities are warranted to identify patients who are most likely to achieve durable response and reduce the risk of disease progression. Ongoing trials with current and investigational agents may further elucidate their place in therapy and therapeutic benefits.
Collapse
|
44
|
Hofland T, de Weerdt I, ter Burg H, de Boer R, Tannheimer S, Tonino SH, Kater AP, Eldering E. Dissection of the Effects of JAK and BTK Inhibitors on the Functionality of Healthy and Malignant Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2019; 203:2100-2109. [DOI: 10.4049/jimmunol.1900321] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/10/2019] [Indexed: 02/07/2023]
|
45
|
van Bruggen JAC, Martens AWJ, Fraietta JA, Hofland T, Tonino SH, Eldering E, Levin MD, Siska PJ, Endstra S, Rathmell JC, June CH, Porter DL, Melenhorst JJ, Kater AP, van der Windt GJW. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8 + T cells and impede CAR T-cell efficacy. Blood 2019; 134:44-58. [PMID: 31076448 PMCID: PMC7022375 DOI: 10.1182/blood.2018885863] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/20/2019] [Indexed: 01/02/2023] Open
Abstract
In chronic lymphocytic leukemia (CLL), acquired T-cell dysfunction impedes development of effective immunotherapeutic strategies, through as-yet unresolved mechanisms. We have previously shown that CD8+ T cells in CLL exhibit impaired activation and reduced glucose uptake after stimulation. CD8+ T cells in CLL patients are chronically exposed to leukemic B cells, which potentially impacts metabolic homeostasis resulting in aberrant metabolic reprogramming upon stimulation. Here, we report that resting CD8+ T cells in CLL have reduced intracellular glucose transporter 1 (GLUT1) reserves, and have an altered mitochondrial metabolic profile as displayed by increased mitochondrial respiration, membrane potential, and levels of reactive oxygen species. This coincided with decreased levels of peroxisome proliferator-activated receptor γ coactivator 1-α, and in line with that, CLL-derived CD8+ T cells showed impaired mitochondrial biogenesis upon stimulation. In search of a therapeutic correlate of these findings, we analyzed mitochondrial biogenesis in CD19-directed chimeric antigen receptor (CAR) CD8+ T cells prior to infusion in CLL patients (who were enrolled in NCT01747486 and NCT01029366 [https://clinicaltrials.gov]). Interestingly, in cases with a subsequent complete response, the infused CD8+ CAR T cells had increased mitochondrial mass compared with nonresponders, which positively correlated with the expansion and persistence of CAR T cells. Our findings demonstrate that GLUT1 reserves and mitochondrial fitness of CD8+ T cells are impaired in CLL. Therefore, boosting mitochondrial biogenesis in CAR T cells might improve the efficacy of CAR T-cell therapy and other emerging cellular immunotherapies.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Female
- Humans
- Immunotherapy, Adoptive
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Male
- Middle Aged
- Mitochondria/metabolism
- Organelle Biogenesis
- Receptors, Chimeric Antigen
Collapse
Affiliation(s)
- Jaco A C van Bruggen
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Anne W J Martens
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Joseph A Fraietta
- Department of Pathology and Laboratory Medicine
- Department of Microbiology
- Center for Cellular Immunotherapies, and
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tom Hofland
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Sanne H Tonino
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Peter J Siska
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; and
| | - Sanne Endstra
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Carl H June
- Department of Pathology and Laboratory Medicine
- Center for Cellular Immunotherapies, and
| | | | - J Joseph Melenhorst
- Department of Pathology and Laboratory Medicine
- Center for Cellular Immunotherapies, and
| | - Arnon P Kater
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
| | - Gerritje J W van der Windt
- Department of Hematology, Cancer Center Amsterdam
- Lymphoma and Myeloma Center Amsterdam, and
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, The Netherlands
| |
Collapse
|
46
|
Nguyen PH, Niesen E, Hallek M. New roles for B cell receptor associated kinases: when the B cell is not the target. Leukemia 2019; 33:576-587. [PMID: 30700840 DOI: 10.1038/s41375-018-0366-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
Abstract
Targeting of B cell receptor associated kinases (BAKs), such as Bruton's tyrosine kinase (BTK) or phosphoinositol-3-kinase (PI3K) delta, by specific inhibitors has revolutionized the therapy of B lymphoid malignancies. BAKs are critical signaling transducers of BCR signaling and seem relevant in B cell lymphoma pathogenesis. The functional relevance of BTK for lymphoid malignancies is strongly supported by the observation that resistance to therapy in CLL patients treated with BTK inhibitors such as ibrutinib is often associated with mutations in genes coding for BTK or Phospholipase-C gamma (PLCɣ). In some contrast, next generation sequencing data show that BAKs are mutated at very low frequency in treatment-naïve B cell lymphomas. Therefore, it remains debatable whether BAKs are essential drivers for lymphoma development. In addition, results obtained by targeted deletion of BAKs such as Lyn and Btk in murine CLL models suggest that BAKs may be essential to shape the dialogue between malignant B cells and the tumor microenvironment (TME). Since BAKs are expressed in multiple cell types, BAK inhibitors may disrupt the lymphoma supportive microenvironment. This concept also explains the typical response to BAK inhibitor treatment, characterized by a long-lasting increase of peripheral blood lymphoid cells, due to a redistribution from the lymphoid homing compartments. In addition, BAK inhibitors have shown some efficacy in solid tumors, probably through mediator cells in the TME. This review summarizes and validates the evidence for BAK inhibitors being part of a class of agents that modulate the (hematopoietic) microenvironment of cancers.
Collapse
Affiliation(s)
- Phuong-Hien Nguyen
- Department I of Internal Medicine, University Hospital of Cologne; Center for Integrated Oncology Cologne-Bonn; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases; Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany
| | - Emanuel Niesen
- Department I of Internal Medicine, University Hospital of Cologne; Center for Integrated Oncology Cologne-Bonn; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases; Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University Hospital of Cologne; Center for Integrated Oncology Cologne-Bonn; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases; Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
47
|
The phase 3 DUO trial: duvelisib vs ofatumumab in relapsed and refractory CLL/SLL. Blood 2018; 132:2446-2455. [PMID: 30287523 DOI: 10.1182/blood-2018-05-850461] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/25/2018] [Indexed: 01/07/2023] Open
Abstract
Duvelisib (also known as IPI-145) is an oral, dual inhibitor of phosphatidylinositol 3-kinase δ and γ (PI3K-δ,γ) being developed for treatment of hematologic malignancies. PI3K-δ,γ signaling can promote B-cell proliferation and survival in clonal B-cell malignancies, such as chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL). In a phase 1 study, duvelisib showed clinically meaningful activity and acceptable safety in CLL/SLL patients. We report here the results of DUO, a global phase 3 randomized study of duvelisib vs ofatumumab monotherapy for patients with relapsed or refractory (RR) CLL/SLL. Patients were randomized 1:1 to oral duvelisib 25 mg twice daily (n = 160) or ofatumumab IV (n = 159). The study met the primary study end point by significantly improving progression-free survival per independent review committee assessment compared with ofatumumab for all patients (median, 13.3 months vs 9.9 months; hazard ratio [HR] = 0.52; P < .0001), including those with high-risk chromosome 17p13.1 deletions [del(17p)] and/or TP53 mutations (HR = 0.40; P = .0002). The overall response rate was significantly higher with duvelisib (74% vs 45%; P < .0001) regardless of del(17p) status. The most common adverse events were diarrhea, neutropenia, pyrexia, nausea, anemia, and cough on the duvelisib arm, and neutropenia and infusion reactions on the ofatumumab arm. The DUO trial data support duvelisib as a potentially effective treatment option for patients with RR CLL/SLL. This trial was registered at www.clinicaltrials.gov as #NCT02004522.
Collapse
|
48
|
Jurj A, Pop L, Petrushev B, Pasca S, Dima D, Frinc I, Deak D, Desmirean M, Trifa A, Fetica B, Gafencu G, Selicean S, Moisoiu V, Micu WT, Berce C, Sacu A, Moldovan A, Colita A, Bumbea H, Tanase A, Dascalescu A, Zdrenghea M, Stiufiuc R, Leopold N, Tetean R, Burzo E, Tomuleasa C, Berindan-Neagoe I. Exosome-carried microRNA-based signature as a cellular trigger for the evolution of chronic lymphocytic leukemia into Richter syndrome. Crit Rev Clin Lab Sci 2018; 55:501-515. [PMID: 30238808 DOI: 10.1080/10408363.2018.1499707] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Even if considered a cumulative and not a proliferative CD5+ B-cell neoplasm, chronic lymphocytic leukemia (CLL) has a proliferation rate higher than that recognized earlier, especially in the lymphoid tissues. Some patients with CLL develop a clinical syndrome entitled Richter syndrome (RS). Understanding CLL genetics and epigenetics may help to elucidate the molecular basics of the clinical heterogeneity of this type of malignancy. In the present project we aimed to identify a microRNA species that can predict the evolution of therapy-resistant CLL towards RS. In the first phase of our study, microRNA-19b was identified as a possible target, and in the second phase, we transfected three different CLL cell lines with microRNA-19b mimic and inhibitor and assessed the potential role on leukemia cells in vitro. The mechanism by which miR-19b acts were identified as the upregulation of Ki67 and downregulation of p53. This was further supported through RT-PCR and western blotting on CLL cell lines, as well as by next generation sequencing on two patients diagnosed with CLL that evolved into RS.
Collapse
Affiliation(s)
- Ancuta Jurj
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Laura Pop
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Bobe Petrushev
- b Department of Pathology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Sergiu Pasca
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Delia Dima
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Ioana Frinc
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Dalma Deak
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Minodora Desmirean
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Adrian Trifa
- c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Bogdan Fetica
- b Department of Pathology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Grigore Gafencu
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Sonia Selicean
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Vlad Moisoiu
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Wilhelm-Thomas Micu
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Cristian Berce
- e Center for Experimental Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Alexandra Sacu
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Alin Moldovan
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania.,f Department of Hematology , Carol Davilla University of Medicine and Pharmacy , Bucharest , Romania
| | - Andrei Colita
- g Department of Hematology , Coltea Hospital , Bucharest , Romania
| | - Horia Bumbea
- f Department of Hematology , Carol Davilla University of Medicine and Pharmacy , Bucharest , Romania.,h Department of Hematology , University Hospital , Bucharest , Romania
| | - Alina Tanase
- h Department of Hematology , University Hospital , Bucharest , Romania.,i Department of Hematology , Fundeni Clinical Hospital , Bucharest , Romania
| | - Angela Dascalescu
- j Department of Hematology , Grigore T. Popa University of Medicine and Pharmacy , Iasi , Romania.,k Department of Hematology , Regional Institute of Oncology , Iasi , Romania
| | - Mihnea Zdrenghea
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Rares Stiufiuc
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Nicolae Leopold
- l Department of Physics , Babes Bolyai University , Cluj Napoca , Romania
| | - Romulus Tetean
- l Department of Physics , Babes Bolyai University , Cluj Napoca , Romania
| | - Emil Burzo
- l Department of Physics , Babes Bolyai University , Cluj Napoca , Romania.,m Romanian Academy , Romania
| | - Ciprian Tomuleasa
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania.,c Department of Hematology , Ion Chiricuta Oncology Institute , Cluj Napoca , Romania
| | - Ioana Berindan-Neagoe
- a Research Center for Functional Genomic, Biomedicine and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| |
Collapse
|
49
|
Improving CLL Vγ9Vδ2-T-cell fitness for cellular therapy by ex vivo activation and ibrutinib. Blood 2018; 132:2260-2272. [PMID: 30213872 DOI: 10.1182/blood-2017-12-822569] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/01/2018] [Indexed: 12/27/2022] Open
Abstract
The efficacy of autologous (αβ) T-cell-based treatment strategies in chronic lymphocytic leukemia (CLL) has been modest. The Vγ9Vδ2-T cell subset consists of cytotoxic T lymphocytes with potent antilymphoma activity via a major histocompatibility complex-independent mechanism. We studied whether Vγ9Vδ2-T cells can be exploited as autologous effector lymphocytes in CLL. Healthy control Vγ9Vδ2-T cells were activated by and had potent cytolytic activity against CLL cells. However, CLL-derived Vγ9Vδ2-T cells proved dysfunctional with respect to effector cytokine production and degranulation, despite an increased frequency of the effector-type subset. Consequently, cytotoxicity against malignant B cells was hampered. A comparable dysfunctional phenotype was observed in healthy Vγ9Vδ2-T cells after coculture with CLL cells, indicating a leukemia-induced mechanism. Gene-expression profiling implicated alterations in synapse formation as a conceivable contributor to compromised Vγ9Vδ2-T-cell function in CLL patients. Dysfunction of Vγ9Vδ2-T cells was fully reversible upon activation with autologous monocyte-derived dendritic cells (moDCs). moDC activation resulted in efficient expansion and predominantly yielded Vγ9Vδ2-T cells with a memory phenotype. Furthermore, ibrutinib treatment promoted an antitumor T helper 1 (TH1) phenotype in Vγ9Vδ2-T cells, and we demonstrated binding of ibrutinib to IL-2-inducible kinase (ITK) in Vγ9Vδ2-T cells. Taken together, CLL-mediated dysfunction of autologous Vγ9Vδ2-T cells is fully reversible, resulting in potent cytotoxicity toward CLL cells. Our data support the potential use of Vγ9Vδ2-T cells as effector T cells in CLL immunotherapy and favor further exploration of combining Vγ9Vδ2-T-cell-based therapy with ibrutinib.
Collapse
|
50
|
Constitutive IP 3 signaling underlies the sensitivity of B-cell cancers to the Bcl-2/IP 3 receptor disruptor BIRD-2. Cell Death Differ 2018; 26:531-547. [PMID: 29899382 PMCID: PMC6370760 DOI: 10.1038/s41418-018-0142-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/03/2022] Open
Abstract
Anti-apoptotic Bcl-2 proteins are upregulated in different cancers, including diffuse large B-cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL), enabling survival by inhibiting pro-apoptotic Bcl-2-family members and inositol 1,4,5-trisphosphate (IP3) receptor (IP3R)-mediated Ca2+-signaling. A peptide tool (Bcl-2/IP3R Disruptor-2; BIRD-2) was developed to abrogate the interaction of Bcl-2 with IP3Rs by targeting Bcl-2′s BH4 domain. BIRD-2 triggers cell death in primary CLL cells and in DLBCL cell lines. Particularly, DLBCL cells with high levels of IP3R2 were sensitive to BIRD-2. Here, we report that BIRD-2-induced cell death in DLBCL cells does not only depend on high IP3R2-expression levels, but also on constitutive IP3 signaling, downstream of the tonically active B-cell receptor. The basal Ca2+ level in SU-DHL-4 DLBCL cells was significantly elevated due to the constitutive IP3 production. This constitutive IP3 signaling fulfilled a pro-survival role, since inhibition of phospholipase C (PLC) using U73122 (2.5 µM) caused cell death in SU-DHL-4 cells. Milder inhibition of IP3 signaling using a lower U73122 concentration (1 µM) or expression of an IP3 sponge suppressed both BIRD-2-induced Ca2+ elevation and apoptosis in SU-DHL-4 cells. Basal PLC/IP3 signaling also fulfilled a pro-survival role in other DLBCL cell lines, including Karpas 422, RI-1 and SU-DHL-6 cells, whereas PLC inhibition protected these cells against BIRD-2-evoked apoptosis. Finally, U73122 treatment also suppressed BIRD-2-induced cell death in primary CLL, both in unsupported systems and in co-cultures with CD40L-expressing fibroblasts. Thus, constitutive IP3 signaling in lymphoma and leukemia cells is not only important for cancer cell survival, but also represents a vulnerability, rendering cancer cells dependent on Bcl-2 to limit IP3R activity. BIRD-2 seems to switch constitutive IP3 signaling from pro-survival into pro-death, presenting a plausible therapeutic strategy.
Collapse
|