1
|
Hoferkova E, Seda V, Kadakova S, Verner J, Loja T, Matulova K, Skuhrova Francova H, Ondrouskova E, Filip D, Blavet N, Boudny M, Mladonicka Pavlasova G, Vecera J, Ondrisova L, Pavelkova P, Hlavac K, Kostalova L, Michaelou A, Pospisilova S, Dorazilova J, Chochola V, Jaros J, Doubek M, Jarosova M, Hampl A, Vojtova L, Kren L, Mayer J, Mraz M. Stromal cells engineered to express T cell factors induce robust CLL cell proliferation in vitro and in PDX co-transplantations allowing the identification of RAF inhibitors as anti-proliferative drugs. Leukemia 2024; 38:1699-1711. [PMID: 38877102 PMCID: PMC11286525 DOI: 10.1038/s41375-024-02284-w] [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: 03/11/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 06/16/2024]
Abstract
Several in vitro models have been developed to mimic chronic lymphocytic leukemia (CLL) proliferation in immune niches; however, they typically do not induce robust proliferation. We prepared a novel model based on mimicking T-cell signals in vitro and in patient-derived xenografts (PDXs). Six supportive cell lines were prepared by engineering HS5 stromal cells with stable expression of human CD40L, IL4, IL21, and their combinations. Co-culture with HS5 expressing CD40L and IL4 in combination led to mild CLL cell proliferation (median 7% at day 7), while the HS5 expressing CD40L, IL4, and IL21 led to unprecedented proliferation rate (median 44%). The co-cultures mimicked the gene expression fingerprint of lymph node CLL cells (MYC, NFκB, and E2F signatures) and revealed novel vulnerabilities in CLL-T-cell-induced proliferation. Drug testing in co-cultures revealed for the first time that pan-RAF inhibitors fully block CLL proliferation. The co-culture model can be downscaled to five microliter volume for large drug screening purposes or upscaled to CLL PDXs by HS5-CD40L-IL4 ± IL21 co-transplantation. Co-transplanting NSG mice with purified CLL cells and HS5-CD40L-IL4 or HS5-CD40L-IL4-IL21 cells on collagen-based scaffold led to 47% or 82% engraftment efficacy, respectively, with ~20% of PDXs being clonally related to CLL, potentially overcoming the need to co-transplant autologous T-cells in PDXs.
Collapse
Affiliation(s)
- Eva Hoferkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Vaclav Seda
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sona Kadakova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jan Verner
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Loja
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Kvetoslava Matulova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Hana Skuhrova Francova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eva Ondrouskova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Daniel Filip
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Nicolas Blavet
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Miroslav Boudny
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | - Josef Vecera
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Laura Ondrisova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petra Pavelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Krystof Hlavac
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Kostalova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Androniki Michaelou
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Sarka Pospisilova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Dorazilova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Vaclav Chochola
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Josef Jaros
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michael Doubek
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marie Jarosova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ales Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lucy Vojtova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Leos Kren
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marek Mraz
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
2
|
Taghiloo S, Asgarian-Omran H. Cross-talk between leukemic and immune cells at the tumor microenvironment in chronic lymphocytic leukemia: An update review. Eur J Haematol 2024; 113:4-15. [PMID: 38698678 DOI: 10.1111/ejh.14224] [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: 02/20/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a mature-type B cell malignancy correlated with significant changes and defects in both the innate and adaptive arms of the immune system, together with a high dependency on the tumor microenvironment. Overall, the tumor microenvironment (TME) in CLL provides a supportive niche for leukemic cells to grow and survive, and interactions between CLL cells and the TME can contribute to disease progression and treatment resistance. Therefore, the increasing knowledge of the complicated interaction between immune cells and tumor cells, which is responsible for immune evasion and cancer progression, has provided an opportunity for the development of new therapeutic approaches. In this review, we outline tumor microenvironment-driven contributions to the licensing of immune escape mechanisms in CLL patients.
Collapse
Affiliation(s)
- Saeid Taghiloo
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Gastrointestinal Cancer Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
3
|
Ma M, Xie Y, Liu J, Wu L, Liu Y, Qin X. Biological effects of IL-21 on immune cells and its potential for cancer treatment. Int Immunopharmacol 2024; 126:111154. [PMID: 37977064 DOI: 10.1016/j.intimp.2023.111154] [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: 08/20/2023] [Revised: 10/28/2023] [Accepted: 10/29/2023] [Indexed: 11/19/2023]
Abstract
Interleukin-21 (IL-21), a member of the IL-2 cytokine family, is one of the most important effector and messenger molecules in the immune system. Produced by various immune cells, IL-21 has pleiotropic effects on innate and adaptive immune responses via regulation of natural killer, T, and B cells. An anti-tumor role of IL-21 has also been reported in the literature, as it may support cell proliferation or on the contrary induce growth arrest or apoptosis of the tumor cell. Anti-tumor effect of IL-21 enhances when combined with other agents that target tumor cells, immune regulatory circuits, or other immune-enhancing molecules. Therefore, understanding the biology of IL-21 in the tumor microenvironment (TME) and reducing its systemic toxic and side effects is crucial to ensure the maximum benefits of anti-tumor treatment strategies. In this review, we provide a comprehensive overview on the biological functions, roles in tumors, and the recent advances in preclinical and clinical research of IL-21 in tumor immunotherapy.
Collapse
Affiliation(s)
- Meichen Ma
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Xie
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yong Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 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
|
5
|
Williams MV, Mena-Palomo I, Cox B, Ariza ME. EBV dUTPase: A Novel Modulator of Inflammation and the Tumor Microenvironment in EBV-Associated Malignancies. Cancers (Basel) 2023; 15:855. [PMID: 36765813 PMCID: PMC9913121 DOI: 10.3390/cancers15030855] [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: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
There is increasing evidence that put into question the classical dogma that the Epstein-Barr virus (EBV) exists in cells as either a lytic virus in which new progeny is produced or in a latent state in which no progeny is produced. Notably, a third state has now been described, known as the abortive-lytic phase, which is characterized by the expression of some immediate early (IE) and early (E) genes, but no new virus progeny is produced. While the function of these IE and E gene products is not well understood, several recent studies support the concept they may contribute to tumor promotion by altering the tumor microenvironment (TME). The mechanisms by which these viral gene products may contribute to tumorigenesis remain unclear; however, it has been proposed that some of them promote cellular growth, immune evasion, and/or inhibit apoptosis. One of these EBV early gene products is the deoxyuridine triphosphate nucleotidohydrolase (dUTPase) encoded by BLLF3, which not only contributes to the establishment of latency through the production of activin A and IL-21, but it may also alter the TME, thus promoting oncogenesis.
Collapse
Affiliation(s)
- Marshall V. Williams
- Department of Cancer Biology and Genetics (CBG), The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research (IBMR), The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Irene Mena-Palomo
- Institute for Behavioral Medicine Research (IBMR), The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Brandon Cox
- Institute for Behavioral Medicine Research (IBMR), The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Maria Eugenia Ariza
- Department of Cancer Biology and Genetics (CBG), The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research (IBMR), The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Old and New Facts and Speculations on the Role of the B Cell Receptor in the Origin of Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:ijms232214249. [PMID: 36430731 PMCID: PMC9693457 DOI: 10.3390/ijms232214249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
The engagement of the B cell receptor (BcR) on the surface of leukemic cells represents a key event in chronic lymphocytic leukemia (CLL) since it can lead to the maintenance and expansion of the neoplastic clone. This notion was initially suggested by observations of the CLL BcR repertoire and of correlations existing between certain BcR features and the clinical outcomes of single patients. Based on these observations, tyrosine kinase inhibitors (TKIs), which block BcR signaling, have been introduced in therapy with the aim of inhibiting CLL cell clonal expansion and of controlling the disease. Indeed, the impressive results obtained with these compounds provided further proof of the role of BcR in CLL. In this article, the key steps that led to the determination of the role of BcR are reviewed, including the features of the CLL cell repertoire and the fine mechanisms causing BcR engagement and cell signaling. Furthermore, we discuss the biological effects of the engagement, which can lead to cell survival/proliferation or apoptosis depending on certain intrinsic cell characteristics and on signals that the micro-environment can deliver to the leukemic cells. In addition, consideration is given to alternative mechanisms promoting cell proliferation in the absence of BcR signaling, which can explain in part the incomplete effectiveness of TKI therapies. The role of the BcR in determining clonal evolution and disease progression is also described. Finally, we discuss possible models to explain the selection of a special BcR set during leukemogenesis. The BcR may deliver activation signals to the cells, which lead to their uncontrolled growth, with the possible collaboration of other still-undefined events which are capable of deregulating the normal physiological response of B cells to BcR-delivered stimuli.
Collapse
|
7
|
Matis S, Grazia Recchia A, Colombo M, Cardillo M, Fabbi M, Todoerti K, Bossio S, Fabris S, Cancila V, Massara R, Reverberi D, Emionite L, Cilli M, Cerruti G, Salvi S, Bet P, Pigozzi S, Fiocca R, Ibatici A, Angelucci E, Gentile M, Monti P, Menichini P, Fronza G, Torricelli F, Ciarrocchi A, Neri A, Fais F, Tripodo C, Morabito F, Ferrarini M, Cutrona G. MiR-146b-5p regulates IL-23 receptor complex expression in chronic lymphocytic leukemia cells. Blood Adv 2022; 6:5593-5612. [PMID: 35819446 PMCID: PMC9647700 DOI: 10.1182/bloodadvances.2021005726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 06/30/2022] [Indexed: 11/20/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells express the interleukin-23 receptor (IL-23R) chain, but the expression of the complementary IL-12Rβ1 chain requires cell stimulation via surface CD40 molecules (and not via the B-cell receptor [BCR]). This stimulation induces the expression of a heterodimeric functional IL-23R complex and the secretion of IL-23, initiating an autocrine loop that drives leukemic cell expansion. Based on the observation in 224 untreated Binet stage A patients that the cases with the lowest miR-146b-5p concentrations had the shortest time to first treatment (TTFT), we hypothesized that miR-146b-5p could negatively regulate IL-12Rβ1 side chain expression and clonal expansion. Indeed, miR-146b-5p significantly bound to the 3'-UTR region of the IL-12Rβ1 mRNA in an in vitro luciferase assay. Downregulation of miR-146b-5p with specific miRNA inhibitors in vitro led to the upregulation of the IL-12Rβ1 side chain and expression of a functional IL-23R complex similar to that observed after stimulation of the CLL cell through the surface CD40 molecules. Expression of miR-146b-5p with miRNA mimics in vitro inhibited the expression of the IL-23R complex after stimulation with CD40L. Administration of a miR-146b-5p mimic to NSG mice, successfully engrafted with CLL cells, caused tumor shrinkage, with a reduction of leukemic nodules and of IL-12Rβ1-positive CLL cells in the spleen. Our findings indicate that IL-12Rβ1 expression, a crucial checkpoint for the functioning of the IL-23 and IL-23R complex loop, is under the control of miR-146b-5p, which may represent a potential target for therapy since it contributes to the CLL pathogenesis. This trial is registered at www.clinicaltrials.gov as NCT00917540.
Collapse
Affiliation(s)
- Serena Matis
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Grazia Recchia
- Hematology Unit AO of Cosenza, Cosenza, Italy
- Biothecnology Research Unit, AO, Cosenza, Italy
| | - Monica Colombo
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Martina Cardillo
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Marina Fabbi
- Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Katia Todoerti
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sabrina Bossio
- Hematology Unit AO of Cosenza, Cosenza, Italy
- Biothecnology Research Unit, AO, Cosenza, Italy
| | - Sonia Fabris
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo School of Medicine, Palermo, Italy
| | - Rosanna Massara
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniele Reverberi
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Cilli
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giannamaria Cerruti
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sandra Salvi
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Bet
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Pigozzi
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical and Diagnostic Sciences (DISC), University of Genoa, Genoa, Italy
| | - Roberto Fiocca
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical and Diagnostic Sciences (DISC), University of Genoa, Genoa, Italy
| | - Adalberto Ibatici
- Hematology Unit and Transplant Center, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Emanuele Angelucci
- Hematology Unit and Transplant Center, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Massimo Gentile
- Hematology Unit AO of Cosenza, Cosenza, Italy
- Biothecnology Research Unit, AO, Cosenza, Italy
| | - Paola Monti
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Menichini
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gilberto Fronza
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Federica Torricelli
- Laboratory of Translational Research, Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonino Neri
- Scientific Directorate, Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Franco Fais
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo School of Medicine, Palermo, Italy
| | - Fortunato Morabito
- Biothecnology Research Unit, AO, Cosenza, Italy
- Hematology and Bone Marrow Transplant Unit, Hemato-Oncology Department, Augusta Victoria Hospital, East Jerusalem, Israel
| | - Manlio Ferrarini
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
8
|
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
|
9
|
Datta M, Jumaa H. Immunoglobulin Gene Sequence as an Inherited and Acquired Risk Factor for Chronic Lymphocytic Leukemia. Cancers (Basel) 2022; 14:cancers14133045. [PMID: 35804817 PMCID: PMC9264995 DOI: 10.3390/cancers14133045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/18/2022] [Accepted: 06/19/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Chronic lymphocytic leukemia (CLL) is the most prevalent among adult leukemias. Over the years, several research efforts discovered a lot of intricate details about the cause of the disease, its mechanism, and the prognostic factors that help to understand the progression and outcome of the disease. Mutations in the immunoglobulin gene sequences in B cells are the most important prognostic factor for CLL. The cells having no to very less mutations show aggressive disease, while those having more mutations are either fairly indolent or non-aggressive. In this review, we discussed the current gain of knowledge about these mutations and their effects in the overall disease pathology. Abstract Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disease characterized by the accumulation of CD5+ CD19+ malignant B cells. Autonomous ligand-independent B-cell signaling is a key process involved in the development of CLL pathogenesis. Together with other cytogenetic alterations, mutations in the immunoglobulin heavy chain variable (IGHV) gene act as a prognostic marker for CLL, with mutated CLL (M-CLL) being far more indolent than unmutated CLL (U-CLL). Recent studies highlight the role of a specific light chain mutation, namely, IGLV3-21R110G, in the development and prognosis of CLL. Such a mutation increases the propensity of homotypic BCR–BCR interaction, leading to cell autonomous signaling. In this article, we review the current findings on immunoglobulin gene sequence mutations as a potential risk factor for developing CLL.
Collapse
|
10
|
Gerousi M, Laidou S, Gemenetzi K, Stamatopoulos K, Chatzidimitriou A. Distinctive Signaling Profiles With Distinct Biological and Clinical Implications in Aggressive CLL Subsets With Stereotyped B-Cell Receptor Immunoglobulin. Front Oncol 2021; 11:771454. [PMID: 34804974 PMCID: PMC8595110 DOI: 10.3389/fonc.2021.771454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
The ontogeny and evolution of chronic lymphocytic leukemia (CLL) are critically dependent on interactions between leukemic cells and their microenvironment, including antigens, the latter recognized through the clonotypic B-cell receptor immunoglobulin (BcR IG). Antigen selection is key to the pathogenesis of CLL, as evidenced by the remarkable skewing of the BcR IG gene repertoire, culminating in BcR IG stereotypy, referring to the existence of subsets of patients with (quasi)identical BcR IG. Notably, certain of these subsets have been found to display distinct, subset-biased biological background, clinical presentation, and outcome, including the response to treatment. This points to BcR IG centrality while also emphasizing the need to dissect the signaling pathways triggered by the distinctive BcR IG expressed by different subsets, particularly those with aggressive clinical behavior. In this mini-review, we discuss the current knowledge on the implicated signaling pathways as well as the recurrent gene mutations in these pathways that characterize major aggressive stereotyped subsets. Special emphasis is given on the intertwining of BcR IG and Toll-like receptor (TLR) signaling and the molecular characterization of signaling activation, which has revealed novel players implicated in shaping clinical aggressiveness in CLL, e.g., the histone methyltransferase EZH2 and the transcription factor p63.
Collapse
Affiliation(s)
- Marina Gerousi
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Stamatia Laidou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Katerina Gemenetzi
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Chatzidimitriou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Garcia-Lacarte M, Grijalba SC, Melchor J, Arnaiz-Leché A, Roa S. The PD-1/PD-L1 Checkpoint in Normal Germinal Centers and Diffuse Large B-Cell Lymphomas. Cancers (Basel) 2021; 13:4683. [PMID: 34572910 PMCID: PMC8471895 DOI: 10.3390/cancers13184683] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Besides a recognized role of PD-1/PD-L1 checkpoint in anti-tumour immune evasion, there is accumulating evidence that PD-1/PD-L1 interactions between B and T cells also play an important role in normal germinal center (GC) reactions. Even when smaller in number, T follicular helper cells (TFH) and regulatory T (TFR) or B (Breg) cells are involved in positive selection of GC B cells and may result critical in the lymphoma microenvironment. Here, we discuss a role of PD-1/PD-L1 during tumour evolution in diffuse large B cell lymphoma (DLBCL), a paradigm of GC-derived lymphomagenesis. We depict a progression model, in two phases, where malignant B cells take advantage of positive selection signals derived from correct antigen-presentation and PD-1/PD-L1 inter-cellular crosstalks to survive and initiate tumour expansion. Later, a constant pressure for the accumulation of genetic/epigenetic alterations facilitates that DLBCL cells exhibit higher PD-L1 levels and capacity to secrete IL-10, resembling Breg-like features. As a result, a complex immunosuppressive microenvironment is established where DLBCL cells sustain proliferation and survival by impairing regulatory control of TFR cells and limiting IL-21-mediated anti-tumour functions of TFH cells and maximize the use of PD-1/PD-L1 signaling to escape from CD8+ cytotoxic activity. Integration of these molecular and cellular addictions into a framework may contribute to the better understanding of the lymphoma microenvironment and contribute to the rationale for novel PD-1/PD-L1-based combinational immunotherapies in DLBCL.
Collapse
Affiliation(s)
- Marcos Garcia-Lacarte
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
- Hemato-Oncology Program, Cima University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Sara C. Grijalba
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
| | - Javier Melchor
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
- Hemato-Oncology Program, Cima University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Adrián Arnaiz-Leché
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
| | - Sergio Roa
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
- Hemato-Oncology Program, Cima University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Network Center for Biomedical Research in Cancer—Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
12
|
CD4+ T cells sustain aggressive chronic lymphocytic leukemia in Eμ-TCL1 mice through a CD40L-independent mechanism. Blood Adv 2021; 5:2817-2828. [PMID: 34269799 DOI: 10.1182/bloodadvances.2020003795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is caused by the progressive accumulation of mature CD5+ B cells in secondary lymphoid organs. In vitro data suggest that CD4+ T lymphocytes also sustain survival and proliferation of CLL clones through CD40L/CD40 interactions. In vivo data in animal models are conflicting. To clarify this clinically relevant biological issue, we generated genetically modified Eμ-TCL1 mice lacking CD4+ T cells (TCL1+/+AB0), CD40 (TCL1+/+CD40-/-), or CD8+ T cells (TCL1+/+TAP-/-), and we monitored the appearance and progression of a disease that mimics aggressive human CLL by flow cytometry and immunohistochemical analyses. Findings were confirmed by adoptive transfer of leukemic cells into mice lacking CD4+ T cells or CD40L or mice treated with antibodies depleting CD4 T cells or blocking CD40L/CD40 interactions. CLL clones did not proliferate in mice lacking or depleted of CD4+ T cells, thus confirming that CD4+ T cells are essential for CLL development. By contrast, CD8+ T cells exerted an antitumor activity, as indicated by the accelerated disease progression in TCL1+/+TAP-/- mice. Antigen specificity of CD4+ T cells was marginal for CLL development, because CLL clones efficiently proliferated in transgenic mice whose CD4 T cells had a T-cell receptor with CLL-unrelated specificities. Leukemic clones also proliferated when transferred into wild-type mice treated with monoclonal antibodies blocking CD40 or into CD40L-/- mice, and TCL1+/+CD40-/- mice developed frank CLL. Our data demonstrate that CD8+ T cells restrain CLL progression, whereas CD4+ T cells support the growth of leukemic clones in TCL1 mice through CD40-independent and apparently noncognate mechanisms.
Collapse
|
13
|
Yang Z, Wu CAM, Targ S, Allen CDC. IL-21 is a broad negative regulator of IgE class switch recombination in mouse and human B cells. J Exp Med 2020; 217:133860. [PMID: 32130409 PMCID: PMC7201927 DOI: 10.1084/jem.20190472] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/24/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
IgE antibodies may elicit potent allergic reactions, and their production is tightly controlled. The tendency to generate IgE has been thought to reflect the balance between type 1 and type 2 cytokines, with the latter promoting IgE. Here, we reevaluated this paradigm by a direct cellular analysis, demonstrating that IgE production was not limited to type 2 immune responses yet was generally constrained in vivo. IL-21 was a critical negative regulator of IgE responses, whereas IFN-γ, IL-6, and IL-10 were dispensable. Follicular helper T cells were the primary source of IL-21 that inhibited IgE responses by directly engaging the IL-21 receptor on B cells and triggering STAT3-dependent signaling. We reconciled previous discordant results between mouse and human B cells and revealed that the inhibition of IgE class switch recombination by IL-21 was attenuated by CD40 signaling, whereas IgG1 class switch recombination was potentiated by IL-21 in the context of limited IL-4. These findings establish key features of the extrinsic regulation of IgE production by cytokines.
Collapse
Affiliation(s)
- Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Chung-An M Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Sasha Targ
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA.,Department of Anatomy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
14
|
Haselager MV, Kater AP, Eldering E. Proliferative Signals in Chronic Lymphocytic Leukemia; What Are We Missing? Front Oncol 2020; 10:592205. [PMID: 33134182 PMCID: PMC7578574 DOI: 10.3389/fonc.2020.592205] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells cycle between lymphoid tissue sites where they actively proliferate, and the peripheral blood (PB) where they become quiescent. Strong evidence exists for a crucial role of B cell receptor (BCR) triggering, either by (self-)antigen or by receptor auto-engagement in the lymph node (LN) to drive CLL proliferation and provide adhesion. The clinical success of Bruton's tyrosine kinase (BTK) inhibitors is widely accepted to be based on blockade of the BCR signal. Additional signals in the LN that support CLL survival derive from surrounding cells, such as CD40L-presenting T helper cells, myeloid and stromal cells. It is not quite clear if and to what extent these non-BCR signals contribute to proliferation in situ. In vitro BCR triggering, in contrast, leads to low-level activation and does not result in cell division. Various combinations of non-BCR signals delivered via co-stimulatory receptors, Toll-like receptors (TLRs), and/or soluble cytokines are applied, leading to comparatively modest and short-lived CLL proliferation in vitro. Thus, an unresolved gap exists between the condition in the patient as we now understand it and applicable knowledge that can be harnessed in the laboratory for future therapeutic applications. Even in this era of targeted drugs, CLL remains largely incurable with frequent relapses and emergence of resistance. Therefore, we require better insight into all aspects of CLL growth and potential rewiring of signaling pathways. We aim here to provide an overview of in vivo versus in vitro signals involved in CLL proliferation, point out areas of missing knowledge and suggest future directions for research.
Collapse
Affiliation(s)
- Marco V. Haselager
- Department of Experimental Immunology, Academic University Medical Center, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Cancer Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, Netherlands
| | - Arnon P. Kater
- Cancer Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, Netherlands
- Department of Hematology, Academic University Medical Center, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic University Medical Center, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Cancer Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, Netherlands
| |
Collapse
|
15
|
Giannoni P, Marini C, Cutrona G, Matis S, Capra MC, Puglisi F, Luzzi P, Pigozzi S, Gaggero G, Neri A, Todoerti K, Morabito F, Ibatici A, Miglino M, Bergamaschi M, Bruno S, Sambuceti GM, Ravetti JL, Ferrarini M, Fais F, de Totero D. Chronic lymphocytic leukemia cells impair osteoblastogenesis and promote osteoclastogenesis: role of TNFα, IL-6 and IL-11 cytokines. Haematologica 2020; 106:2598-2612. [PMID: 32855274 PMCID: PMC8485691 DOI: 10.3324/haematol.2019.231456] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Indexed: 01/18/2023] Open
Abstract
Bone skeletal alterations are no longer considered a rare event in Chronic Lymphocytic Leukemia (CLL), especially at more advanced stages of the disease. This study is aimed at elucidating the mechanisms underlying this phenomenon. Bone marrow stromal cells, induced to differentiate toward osteoblasts in osteogenic medium, appeared unable to complete their maturation upon co-culture with CLL cells, CLL cells-derived conditioned media (CLL-cm) or CLL-sera (CLL-sr). Inhibition of osteoblast differentiation was documented by decreased levels of RUNX2 and osteocalcin mRNA expression, by increased osteopontin and DKK-1 mRNA levels, and by a marked reduction of mineralized matrix deposition. The addition of neutralizing TNFα, IL-11 or anti-IL-6R monoclonal antibodies to these co-cultures resulted into restoration of bone mineralization, indicating the involvement of these cytokines: these findings were further supported by silencing TNFα, IL-11 and IL-6 in leukemic cells. We also demonstrated that the addition of CLL-cm to monocytes, previously stimulated with MCSF and RANKL, significantly amplified the formation of large mature osteoclasts as well as their bone resorption activity. Moreover enhanced osteoclastogenesis, induced by CLL-cm, was significantly reduced by treating cultures with the anti-TNFα moAb Infliximab; an analogous effect was observed by the use of the BTK inhibitor Ibrutinib. CLL cells, co-cultured with mature osteoclasts, were interestingly protected from apoptosis and upregulated Ki-67. These experimental results parallel the direct correlation between TNFα amounts in CLL sera and the degree of compact bone erosion we previously described, further strengthening the indication of a reciprocal influence between leukemic cells expansion and bone structure derangement.
Collapse
Affiliation(s)
- Paolo Giannoni
- Dept. of Experimental Medicine, Biology Section, University of Genoa, Italy
| | - Cecilia Marini
- CNR Institute of Bioimages and Molecular Physiology, Milan, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Serena Matis
- Molecular Pathology Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Francesca Puglisi
- Dept. of Experimental Medicine, Biology Section, University of Genoa, Italy
| | - Paola Luzzi
- Dept. of Experimental Medicine, Biology Section, University of Genoa, Italy
| | - Simona Pigozzi
- Pathological Anatomy Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Gabriele Gaggero
- Pathologycal Anatomy Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Antonino Neri
- Dept. of Oncology and Hemato-Oncology,University of Milan, Italy
| | - Katia Todoerti
- Dept. of Oncology and Hemato-Oncology,University of Milan, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Azienda Ospedaliera Aprigliano, Cosenza, Italy
| | - Adalberto Ibatici
- Hematology Clinic, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Maurizio Miglino
- Hematology Clinic, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Silvia Bruno
- Dept. of Experimental Medicine, Anatomy Section, University of Genoa, Italy
| | | | - Jean Louis Ravetti
- Pathological Anatomy Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Manlio Ferrarini
- Dept. of Experimental Medicine, Anatomy Section, University of Genoa, Italy
| | - Franco Fais
- Molecular Pathology Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela de Totero
- Molecular Pathology Unit, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy;
| |
Collapse
|
16
|
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: 5.5] [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
|
17
|
Chabab G, Bonnefoy N, Lafont V. IL-21 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:73-82. [PMID: 32060889 DOI: 10.1007/978-3-030-38315-2_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IL-21 is an immunomodulatory cytokine produced by natural killer (NK) cells and T cells that has pleiotropic roles in immune and nonimmune cells. IL-21 can modulate innate and specific immunity activities. It is a potent stimulator of T and natural killer cell-mediated antitumor immunity but also has pro-inflammatory functions in many tissues and is involved in oncogenesis. It is important to understand IL-21 biology in these different situations to ensure the maximal benefit of therapeutic strategies targeting this cytokine. This chapter summarizes IL-21 characteristics and signaling, its role in immune system components, and its use in cancer immunotherapies.
Collapse
Affiliation(s)
- Ghita Chabab
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Nathalie Bonnefoy
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Virginie Lafont
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.
| |
Collapse
|
18
|
Shahid A, Bharadwaj M. The connection between the Th17 cell related cytokines and cancer stem cells in cancer: Novel therapeutic targets. Immunol Lett 2019; 213:9-20. [PMID: 31278971 DOI: 10.1016/j.imlet.2019.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
Abstract
Cancer Stem Cells (CSCs) are the subpopulation of cells present in the different types of cancers with capabilities of self-renewal, differentiation, and tumorigenicity when transplanted into an animal host. The research work on the CSC has been providing a promising approach for the improvement of cancer therapies in the future. The CSCs have a close connection with the cytokines related with the T helper 17 (Th17) cell and other factors present in the tumor microenvironment, and these play a pivotal role in tumor progression and metastasis. The properties of CSCs are well defined in various type of tumor which is mainly developed by chemically and spontaneously in murine cancer model but in human defined primarily on acute myeloid leukemia, glioma, and breast cancer. The role of Th1, Th2, Natural Killer cells are well described in the cancer biology, but the Th17 cells are the subset which is recently exploited, and lots of research are going on. In this Review, we summarize current findings of the characteristics and functions of the Th17 cell and its signature cytokines in different cancers and their interconnections with cancer stem cells and with their markers. We have also discussed the functional properties of CSCs and how the CSCs markers can be distinguished from normal stem cells markers. We have also talked about the strategies that are efficiently targeting of CSCs and Th17 cells in different cancers.
Collapse
Affiliation(s)
- Ayaz Shahid
- Molecular Biology Group, National Institute of Cancer Prevention and Research, Indian Council of Medical Research (ICMR), Department of Health Research, Noida, 201301, India
| | - Mausumi Bharadwaj
- Molecular Biology Group, National Institute of Cancer Prevention and Research, Indian Council of Medical Research (ICMR), Department of Health Research, Noida, 201301, India.
| |
Collapse
|
19
|
Schleiss C, Ilias W, Tahar O, Güler Y, Miguet L, Mayeur-Rousse C, Mauvieux L, Fornecker LM, Toussaint E, Herbrecht R, Bertrand F, Maumy-Bertrand M, Martin T, Fournel S, Georgel P, Bahram S, Vallat L. BCR-associated factors driving chronic lymphocytic leukemia cells proliferation ex vivo. Sci Rep 2019; 9:701. [PMID: 30679590 PMCID: PMC6345919 DOI: 10.1038/s41598-018-36853-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/21/2018] [Indexed: 01/18/2023] Open
Abstract
A chronic antigenic stimulation is believed to sustain the leukemogenic development of chronic lymphocytic leukemia (CLL) and most of lymphoproliferative malignancies developed from mature B cells. Reproducing a proliferative stimulation ex vivo is critical to decipher the mechanisms of leukemogenesis in these malignancies. However, functional studies of CLL cells remains limited since current ex vivo B cell receptor (BCR) stimulation protocols are not sufficient to induce the proliferation of these cells, pointing out the need of mandatory BCR co-factors in this process. Here, we investigated benefits of several BCR co-stimulatory molecules (IL-2, IL-4, IL-15, IL-21 and CD40 ligand) in multiple culture conditions. Our results demonstrated that BCR engagement (anti-IgM ligation) concomitant to CD40 ligand, IL-4 and IL-21 stimulation allowed CLL cells proliferation ex vivo. In addition, we established a proliferative advantage for ZAP70 positive CLL cells, associated to an increased phosphorylation of ZAP70/SYK and STAT6. Moreover, the use of a tri-dimensional matrix of methylcellulose and the addition of TLR9 agonists further increased this proliferative response. This ex vivo model of BCR stimulation with T-derived cytokines is a relevant and efficient model for functional studies of CLL as well as lymphoproliferative malignancies.
Collapse
Affiliation(s)
- Cédric Schleiss
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR-S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France
| | - Wassila Ilias
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR-S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France
| | - Ouria Tahar
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR-S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France.,Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg, France
| | - Yonca Güler
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France
| | - Laurent Miguet
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France.,Laboratoire d'Hématologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Caroline Mayeur-Rousse
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France.,Laboratoire d'Hématologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laurent Mauvieux
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France.,Laboratoire d'Hématologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Luc-Matthieu Fornecker
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France.,Service d'Hématologie Adulte, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Elise Toussaint
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France.,Service d'Hématologie Adulte, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Raoul Herbrecht
- Université de Strasbourg, INSERM, IRFAC UMR-S1113, Strasbourg, France.,Service d'Hématologie Adulte, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Frédéric Bertrand
- Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France.,Institut de Recherche Mathématique Avancée IRMA, CNRS UMR 7501, Strasbourg, France
| | - Myriam Maumy-Bertrand
- Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France.,Institut de Recherche Mathématique Avancée IRMA, CNRS UMR 7501, Strasbourg, France
| | - Thierry Martin
- Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France.,CNRS UPR 9021 - Immunologie et Chimie Thérapeutiques, Institut de Biologie Moléculaire et cellulaire (IBMC), Strasbourg, France
| | - Sylvie Fournel
- CNRS UMR7199, Université de Strasbourg, Illkirch, France
| | - Philippe Georgel
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR-S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France
| | - Seiamak Bahram
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR-S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France. .,Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France. .,Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg, France.
| | - Laurent Vallat
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR-S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France. .,Fédération Hospitalo-Universitaire (FHU) OMICARE, Université de Strasbourg, Strasbourg, France. .,Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg, France. .,Université de Strasbourg, INSERM, IRFAC UMR-S1113, and Laboratoire d'Hématologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
20
|
Papakonstantinou N, Ntoufa S, Tsagiopoulou M, Moysiadis T, Bhoi S, Malousi A, Psomopoulos F, Mansouri L, Laidou S, Papazoglou D, Gounari M, Pasentsis K, Plevova K, Kuci-Emruli V, Duran-Ferrer M, Davis Z, Ek S, Rossi D, Gaidano G, Ritgen M, Oscier D, Stavroyianni N, Pospisilova S, Davi F, Ghia P, Hadzidimitriou A, Belessi C, Martin-Subero JI, Pott C, Rosenquist R, Stamatopoulos K. Integrated epigenomic and transcriptomic analysis reveals TP63 as a novel player in clinically aggressive chronic lymphocytic leukemia. Int J Cancer 2019; 144:2695-2706. [PMID: 30447004 DOI: 10.1002/ijc.31999] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 10/01/2018] [Accepted: 10/29/2018] [Indexed: 01/19/2023]
Abstract
Chronic lymphocytic leukemia (CLL) stereotyped subsets #6 and #8 include cases expressing unmutated B cell receptor immunoglobulin (BcR IG) (U-CLL). Subset #6 (IGHV1-69/IGKV3-20) is less aggressive compared to subset #8 (IGHV4-39/IGKV1(D)-39) which has the highest risk for Richter's transformation among all CLL. The underlying reasons for this divergent clinical behavior are not fully elucidated. To gain insight into this issue, here we focused on epigenomic signatures and their links with gene expression, particularly investigating genome-wide DNA methylation profiles in subsets #6 and #8 as well as other U-CLL cases not expressing stereotyped BcR IG. We found that subset #8 showed a distinctive DNA methylation profile compared to all other U-CLL cases, including subset #6. Integrated analysis of DNA methylation and gene expression revealed significant correlation for several genes, particularly highlighting a relevant role for the TP63 gene which was hypomethylated and overexpressed in subset #8. This observation was validated by quantitative PCR, which also revealed TP63 mRNA overexpression in additional nonsubset U-CLL cases. BcR stimulation had distinct effects on p63 protein expression, particularly leading to induction in subset #8, accompanied by increased CLL cell survival. This pro-survival effect was also supported by siRNA-mediated downregulation of p63 expression resulting in increased apoptosis. In conclusion, we report that DNA methylation profiles may vary even among CLL patients with similar somatic hypermutation status, supporting a compartmentalized approach to dissecting CLL biology. Furthermore, we highlight p63 as a novel prosurvival factor in CLL, thus identifying another piece of the complex puzzle of clinical aggressiveness.
Collapse
Affiliation(s)
- Nikos Papakonstantinou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Stavroula Ntoufa
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria Tsagiopoulou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Theodoros Moysiadis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Sujata Bhoi
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andigoni Malousi
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, Greece
| | - Fotis Psomopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Larry Mansouri
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Stamatia Laidou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Despoina Papazoglou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Maria Gounari
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Konstantinos Pasentsis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Karla Plevova
- Center of Molecular Biology and Gene Therapy, Department of Internal Medicine-Hematology and Oncology, University Hospital Brno and Medical Faculty of the Masaryk University, Brno, Czech republic
| | - Venera Kuci-Emruli
- Department of Immunotechnology, Faculty of Engineering, Lund University, Sweden
| | - Marti Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Departamento de Fundamentos Clínicos, Universitat de Barcelona, Barcelona, Spain
| | - Zadie Davis
- Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK
| | - Sara Ek
- Department of Immunotechnology, Faculty of Engineering, Lund University, Sweden
| | - Davide Rossi
- Hematology, Oncology Institute of Southern Switzerland and Institute of Oncology Research, Bellinzona, Switzerland
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Matthias Ritgen
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - David Oscier
- Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK
| | - Niki Stavroyianni
- Hematology Department and HCT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Sarka Pospisilova
- Center of Molecular Biology and Gene Therapy, Department of Internal Medicine-Hematology and Oncology, University Hospital Brno and Medical Faculty of the Masaryk University, Brno, Czech republic
| | - Frederic Davi
- Hematology Department and University Pierre et Marie Curie, Paris, France
| | - Paolo Ghia
- Division of Experimental Oncology, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Anastasia Hadzidimitriou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | | | - Jose I Martin-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Departamento de Fundamentos Clínicos, Universitat de Barcelona, Barcelona, Spain
| | - Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Yan L, Zhang J, Guo D, Ma J, Shui SF, Han XW. IL-21R functions as an oncogenic factor and is regulated by the lncRNA MALAT1/miR-125a-3p axis in gastric cancer. Int J Oncol 2018; 54:7-16. [PMID: 30387833 PMCID: PMC6255062 DOI: 10.3892/ijo.2018.4612] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022] Open
Abstract
Interleukin-21 receptor (IL-21R) is involved in the immunological regulation of immune cells and tumor progression in multiple malignancies. However, the potential molecular mechanisms through which non-coding RNAs (ncRNAs) modulate IL-21R signaling in gastric cancer (GC) remain elusive. In this study, the expression of IL-21R was detected by RT-qPCR and western blot analysis in GC cell lines. The association between IL-21R expression and clinicopathological characteristics and the prognosis of patients with GC was analyzed by immunohistochemistry and Kaplan-Meier plotter analysis. The biological functions of IL-21R were analyzed by a series of in vitro and in vivo experiments, and its regulation by ncRNAs was predicted by bioinformatics analysis and confirmed by luciferase assays and rescue experiments. As a result, the expression of IL-21R was found to be significantly increased in GC cell lines and tissues as compared with normal tissues, and was associated with tumor size and lymphatic metastasis, acting as an independent prognostic factor of poor survival and recurrence in patients with GC. The knockdown of IL-21R markedly suppressed GC cell proliferation and invasion, and IL-21R expression was further validated to be negatively regulated by miR-125a-3p (miR-125a). The overexpression of IL-21R reversed the tumor suppressive effects of miR-125a in vitro and in vivo. Moreover, lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) acted as a sponge of miR-125a to modulate the IL-21R signaling pathway in GC cells and represented a risk factor for survival and recurrence in patients with GC. Taken together, the findings of this study reveal an oncogenic role for IL-21R in gastric tumorigenesis and verify that its activation is partly due to the dysregulation of the lncRNA MALAT1/miR-125a axis. These findings may provide a potential prognostic marker for patients with GC.
Collapse
Affiliation(s)
- Lei Yan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Jing Zhang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Dong Guo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Ji Ma
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Shao-Feng Shui
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Xin-Wei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| |
Collapse
|
22
|
Sharpe C, Davis J, Mason K, Tam C, Ritchie D, Koldej R. Comparison of gene expression and flow cytometry for immune profiling in chronic lymphocytic leukaemia. J Immunol Methods 2018; 463:97-104. [PMID: 30267664 DOI: 10.1016/j.jim.2018.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
Understanding how cancer and cancer therapies affect the immune system is integral to the rational application of immunotherapies. Flow cytometry is the gold standard method of peripheral blood immune cell profiling. However, the requirement for viable cells can limit its applicability, especially in studies of retrospective clinical cohorts. We aimed to determine if gene expression, analysed using the NanoString platform, could be used to quantify the immune populations present in cryopreserved peripheral blood mononuclear cell (PBMC) samples from patients with chronic lymphocytic leukaemia. Cell abundance scores derived from gene expression analysis were significantly correlated with the population frequency quantified by flow cytometry for all subsets analysed, including T cells, NK cells and Monocytes. This study demonstrates that gene expression analysis can be applied to cryopreserved PBMC and provides a concordant and complementary understanding of the immune profile to flow cytometry.
Collapse
Affiliation(s)
- Chia Sharpe
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia.
| | - Joanne Davis
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kylie Mason
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia; Clinical Haematology and Bone Marrow Transplantation Service, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Constantine Tam
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Department of Haematology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - David Ritchie
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia; Clinical Haematology and Bone Marrow Transplantation Service, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Rachel Koldej
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Bousoik E, Montazeri Aliabadi H. "Do We Know Jack" About JAK? A Closer Look at JAK/STAT Signaling Pathway. Front Oncol 2018; 8:287. [PMID: 30109213 PMCID: PMC6079274 DOI: 10.3389/fonc.2018.00287] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022] Open
Abstract
Janus tyrosine kinase (JAK) family of proteins have been identified as crucial proteins in signal transduction initiated by a wide range of membrane receptors. Among the proteins in this family JAK2 has been associated with important downstream proteins, including signal transducers and activators of transcription (STATs), which in turn regulate the expression of a variety of proteins involved in induction or prevention of apoptosis. Therefore, the JAK/STAT signaling axis plays a major role in the proliferation and survival of different cancer cells, and may even be involved in resistance mechanisms against molecularly targeted drugs. Despite extensive research focused on the protein structure and mechanisms of activation of JAKs, and signal transduction through these proteins, their importance in cancer initiation and progression seem to be underestimated. This manuscript is an attempt to highlight the role of JAK proteins in cancer biology, the most recent developments in targeting JAKs, and the central role they play in intracellular cross-talks with other signaling cascades.
Collapse
Affiliation(s)
- Emira Bousoik
- Department of Biomedical and Pharmaceutical Sciences, Center for Targeted Drug Delivery, School of Pharmacy, Chapman University, Irvine, CA, United States.,School of Pharmacy, Omar Al-Mukhtar University, Dèrna, Libya
| | - Hamidreza Montazeri Aliabadi
- Department of Biomedical and Pharmaceutical Sciences, Center for Targeted Drug Delivery, School of Pharmacy, Chapman University, Irvine, CA, United States
| |
Collapse
|
24
|
Wu Y, van Besouw NM, Shi Y, Hoogduijn MJ, Wang L, Baan CC. The Biological Effects of IL-21 Signaling on B-Cell-Mediated Responses in Organ Transplantation. Front Immunol 2016; 7:319. [PMID: 27602031 PMCID: PMC4994014 DOI: 10.3389/fimmu.2016.00319] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/08/2016] [Indexed: 02/05/2023] Open
Abstract
Antibody-mediated rejection has emerged as one of the major issues limiting the success of organ transplantation. It exerts a highly negative impact on graft function and outcome, and effective treatment is lacking. The triggers for antibody development, and the mechanisms leading to graft dysfunction and failure, are incompletely understood. The production of antibodies is dependent on instructions from various immunocytes including CD4 T-helper cells that secrete interleukin (IL)-21 and interact with antigen-specific B-cells via costimulatory molecules. In this article, we discuss the role of IL-21 in the activation and differentiation of B-cells and consider the mechanisms of IL-21 and B-cell interaction. An improved understanding of the biological mechanisms involved in antibody-mediated complications after organ transplantation could lead to the development of novel therapeutic strategies, which control humoral alloreactivity, potentially preventing and treating graft-threatening antibody-mediated rejection.
Collapse
Affiliation(s)
- Yongkang Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China; Sector Nephrology & Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nicole M van Besouw
- Sector Nephrology & Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam , Rotterdam , Netherlands
| | - Yunying Shi
- Department of Nephrology, West China Hospital, Sichuan University , Chengdu , China
| | - Martin J Hoogduijn
- Sector Nephrology & Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam , Rotterdam , Netherlands
| | - Lanlan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University , Chengdu , China
| | - Carla C Baan
- Sector Nephrology & Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam , Rotterdam , Netherlands
| |
Collapse
|
25
|
Bhatt S, Sarosiek KA, Lossos IS. Interleukin 21 - its potential role in the therapy of B-cell lymphomas. Leuk Lymphoma 2016; 58:17-29. [PMID: 27405876 DOI: 10.1080/10428194.2016.1201568] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Interleukin-21 (IL-21), a member of IL-2 cytokine family, has pleotropic biological effects on lymphoid and myeloid cells. During the past 15 years, since the discovery of IL-21, great advances have been made regarding its biological activity and the mechanisms controlling IL-21-mediated cellular responses, especially in hematological malignancies. Preclinical studies have shown that IL-21R is expressed on healthy and neoplastic B-cells and exogenous IL-21 can induce direct apoptosis of IL-21R expressing B-cell non-Hodgkin lymphomas (NHL), making it a potentially attractive anti-lymphoma therapy. However, in some hematological malignancies such as multiple myeloma, Hodgkin lymphoma and Burkitt lymphoma, IL-21 can induce proliferation of neoplastic B-cells. In NHL, the underlying mechanism of cell death was found to be different between the various subtypes, including activation of different JAK/STAT signal transduction pathways or other factors. Immunomodulatory effects of IL-21 have also been reported to contribute to its anti-tumor effects as described by earlier studies in solid tumors and B-cell associated malignancies. These effects are predominantly mediated by IL-21's ability to activate cytolytic activities by NK-cells and CD4+/CD8+ T-cells. In this review, we provide an overview of IL-21's effects in NHL, results from clinical trials utilizing IL-21, and propose how IL-21 can be therapeutically exploited for treating these lymphomas.
Collapse
Affiliation(s)
- Shruti Bhatt
- a Dana-Farber Cancer Institute/Harvard Medical School , Boston , MA , USA
| | | | - Izidore S Lossos
- b Department of Molecular and Cellular Pharmacology , University of Miami Miller School of Medicine , Miami , FL , USA.,c Department of Medicine, Division of Hematology-Oncology , Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami , FL , USA
| |
Collapse
|
26
|
Browning RL, Byrd WH, Gupta N, Jones J, Mo X, Hertlein E, Yu L, Muthusamy N, Byrd JC. Lenalidomide Induces Interleukin-21 Production by T Cells and Enhances IL21-Mediated Cytotoxicity in Chronic Lymphocytic Leukemia B Cells. Cancer Immunol Res 2016; 4:698-707. [PMID: 27287425 DOI: 10.1158/2326-6066.cir-15-0291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/14/2016] [Indexed: 11/16/2022]
Abstract
The immunomodulatory drug lenalidomide has demonstrated efficacy in patients with chronic lymphocytic leukemia (CLL), despite a lack of direct cytotoxic effects in vitro The mechanism of lenalidomide efficacy in vivo is thought to occur via a combination of enhanced immune activity and an alteration of tumor cell-microenvironment interactions. We demonstrate in whole blood from patients with CLL that lenalidomide significantly depletes malignant B cells. Lenalidomide also induced production of interleukin-21 (IL21) and its mRNA in T cells from patients with CLL. In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. We show association of cell death with upregulation of Bid by IL21, enhanced upregulation of Bid by the combination therapy, and diminished Lck and downstream BCR signaling activation of Syk and PLCG2. Collectively, we demonstrated an immune cell-tumor cell interaction through lenalidomide-mediated induction of IL21 and IL21R, with enhanced IL21-mediated cytotoxicity, which provides justification for this combination in clinical trials for patients with CLL. Cancer Immunol Res; 4(8); 698-707. ©2016 AACR.
Collapse
Affiliation(s)
- Rebekah L Browning
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - William H Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio. Davidson College, Davidson, North Carolina
| | - Nikhil Gupta
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jeffrey Jones
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Erin Hertlein
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio.
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
27
|
Abstract
IL-21 is a type I cytokine produced by T cells and natural killer T cells that has pleiotropic actions on a wide range of immune and non-immune cell types. Since its discovery in 2000, extensive studies on the biological actions of IL-21 have been performed in vitro and in vivo. Recent reports describing patients with primary immunodeficiency caused by mutations of IL21 or IL21R have further deepened our knowledge of the role of this cytokine in host defense. Elucidation of the molecular mechanisms that mediate IL-21's actions has provided the rationale for targeting IL-21 and IL-21 downstream mediators for therapeutic purposes. The use of next-generation sequencing technology has provided further insights into the complexity of IL-21 signaling and has identified transcription factors and co-factors involved in mediating the actions of this cytokine. In this review, we discuss recent advances in the biology and signaling of IL-21 and how this knowledge can be potentially translated into clinical settings.
Collapse
Affiliation(s)
- Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethseda, Maryland, 20892, USA
| | - Chi-Keung Wan
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethseda, Maryland, 20892, USA
| |
Collapse
|
28
|
Kim N, Nam YS, Im KI, Lim JY, Lee ES, Jeon YW, Cho SG. IL-21-Expressing Mesenchymal Stem Cells Prevent Lethal B-Cell Lymphoma Through Efficient Delivery of IL-21, Which Redirects the Immune System to Target the Tumor. Stem Cells Dev 2015; 24:2808-21. [PMID: 26415081 DOI: 10.1089/scd.2015.0103] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Interleukin (IL)-21, a proinflammatory cytokine, has been developed as an immunotherapeutic approach due to its effects on various lymphocytes, including natural killer (NK) cells and T cells; however, the clinical success in cancer patients has been limited. Recently, mesenchymal stem cells (MSCs) have emerged as vehicles for cancer gene therapy due to their inherent migratory abilities toward tumors. In the present study, we hypothesized that MSCs, genetically modified to express high levels of IL-21 (IL-21/MSCs), can enhance antitumor responses through localized delivery of IL-21. For tumor induction, BALB/c mice were injected intravenously with syngeneic A20 B-cell lymphoma cells to develop a disseminated B-cell lymphoma model. Then, 6 days following tumor induction, the tumor-bearing mice were treated with IL-21/MSCs weekly, four times. Systemic infusion of A20 cells led to hind-leg paralysis as well as severe liver metastasis in the control group. The IL-21/MSC-treated group showed delayed tumor incidence as well as improved survival, whereas the MSC- and recombinant adenovirus-expressing IL-21 (rAD/IL-21)-treated groups did not show significant differences from the untreated mice. These therapeutic effects were associated with high levels of IL-21 delivered to the liver, which prevented the formation of tumor nodules. Furthermore, the infusion of IL-21/MSCs led to induction of effector T and NK cells, while potently inhibiting immune suppressor cells. Our findings demonstrate that IL-21-expressing MSCs have the therapeutic potential to induce potent antitumor effects against disseminated B-cell lymphoma through localized IL-21 delivery and induction of systemic antitumor immunity.
Collapse
Affiliation(s)
- Nayoun Kim
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,2 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease , Seoul, Korea
| | - Young-Sun Nam
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,2 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease , Seoul, Korea
| | - Keon-Il Im
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,2 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease , Seoul, Korea
| | - Jung-Yeon Lim
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,2 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease , Seoul, Korea
| | - Eun-Sol Lee
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,2 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease , Seoul, Korea
| | - Young-Woo Jeon
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,3 Department of Hematology, Catholic Blood and Marrow Transplantation Center, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine , Seoul, Korea
| | - Seok-Goo Cho
- 1 Institute for Translational Research and Molecular Imaging, The Catholic University of Korea College of Medicine , Seoul, Korea.,2 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease , Seoul, Korea.,3 Department of Hematology, Catholic Blood and Marrow Transplantation Center, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine , Seoul, Korea
| |
Collapse
|
29
|
De Cecco L, Capaia M, Zupo S, Cutrona G, Matis S, Brizzolara A, Orengo AM, Croce M, Marchesi E, Ferrarini M, Canevari S, Ferrini S. Interleukin 21 Controls mRNA and MicroRNA Expression in CD40-Activated Chronic Lymphocytic Leukemia Cells. PLoS One 2015; 10:e0134706. [PMID: 26305332 PMCID: PMC4549109 DOI: 10.1371/journal.pone.0134706] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/13/2015] [Indexed: 12/22/2022] Open
Abstract
Several factors support CLL cell survival in the microenvironment. Under different experimental conditions, IL21 can either induce apoptosis or promote CLL cell survival. To investigate mechanisms involved in the effects of IL21, we studied the ability of IL21 to modulate gene and miRNA expressions in CD40-activated CLL cells. IL21 was a major regulator of chemokine production in CLL cells and it modulated the expression of genes involved in cell movement, metabolism, survival and apoptosis. In particular, IL21 down-regulated the expression of the chemokine genes CCL4, CCL3, CCL3L1, CCL17, and CCL2, while it up-regulated the Th1-related CXCL9 and CXCL10. In addition, IL21 down-regulated the expression of genes encoding signaling molecules, such as CD40, DDR1 and PIK3CD. IL21 modulated a similar set of genes in CLL and normal B-cells (e.g. chemokine genes), whereas other genes, including MYC, TNF, E2F1, EGR2 and GAS-6, were regulated only in CLL cells. An integrated analysis of the miRNome and gene expression indicated that several miRNAs were under IL21 control and these could, in turn, influence the expression of potential target genes. We focused on hsa-miR-663b predicted to down-regulate several relevant genes. Transfection of hsa-miR-663b or its specific antagonist showed that this miRNA regulated CCL17, DDR1, PIK3CD and CD40 gene expression. Our data indicated that IL21 modulates the expression of genes mediating the crosstalk between CLL cells and their microenvironment and miRNAs may take part in this process.
Collapse
Affiliation(s)
- Loris De Cecco
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Capaia
- Laboratory of Biotherapy, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Simona Zupo
- Laboratory of Molecular Diagnostics, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Giovanna Cutrona
- Laboratory of Molecular Diagnostics, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Serena Matis
- Scientific Direction, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Antonella Brizzolara
- Laboratory of Biotherapy, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Anna Maria Orengo
- Laboratory of Biotherapy, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Michela Croce
- Laboratory of Biotherapy, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Edoardo Marchesi
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Manlio Ferrarini
- Scientific Direction, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Silvana Canevari
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- * E-mail: (SF); (SC)
| | - Silvano Ferrini
- Laboratory of Biotherapy, IRCCS-AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
- * E-mail: (SF); (SC)
| |
Collapse
|
30
|
Direct and immune-mediated cytotoxicity of interleukin-21 contributes to antitumor effects in mantle cell lymphoma. Blood 2015; 126:1555-64. [PMID: 26194763 DOI: 10.1182/blood-2015-01-624585] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 07/06/2015] [Indexed: 12/18/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a distinct subtype of non-Hodgkin lymphoma characterized by overexpression of cyclin D1 in 95% of patients. MCL patients experience frequent relapses resulting in median survival of 3 to 5 years, requiring more efficient therapeutic regimens. Interleukin (IL)-21, a member of the IL-2 cytokine family, possesses potent antitumor activity against a variety of cancers not expressing the IL-21 receptor (IL-21R) through immune activation. Previously, we established that IL-21 exerts direct cytotoxicity on IL-21R-expressing diffuse large B-cell lymphoma cells. Herein, we demonstrate that IL-21 possesses potent cytotoxicity against MCL cell lines and primary tumors. We identify that IL-21-induced direct cytotoxicity is mediated through signal transducer and activator of transcription 3-dependent cMyc upregulation, resulting in activation of Bax and inhibition of Bcl-2 and Bcl-XL. IL-21-mediated cMyc upregulation is only observed in IL-21-sensitive cells. Further, we demonstrate that IL-21 leads to natural killer (NK)-cell-dependent lysis of MCL cell lines that were resistant to direct cytotoxicity. In vivo treatment with IL-21 results in complete FC-muMCL1 tumor regression in syngeneic mice via NK- and T-cell-dependent mechanisms. Together, these data indicate that IL-21 has potent antitumor activity against MCL cells via direct cytotoxic and indirect, immune-mediated effects.
Collapse
|
31
|
Browning RL, Mo X, Muthusamy N, Byrd JC. CpG oligodeoxynucleotide CpG-685 upregulates functional interleukin-21 receptor on chronic lymphocytic leukemia B cells through an NF-κB mediated pathway. Oncotarget 2015; 6:15931-9. [PMID: 26158860 PMCID: PMC4599247 DOI: 10.18632/oncotarget.3285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/05/2015] [Indexed: 11/25/2022] Open
Abstract
CpG oligodeoxynucleotides (ODNs) upregulate the interleukin-21 receptor (IL21R) and enhance IL-21-mediated cytotoxicity in chronic lymphocytic leukemia (CLL) B cells. We demonstrate that treatment of CLL B cells with the ODN CpG-685 leads to increased IL21R expression, and that this increased expression enhances the effects of IL-21 treatment as evidenced by increased phosphorylation of JAK1, STAT1, and STAT3, as compared to IL-21 treatment without prior CpG stimulation. Induction of IL21R by CpG-685 also enhanced IL-21-mediated cytotoxicity. The mechanism by which CpG ODNs upregulate IL21R has not been elucidated, although IL21R regulation in T cells has been shown to be linked to T cell receptor-induced Sp1 binding to the IL21R promoter. Here, we demonstrate that luciferase reporter constructs containing the Sp1 binding site have increased basal luciferase activity compared to constructs lacking the Sp1 binding site, but fail to increase luciferase activity with CpG-685 stimulation in CLL B cells. By treating CLL cells with an NF-κB inhibitor, we inhibit the CpG ODN-mediated induction of IL21R, thus demonstrating that CpG-685 upregulates IL21R through an NF-κB mediated pathway. These findings suggest an alternative mechanism for induction of IL-21 receptor in CLL B cells and provide a basis for creation of future combination therapies.
Collapse
MESH Headings
- Humans
- I-kappa B Kinase/antagonists & inhibitors
- I-kappa B Kinase/metabolism
- Interleukin-21 Receptor alpha Subunit/biosynthesis
- Interleukin-21 Receptor alpha Subunit/genetics
- Interleukin-21 Receptor alpha Subunit/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- NF-kappa B/metabolism
- Oligodeoxyribonucleotides/genetics
- Oligodeoxyribonucleotides/pharmacology
- Sp1 Transcription Factor/metabolism
- Tumor Cells, Cultured
- Up-Regulation
Collapse
Affiliation(s)
- Rebekah L. Browning
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - John C. Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
32
|
IL-21: a pleiotropic cytokine with potential applications in oncology. J Immunol Res 2015; 2015:696578. [PMID: 25961061 PMCID: PMC4413888 DOI: 10.1155/2015/696578] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/19/2015] [Accepted: 03/25/2015] [Indexed: 12/29/2022] Open
Abstract
Interleukin- (IL-) 21 is a pleiotropic cytokine that regulates the activity of both innate and specific immunity. Indeed, it costimulates T and natural killer (NK) cell proliferation and function and regulates B cell survival and differentiation and the function of dendritic cells. In addition, IL-21 exerts divergent effects on different lymphoid cell leukemia and lymphomas, as it may support cell proliferation or on the contrary induce growth arrest or apoptosis of the neoplastic lymphoid cells. Several preclinical studies showed that IL-21 has antitumor activity in different tumor models, through mechanism involving the activation of NK and T or B cell responses. Moreover, IL-21's antitumor activity can be potentiated by its combination with other immune-enhancing molecules, monoclonal antibodies recognizing tumor antigens, chemotherapy, or molecular targeted agents. Clinical phase I-II studies of IL-21 in cancer patients showed immune stimulatory properties, acceptable toxicity profile, and antitumor effects in a fraction of patients. In view of its tolerability, IL-21 is also suitable for combinational therapeutic regimens with other agents. This review will summarize the biological functions of IL-21, and address its role in lymphoid malignancies and preclinical and clinical studies of cancer immunotherapy.
Collapse
|
33
|
Ghalamfarsa G, Jadidi-Niaragh F, Amiri MM, Razavi SM, Saboor-Yaraghi AA, Shokri F. All-trans-retinoic Acid differentially regulates proliferation of normal and leukemic B cells from different subsets of chronic lymphocytic leukemia. Nutr Cancer 2015; 67:285-91. [PMID: 25611428 DOI: 10.1080/01635581.2015.989368] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
All-trans-retinoic acid (ATRA) has been shown to modulate cell growth and differentiation in a variety of tumor cell types, but little is known regarding its precise role in regulation of leukemic B cells from different subsets of chronic lymphocytic leukemia (CLL). Previously, we showed that IL-21 significantly inhibits the CpG-mediated proliferation of CLL B cells in progressive compared to nonprogressive patients. In the present study, the effect of ATRA (10(-7) mol/L) on in vitro proliferation and apoptosis of B cells was investigated in 24 CLL patients and 8 normal subjects. Our results showed that ATRA markedly enhanced CpG-mediated proliferation of normal B cells, but it slightly inhibited CpG-induced proliferation of CLL B cells [stimulation index (SI): 105.6 vs. 14.7, P = 0.0001]. Although addition of IL-21 counteracted the proliferative effect of ATRA in normal B cells, it significantly enhanced the growth of tumor B cells in presence of CpG and ATRA. This stimulatory effect was restricted to nonprogressive and unmutated patients compared to progressive and mutated groups, respectively. Our results suggest that ATRA acts differentially on normal and CLL B cells and might have therapeutic implication in patients with progressive disease.
Collapse
Affiliation(s)
- Ghasem Ghalamfarsa
- a Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran and Immunology Research Center, Medical School , Mashhad University of Medical Sciences , Mashhad , Iran
| | | | | | | | | | | |
Collapse
|
34
|
Rasul E, Salamon D, Nagy N, Leveau B, Banati F, Szenthe K, Koroknai A, Minarovits J, Klein G, Klein E. The MEC1 and MEC2 lines represent two CLL subclones in different stages of progression towards prolymphocytic leukemia. PLoS One 2014; 9:e106008. [PMID: 25162594 PMCID: PMC4146575 DOI: 10.1371/journal.pone.0106008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/25/2014] [Indexed: 11/23/2022] Open
Abstract
The EBV carrying lines MEC1 and MEC2 were established earlier from explants of blood derived cells of a chronic lymphocytic leukemia (CLL) patient at different stages of progression to prolymphocytoid transformation (PLL). This pair of lines is unique in several respects. Their common clonal origin was proven by the rearrangement of the immunoglobulin genes. The cells were driven to proliferation in vitro by the same indigenous EBV strain. They are phenotypically different and represent subsequent subclones emerging in the CLL population. Furthermore they reflect the clinical progression of the disease. We emphasize that the support for the expression of the EBV encoded growth program is an important differentiation marker of the CLL cells of origin that was shared by the two subclones. It can be surmised that proliferation of EBV carrying cells in vitro, but not in vivo, reflects the efficient surveillance that functions even in the severe leukemic condition. The MEC1 line arose before the aggressive clinical stage from an EBV carrying cell within the subclone that was in the early prolymphocytic transformation stage while the MEC2 line originated one year later, from the subsequent subclone with overt PLL characteristics. At this time the disease was disseminated and the blood lymphocyte count was considerably elevated. The EBV induced proliferation of the MEC cells belonging to the subclones with markers of PLL agrees with earlier reports in which cells of PLL disease were infected in vitro and immortalized to LCL. They prove also that the expression of EBV encoded set of proteins can be determined at the event of infection. This pair of lines is particularly important as they provide in vitro cells that represent the subclonal evolution of the CLL disease. Furthermore, the phenotype of the MEC1 cells shares several characteristics of ex vivo CLL cells.
Collapse
MESH Headings
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- B-Lymphocytes/virology
- Biomarkers/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Clonal Evolution/immunology
- Clone Cells/immunology
- Clone Cells/pathology
- Clone Cells/virology
- Disease Progression
- Epstein-Barr Virus Nuclear Antigens/genetics
- Epstein-Barr Virus Nuclear Antigens/metabolism
- Gene Expression
- Herpesvirus 4, Human/physiology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/virology
- Leukemia, Prolymphocytic/immunology
- Leukemia, Prolymphocytic/pathology
- Leukemia, Prolymphocytic/virology
- Lymphocyte Count
- Time Factors
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/metabolism
- Viral Proteins/genetics
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Eahsan Rasul
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Instititet, Stockholm, Sweden
| | - Daniel Salamon
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Instititet, Stockholm, Sweden
| | - Noemi Nagy
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Instititet, Stockholm, Sweden
| | - Benjamin Leveau
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Instititet, Stockholm, Sweden
| | - Ferenc Banati
- RT-Europe Nonprofit Research Ltd, Mosonmagyaróvár, Hungary
| | - Kalman Szenthe
- RT-Europe Nonprofit Research Ltd, Mosonmagyaróvár, Hungary
| | - Anita Koroknai
- Microbiological Research Group, National Center for Epidemiology, Budapest, Hungary
| | - Janos Minarovits
- Microbiological Research Group, National Center for Epidemiology, Budapest, Hungary
- University of Szeged, Faculty of Dentistry, Department of Oral Biology and Experimental Dental Research, Szeged, Hungary
| | - George Klein
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Instititet, Stockholm, Sweden
| | - Eva Klein
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Instititet, Stockholm, Sweden
| |
Collapse
|
35
|
Giannoni P, Pietra G, Travaini G, Quarto R, Shyti G, Benelli R, Ottaggio L, Mingari MC, Zupo S, Cutrona G, Pierri I, Balleari E, Pattarozzi A, Calvaruso M, Tripodo C, Ferrarini M, de Totero D. Chronic lymphocytic leukemia nurse-like cells express hepatocyte growth factor receptor (c-MET) and indoleamine 2,3-dioxygenase and display features of immunosuppressive type 2 skewed macrophages. Haematologica 2014; 99:1078-87. [PMID: 24561793 DOI: 10.3324/haematol.2013.091405] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatocyte growth factor, produced by stromal and follicular dendritic cells, and present at high concentrations in the sera of patients with chronic lymphocytic leukemia, prolongs the survival of leukemic B cells by interacting with their receptor, c-MET. It is, however, unknown whether hepatocyte growth factor influences microenvironmental cells, such as nurse-like cells, which deliver survival signals to the leukemic clone. We evaluated the expression of c-MET on nurse-like cells and monocytes from patients with chronic lymphocytic leukemia and searched for phenotypic/functional features supposed to be influenced by the hepatocyte growth factor/c-MET interaction. c-MET is expressed at high levels on nurse-like cells and at significantly higher levels than normal on monocytes from patients. Moreover, the hepatocyte growth factor/c-MET interaction activates STAT3(TYR705) phosphorylation in nurse-like cells. Indoleamine 2,3-dioxygenase, an enzyme modulating T-cell proliferation and induced on normal monocytes after hepatocyte growth factor treatment, was detected together with interleukin-10 on nurse-like cells, and on freshly-prepared patients' monocytes. Immunohistochemical/immunostaining analyses demonstrated the presence of c-MET(+) and indoleamine 2,3-dioxygenase(+) cells in lymph node biopsies, co-expressed with CD68 and vimentin. Furthermore nurse-like cells and chronic lymphocytic monocytes significantly inhibited T-cell proliferation, prevented by anti-transforming growth factor beta and interleukin-10 antibodies and indoleamine 2,3-dioxygenase inhibitors, and supported CD4(+)CD25(high+)/FOXP3(+) T regulatory cell expansion. We suggest that nurse-like cells display features of immunosuppressive type 2 macrophages: higher hepatocyte growth factor levels, produced by leukemic or other microenvironmental surrounding cells, may cooperate to induce M2 polarization. Hepatocyte growth factor may thus have a dual pathophysiological role: directly through enhancement of survival of the leukemic clone and indirectly by favoring T-cell immunosuppression.
Collapse
Affiliation(s)
- Paolo Giannoni
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Gabriella Pietra
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Giorgia Travaini
- Transfer Gene Laboratory, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Rodolfo Quarto
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Genti Shyti
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy
| | - Roberto Benelli
- Immunology, Dept. of Integrated Oncology Therapies, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Laura Ottaggio
- Pathology and Molecular Diagnostic Laboratory, Dept. of Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Maria Cristina Mingari
- Dept. of Experimental Medicine (Di.Me.S), University of Genoa, Genoa, Italy Excellence Center for Biomedical Research, University of Genoa, Genoa, Italy Immunology, Dept. of Integrated Oncology Therapies, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Simona Zupo
- Pathology and Molecular Diagnostic Laboratory, Dept. of Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Giovanna Cutrona
- Pathology and Molecular Diagnostic Laboratory, Dept. of Service, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Ivana Pierri
- Clinical Oncohematology, University of Genoa, Genoa, Italy
| | - Enrico Balleari
- Clinic of Internal Medicine 3, Dept. of Hematology, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Marco Calvaruso
- Pathological Anatomy, Tumor Immunology Unit, Dept. of Science for Health Promotion and Maternal Infantile "G. D'Alessandro", Palermo, Italy
| | - Claudio Tripodo
- Pathological Anatomy, Tumor Immunology Unit, Dept. of Science for Health Promotion and Maternal Infantile "G. D'Alessandro", Palermo, Italy
| | | | | |
Collapse
|
36
|
Huang PY, Best OG, Almazi JG, Belov L, Davis ZA, Majid A, Dyer MJ, Pascovici D, Mulligan SP, Christopherson RI. Cell surface phenotype profiles distinguish stable and progressive chronic lymphocytic leukemia. Leuk Lymphoma 2014; 55:2085-92. [DOI: 10.3109/10428194.2013.867486] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Rodrigues L, Bonorino C. Role of IL-15 and IL-21 in viral immunity: applications for vaccines and therapies. Expert Rev Vaccines 2014; 8:167-77. [DOI: 10.1586/14760584.8.2.167] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Bhadra R, Cobb DA, Khan IA. CD40 signaling to the rescue: A CD8 exhaustion perspective in chronic infectious diseases. Crit Rev Immunol 2013; 33:361-78. [PMID: 23971530 DOI: 10.1615/critrevimmunol.2013007444] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chronic infectious diseases such as HIV, HBV, and HCV, among others, cause severe morbidity and mortality globally. Progressive decline in CD8 functionality, survival, and proliferative potential-a phenomenon referred to as CD8 exhaustion-is believed to be responsible for poor pathogen control in chronic infectious diseases. While the role of negative inhibitory receptors such as PD-1 in augmenting CD8 exhaustion has been extensively studied, the role of positive costimulatory receptors remains poorly understood. In this review, we discuss how one such costimulatory pathway, CD40-CD40L, regulates CD8 dysfunction and rescue. While the significance of this pathway has been extensively investigated in models of autoimmunity, acute infectious diseases, and tumor models, the role played by CD40-CD40L in regulating CD8 exhaustion in chronic infectious diseases is just beginning to be understood. Considering that monotherapy with blocking antibodies targeting inhibitory PD-1-PD-L1 pathway is only partially effective at ameliorating CD8 exhaustion and that humanized CD40 agonist antibodies are currently available, a better understanding of the role of the CD40-CD40L pathway in chronic infectious diseases will pave the way for the development of more robust immunotherapeutic and prophylactic vaccination strategies.
Collapse
Affiliation(s)
- Rajarshi Bhadra
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037, USA
| | | | | |
Collapse
|
39
|
Oppezzo P, Dighiero G. "Role of the B-cell receptor and the microenvironment in chronic lymphocytic leukemia''. Blood Cancer J 2013; 3:e149. [PMID: 24056719 PMCID: PMC3789209 DOI: 10.1038/bcj.2013.45] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 07/29/2013] [Accepted: 07/31/2013] [Indexed: 01/09/2023] Open
Abstract
Despite significant progress in treatment, chronic lymphocytic leukemia (CLL) remains an incurable disease. Advances have been made to understand the molecular pathogenesis underlying CLL progression and treatment resistance. We here review the available evidences concerning the role of the B-cell receptor (BCR) and the tumor microenvironment interactions in CLL pathogenesis. Antigen likely has a key role in the selection of the tumoral clone, the mutational status of immunoglobulin genes is a strong prognostic predictor and BCR signaling has been postulated to have a role for CLL trafficking and interaction with the stromal microenvironment. There is also important evidence, favoring a role for the microenvironment in CLL pathogenesis. Most, if not all, proliferative events occur in the lymph nodes and bone marrow, where leukemic cells receive through microenvironment interactions survival signals aiming to avoid apoptosis and acquire favorable tumoral growing conditions. In addition, the tumoral microenvironment appears to be the site where the acquisition of additional genetic lesions in the clone occur, which should greatly influence clinical outcome. The advent of new tyrosine kinase inhibitors which seem to be able to modulate microenvironment interactions and circumvent the p53 deletion have generated significant promise by raising the possibility that they could provide significant progress in disease treatment.
Collapse
Affiliation(s)
- P Oppezzo
- 1] Unit of Recombinant Protein, Institut Pasteur de Montevideo, Montevideo, Uruguay [2] Immunobiology Department, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | | |
Collapse
|
40
|
IL-21 and CD40L signals from autologous T cells can induce antigen-independent proliferation of CLL cells. Blood 2013; 122:3010-9. [PMID: 24014238 DOI: 10.1182/blood-2012-11-467670] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells multiply in secondary lymphoid tissue, but the mechanisms leading to their proliferation are still uncertain. In addition to B-cell receptor (BCR)-triggered signals, other microenvironmental factors might well be involved. In proliferation centers, leukemic B cells are in close contact with CD4(+)CD40L(+) T cells. Therefore, we here dissected the signals provided by autologous activated T cells (Tact) to CLL cells. Although the gene expression profile induced by Tact was highly similar to that induced by sole CD40 signaling, an obvious difference was that Tact induced proliferation of CLL cells. We determined that stimulation with only CD40L+IL-21 was sufficient to induce robust proliferation in CLL cells. We then defined an interleukin (IL)-21-induced gene signature in CLL, containing components of Janus kinase/signal transducer and activator of transcription and apoptosis pathways, and this signature could be detected in lymph node (LN) samples from patients. Finally, we could detect IL-21 RNA and protein in LN, and IL-21 production ex vivo by LN CD4(+)CXCR5(+) follicular helper T cells. These results indicate that in addition to BCR signaling, activated T cells might contribute to CLL cell proliferation via CD40 and IL-21. Targeting these signaling pathways might offer new venues for treatment of CLL.
Collapse
|
41
|
Milner JD, Holland SM. The cup runneth over: lessons from the ever-expanding pool of primary immunodeficiency diseases. Nat Rev Immunol 2013; 13:635-48. [PMID: 23887241 DOI: 10.1038/nri3493] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A recent surge in newly described primary immunodeficiencies (PIDs) has highlighted new physiological and pathophysiological pathways that affect the immune system. Furthermore, the study of individuals with PIDs has substantially improved our understanding of basic cellular and signalling pathways in host defence and immune regulation. Single-gene defects can lead to disease manifestations that range from extremely narrow infectious phenotypes to remarkably broad multisystem effects. Hypomorphic or hypermorphic gene mutations often occur in human diseases; when coupled with the fact that humans are exposed to naturally encountered antigens and pathogens, this helps to make the case that the study of immunological diseases in humans should be at the forefront of basic immunological research.
Collapse
Affiliation(s)
- Joshua D Milner
- Allergic Inflammation Unit, Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
42
|
Krejsa CM, Holly RD, Heipel M, Bannink KM, Johnson R, Roque R, Heffernan J, Hill J, Chin L, Wagener F, Shiota F, Henderson K, Sivakumar PV, Ren HP, Barahmand-pour F, Foster D, Clegg C, Kindsvogel W, Ponce R, Hughes SD, Waggie K. Interleukin-21 enhances rituximab activity in a cynomolgus monkey model of B cell depletion and in mouse B cell lymphoma models. PLoS One 2013; 8:e67256. [PMID: 23825648 PMCID: PMC3692496 DOI: 10.1371/journal.pone.0067256] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 05/15/2013] [Indexed: 11/18/2022] Open
Abstract
Rituximab, a monoclonal antibody targeting CD20 on B cells, is currently used to treat many subtypes of B cell lymphomas. However, treatment is not curative and response rates are variable. Recombinant interleukin-21 (rIL-21) is a cytokine that enhances immune effector function and affects both primary and transformed B cell differentiation. We hypothesized that the combination of rIL-21 plus rituximab would be a more efficacious treatment for B cell malignancies than rituximab alone. We cultured human and cynomolgus monkey NK cells with rIL-21 and found that their activity was increased and proteins associated with antibody dependent cytotoxicity were up-regulated. Studies in cynomolgus monkeys modeled the effects of rIL-21 on rituximab activity against CD20 B cells. In these studies, rIL-21 activated innate immune effectors, increased ADCC and mobilized B cells into peripheral blood. When rIL-21 was combined with rituximab, deeper and more durable B cell depletion was observed. In another series of experiments, IL-21 was shown to have direct antiproliferative activity against a subset of human lymphoma cell lines, and combination of murine IL-21 with rituximab yielded significant survival benefits over either agent alone in xenogeneic mouse tumor models of disseminated lymphoma. Therefore, our results do suggest that the therapeutic efficacy of rituximab may be improved when used in combination with rIL-21.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antibodies, Monoclonal, Murine-Derived/therapeutic use
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- B-Lymphocytes/cytology
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- Cell Line, Tumor
- Disease Models, Animal
- Drug Synergism
- Female
- Humans
- Immunity, Innate/drug effects
- Interleukins/pharmacology
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Macaca fascicularis
- Male
- Mice
- Rituximab
- Survival Analysis
Collapse
Affiliation(s)
- Cecile M. Krejsa
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Rick D. Holly
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Mark Heipel
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Ken M. Bannink
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Rebecca Johnson
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Richard Roque
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Jane Heffernan
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Julie Hill
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Lay Chin
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Felecia Wagener
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Faith Shiota
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Katherine Henderson
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Pallavur V. Sivakumar
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Hong-Ping Ren
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Fariba Barahmand-pour
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Don Foster
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Chris Clegg
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Wayne Kindsvogel
- Department of Research, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Rafael Ponce
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Steven D. Hughes
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
| | - Kim Waggie
- Department of Pre-clinical Development, ZymoGenetics, Incorporated, a Bristol-Myers Squibb Company, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
43
|
Ahearne MJ, Willimott S, Piñon L, Kennedy DB, Miall F, Dyer MJS, Wagner SD. Enhancement of CD154/IL4 proliferation by the T follicular helper (Tfh) cytokine, IL21 and increased numbers of circulating cells resembling Tfh cells in chronic lymphocytic leukaemia. Br J Haematol 2013; 162:360-70. [DOI: 10.1111/bjh.12401] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/16/2013] [Indexed: 01/27/2023]
Affiliation(s)
- Matthew J. Ahearne
- Department of Cancer Studies and Molecular Medicine; MRC Toxicology Unit; University of Leicester; Leicester; UK
| | - Shaun Willimott
- Department of Cancer Studies and Molecular Medicine; MRC Toxicology Unit; University of Leicester; Leicester; UK
| | - Lucia Piñon
- Department of Cancer Studies and Molecular Medicine; MRC Toxicology Unit; University of Leicester; Leicester; UK
| | | | - Fiona Miall
- Department of Haematology; Leicester Royal Infirmary; Leicester; UK
| | | | - Simon D. Wagner
- Department of Cancer Studies and Molecular Medicine; MRC Toxicology Unit; University of Leicester; Leicester; UK
| |
Collapse
|
44
|
Ramsay AD, Rodriguez-Justo M. Chronic lymphocytic leukaemia--the role of the microenvironment pathogenesis and therapy. Br J Haematol 2013; 162:15-24. [PMID: 23617880 DOI: 10.1111/bjh.12344] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Chronic lymphocytic leukaemia/small lymphocytic lymphoma (CLL) is one of the more common forms of B cell malignancy. Although the condition has a variable clinical course, the trend is towards eventual relapse and the disease is considered incurable. Whilst the majority of the circulating CD5-positive neoplastic B cells are arrested in the G0 phase of the cell cycle, those in the bone marrow and lymphoid tissues proliferate at a rate of 0·1-1% of the entire clone per day. This proliferation is supported by the tissue microenvironment, which has been shown to induce upregulation of anti-apoptotic proteins and enhance the survival of the neoplastic cells. Microenvironmental factors are also thought to be important in tumour relapse and resistance to therapy. This review outlines the main signalling pathways involved in these tumour cell-stromal interactions, and includes potential therapeutic strategies based on the manipulation of key components within the CLL microenvironment.
Collapse
Affiliation(s)
- Alan D Ramsay
- Department of Cellular Pathology, University College Hospital London, London, UK.
| | | |
Collapse
|
45
|
Differential regulation of B-cell proliferation by IL21 in different subsets of chronic lymphocytic leukemia. Cytokine 2013; 62:439-45. [PMID: 23579027 DOI: 10.1016/j.cyto.2013.03.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/21/2013] [Accepted: 03/23/2013] [Indexed: 02/06/2023]
Abstract
Interleukin-21 (IL21) plays an important role in B-cell proliferation, survival and differentiation. Contrary to its stimulatory effect in normal B cells, it has been shown that it induces pro-apoptotic effect in leukemic B cells from CLL patients. Little is known regarding the biological function of IL21 in leukemic B cells from progressive and non-progressive CLL patients. In the present study, the proliferative effect of IL21 in combination with TLR9 agonist (CpG) was investigated in B cells isolated from 24 CLL patients and eight normal subjects by radioactive thymidine incorporation assay. B cells were enriched from peripheral blood mononuclear cells by negative selection using magnetic beads (MACS) and immunophenotyped by flow cytometry. Our results showed that IL21 enhanced the proliferative effects of CpG in both normal and leukemic B cells, though no significant differences were observed between CLL patients and healthy controls. Comparison between different subsets of patients revealed that while the combination of IL21 and CpG significantly inhibited the proliferation of B cells from progressive compared to non-progressive patients (p=0.001), it enhanced proliferation of leukemic B cells from IGHV mutated compared to unmutated patients (p=0.001). The inhibitory effect of IL21 on proliferation of normal and leukemic cells was found to be apoptosis-independent. Our findings suggest differential effects of IL21 in different subsets of CLL patients and suggest its potential therapeutic implication in patients with a more progressive disease.
Collapse
|
46
|
Vallières F, Girard D. IL-21 Enhances Phagocytosis in Mononuclear Phagocyte Cells: Identification of Spleen Tyrosine Kinase as a Novel Molecular Target of IL-21. THE JOURNAL OF IMMUNOLOGY 2013; 190:2904-12. [DOI: 10.4049/jimmunol.1201941] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
IL-21 in the bone marrow microenvironment contributes to IgM secretion and proliferation of malignant cells in Waldenstrom macroglobulinemia. Blood 2012; 120:3774-82. [PMID: 22976953 DOI: 10.1182/blood-2012-03-419440] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytokines within the tumor microenvironment play an important role in supporting the growth and survival of B-cell malignancies. One such cytokine, IL-21, promotes the growth of myeloma and Hodgkin lymphoma cells while inducing apoptosis in chronic lymphocytic leukemia. However, the biologic significance of IL-21 has not been examined in Waldenstrom macroglobulinemia (WM), a B-cell lymphoma characterized by elevated serum IgM and a lymphoplasmacytic bone marrow infiltrate. We report here on the presence of IL-21 in the bone marrow of patients with WM and have identified activated T cells as the source of this cytokine. We readily detected the IL-21 receptor on malignant WM B cells and show that IL-21 significantly increases both IgM secretion and cellular proliferation of these cells with no effect on viability. IL-21 rapidly induces phosphorylation of STAT3 in WM cells, and treatment of the WM cell line MWCL-1 with a STAT3 inhibitor abolished the IL-21-mediated increases in cellular proliferation and IgM secretion. IL-21 also increased the expression of known STAT3 targets involved in B-cell differentiation, including BLIMP-1, XBP-1, IL-6, and IL-10. Overall, our data indicate that IL-21 in the bone marrow microenvironment significantly affects the biology of WM tumor cells through a STAT3-dependent mechanism.
Collapse
|
48
|
Timmerman JM, Byrd JC, Andorsky DJ, Yamada RE, Kramer J, Muthusamy N, Hunder N, Pagel JM. A phase I dose-finding trial of recombinant interleukin-21 and rituximab in relapsed and refractory low grade B-cell lymphoproliferative disorders. Clin Cancer Res 2012; 18:5752-60. [PMID: 22893631 DOI: 10.1158/1078-0432.ccr-12-0456] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We conducted a phase I study to determine the safety, maximum-tolerated dose (MTD), and efficacy of weekly bolus recombinant human interleukin-21 (rIL-21) plus rituximab in patients with indolent B-cell malignancies. EXPERIMENTAL DESIGN One week after a lead-in rituximab dose, cohorts of three patients were treated with 30, 100, or 150 μg/kg rIL-21 weekly for four weeks, concurrent with four weekly doses of rituximab. Patients with stable disease or better were eligible for a second course of therapy. RESULTS Twenty-one patients with relapsed small lymphocytic lymphoma/chronic lymphocytic leukemia (SLL/CLL, n = 11), follicular lymphoma (n = 9), or marginal zone lymphoma (n = 1) were enrolled, with 19 completing at least one course of therapy. The MTD for rIL-21 was 100 μg/kg, based on observed toxicities including nausea, vomiting, diarrhea, hypotension, edema, and hypophosphatemia. Clinical responses were seen in 8 of 19 evaluable patients (42%; 3 CR/CRu, 5 PR), with 4 of longer duration than the patient's previous response to rituximab-based treatment (median 9 months vs. 3 months). CONCLUSIONS Outpatient therapy of indolent B-cell malignancies with rituximab and weekly rIL-21 was well tolerated and clinically active, with durable complete remissions in a small subset of patients. Additional studies of rIL-21 and anti-CD20 antibodies in lymphoma and SLL/CLL are warranted.
Collapse
Affiliation(s)
- John M Timmerman
- Division of Hematology & Oncology, University of California Los Angeles Medical Center, Los Angeles, California 90095-1678, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Chronic lymphocytic leukemia (CLL) is a clinically heterogeneous disease, as some patients progress rapidly toward the more advanced studies, whereas others survive for a long period without the need for treatment. This heterogeneity of clinical course was somehow unexplained until studies on the CLL cell features disclosed that the CLL clones were heterogeneous and were characterized by different phenotypic and genotypic features in the different patients. On the basis of these observations, it was determined in retrospective studies that clones characterized by unmutated IGHV genes, and/or CD38 and/or ZAP-70 expression conferred a more severe prognosis to the CLL patients. Here, we present data on prospective studies carried out on Binet A-stage patients, in whom the markers were determined at diagnosis and their predictive value was assessed in comparison with chromosomal abnormalities and gene expression or micro RNA profiles. In addition, hypothesis on the potential pathogenetic role of these markers will be presented.
Collapse
|
50
|
van der Fits L, Out-Luiting JJ, van Leeuwen MA, Samsom JN, Willemze R, Tensen CP, Vermeer MH. Autocrine IL-21 Stimulation Is Involved in the Maintenance of Constitutive STAT3 Activation in Sézary Syndrome. J Invest Dermatol 2012; 132:440-7. [DOI: 10.1038/jid.2011.293] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|