1
|
Lopresti L, Tatangelo V, Baldari CT, Patrussi L. Rewiring the T cell-suppressive cytokine landscape of the tumor microenvironment: a new frontier for precision anti-cancer therapy. Front Immunol 2024; 15:1418527. [PMID: 39281678 PMCID: PMC11392891 DOI: 10.3389/fimmu.2024.1418527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
T lymphocytes that infiltrate the tumor microenvironment (TME) often fail to function as effective anti-cancer agents. Within the TME, cell-to-cell inhibitory interactions play significant roles in dampening their anti-tumor activities. Recent studies have revealed that soluble factors released in the TME by immune and non-immune cells, as well as by tumor cells themselves, contribute to the exacerbation of T cell exhaustion. Our understanding of the cytokine landscape of the TME, their interrelationships, and their impact on cancer development is still at its early stages. In this review, we aim to shed light on Interleukin (IL) -6, IL-9, and IL-10, a small group of JAK/STAT signaling-dependent cytokines harboring T cell-suppressive effects in the TME and summarize their mechanisms of action. Additionally, we will explore how advancements in scientific research can help us overcoming the obstacles posed by cytokines that suppress T cells in tumors, with the ultimate objective of stimulating further investigations for the development of novel therapeutic strategies to counteract their tumor-promoting activities.
Collapse
Affiliation(s)
| | | | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
2
|
Floerchinger A, Seiffert M. Lessons learned from the Eµ-TCL1 mouse model of CLL. Semin Hematol 2024; 61:194-200. [PMID: 38839457 DOI: 10.1053/j.seminhematol.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
The Eµ-TCL1 mouse model has been used for over 20 years to study the pathobiology of chronic lymphocytic leukemia (CLL) and for preclinical testing of novel therapies. A CLL-like disease develops with increasing age in these mice due to a B cell specific overexpression of human TCL1. The reliability of this model to mirror human CLL is controversially discussed, as none of the known driver mutations identified in patients are found in Eµ-TCL1 mice. It has to be acknowledged that this mouse model was key to develop targeted therapies that aim at inhibiting the constitutive B cell receptor (BCR) signaling, a main driver of CLL. Inhibitors of BCR signaling became standard-of-care for a large proportion of patients with CLL as they are highly effective. The Eµ-TCL1 model further advanced our understanding of CLL biology owed to studies that crossed this mouse line with various transgenic mouse models and demonstrated the relevance of CLL-cell intrinsic and -extrinsic drivers of disease. These studies were instrumental in showing the relevance of the tumor microenvironment in the lymphoid tissues for disease progression and immune escape in CLL. It became clear that CLL cells shape and rely on stromal and immune cells, and that immune suppressive mechanisms and T cell exhaustion contribute to CLL progression. Based on this knowledge, new immunotherapy strategies were clinically tested for CLL, but so far with disappointing results. As some of these therapies were effective in the Eµ-TCL1 mouse model, the question arose concerning the translatability of preclinical studies in these mice. The aim of this review is to summarize lessons we have learnt over the last decades by studying CLL-like disease in the Eµ-TCL1 mouse model. The article focuses on pitfalls and limitations of the model, as well as the gained knowledge and potential of using this model for the development of novel treatment strategies to achieve the goal of curing patients with CLL.
Collapse
MESH Headings
- Animals
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Mice
- Disease Models, Animal
- Humans
- Mice, Transgenic
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Tumor Microenvironment/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
Collapse
Affiliation(s)
- Alessia Floerchinger
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Biosciences of the University of Heidelberg, Heidelberg, Germany
| | - Martina Seiffert
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
3
|
Shang J, Hu S, Wang X. Targeting natural killer cells: from basic biology to clinical application in hematologic malignancies. Exp Hematol Oncol 2024; 13:21. [PMID: 38396050 PMCID: PMC10885621 DOI: 10.1186/s40164-024-00481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Natural killer (NK) cell belongs to innate lymphoid cell family that contributes to host immunosurveillance and defense without pre-immunization. Emerging studies have sought to understand the underlying mechanism behind NK cell dysfunction in tumor environments, and provide numerous novel therapeutic targets for tumor treatment. Strategies to enhance functional activities of NK cell have exhibited promising efficacy and favorable tolerance in clinical treatment of tumor patients, such as immune checkpoint blockade (ICB), chimeric antigen receptor NK (CAR-NK) cell, and bi/trispecific killer cell engager (BiKE/TriKE). Immunotherapy targeting NK cell provides remarkable advantages compared to T cell therapy, including a decreased rate of graft versus-host disease (GvHD) and neurotoxicity. Nevertheless, advanced details on how to support the maintenance and function of NK cell to obtain better response rate and longer duration still remain to be elucidated. This review systematically summarizes the profound role of NK cells in tumor development, highlights up-to-date advances and current challenges of therapy targeting NK cell in the clinical treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Juanjuan Shang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Taishan Scholars Program of Shandong Province, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
4
|
Leschiera E, Al-Hity G, Flint MS, Venkataraman C, Lorenzi T, Almeida L, Audebert C. An individual-based model to explore the impact of psychological stress on immune infiltration into tumour spheroids. Phys Biol 2024; 21:026003. [PMID: 38266283 DOI: 10.1088/1478-3975/ad221a] [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: 07/27/2023] [Accepted: 01/24/2024] [Indexed: 01/26/2024]
Abstract
In recentin vitroexperiments on co-culture between breast tumour spheroids and activated immune cells, it was observed that the introduction of the stress hormone cortisol resulted in a decreased immune cell infiltration into the spheroids. Moreover, the presence of cortisol deregulated the normal levels of the pro- and anti-inflammatory cytokines IFN-γand IL-10. We present an individual-based model to explore the interaction dynamics between tumour and immune cells under psychological stress conditions. With our model, we explore the processes underlying the emergence of different levels of immune infiltration, with particular focus on the biological mechanisms regulated by IFN-γand IL-10. The set-up of numerical simulations is defined to mimic the scenarios considered in the experimental study. Similarly to the experimental quantitative analysis, we compute a score that quantifies the level of immune cell infiltration into the tumour. The results of numerical simulations indicate that the motility of immune cells, their capability to infiltrate through tumour cells, their growth rate and the interplay between these cell parameters can affect the level of immune cell infiltration in different ways. Ultimately, numerical simulations of this model support a deeper understanding of the impact of biological stress-induced mechanisms on immune infiltration.
Collapse
Affiliation(s)
- Emma Leschiera
- Léonard de Vinci Pôle Universitaire, Research Center, 92 916 Paris, La Défense, France
- Univ. Bordeaux, CNRS, INRIA, Bordeaux INP, IMB, UMR 5251, F-33400 Talence, France
| | - Gheed Al-Hity
- School of Applied Sciences, University of Brighton, Centre for Stress and Age-related Diseases, Moulsecoomb, Brighton BN2 4GJ, United Kingdom
| | - Melanie S Flint
- School of Applied Sciences, University of Brighton, Centre for Stress and Age-related Diseases, Moulsecoomb, Brighton BN2 4GJ, United Kingdom
| | - Chandrasekhar Venkataraman
- School of Mathematical and Physical Sciences, University of Sussex, Department of Mathematics, Falmer, Brighton BN1 9QH, United Kingdom
| | - Tommaso Lorenzi
- Department of Mathematical Sciences 'G. L. Lagrange', Politecnico di Torino, 10129 Torino, Italy
| | - Luis Almeida
- Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR 7598, 75005 Paris, France
| | - Chloe Audebert
- Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR 7598, 75005 Paris, France
- Sorbonne Université, CNRS, Institut de biologie Paris-Seine (IBPS), Laboratoire de Biologie Computationnelle et Quantitative UMR 7238, 75005 Paris, France
| |
Collapse
|
5
|
Balla B, Tripon F, Lazar E, Bănescu C. Analysis of Mutational Status of IGHV, and Cytokine Polymorphisms as Prognostic Factors in Chronic Lymphocytic Leukemia: The Romanian Experience. Int J Mol Sci 2024; 25:1799. [PMID: 38339076 PMCID: PMC10855205 DOI: 10.3390/ijms25031799] [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: 12/05/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
The aim of the current study was to assess the associations between genetic risk factors (such as the mutational status of the IGHV gene and polymorphisms of the IL-10 and TNF-α genes) and CLL risk, prognosis, and overall survival. Another goal of this study was to evaluate the multivariate effect of the combination of multiple genetic risk factors (mutational status of the IGHV gene, somatic mutations, DNA CNVs, and cytokine SNPs) on the clinical characteristics and survival of patients. A total of 125 CLL patients and 239 healthy controls were included for comparative SNP analysis. IL-10 (rs1800896 and rs1800872) and TNF-α (rs361525 and rs1800750) SNPs and haplotypes were not associated with CLL risk. The absence of hypermutation in the IGHV gene was shown to be of important prognostic value, being associated with short OS. Further individual risk factors for short OS were an age above 65 years at diagnosis and the presence of somatic mutations and/or CNVs. In our multivariable analysis, the presence of somatic mutations and the IL-10 rs1800872 variant allele, and the association of CNVs with the IL-10 rs1800896 variant allele, were identified as risk factors for short OS. Moreover, the OS in unmutated IGHV patients was additionally affected (decreased) by the presence of CNVs and/or somatic mutations. Similarly, IL-10 rs1800896 modulated the OS in unmutated IGHV patients with CNVs.
Collapse
Affiliation(s)
- Beata Balla
- Department of Medical Genetics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (B.B.); (C.B.)
- Center for Advanced Medical and Pharmaceutical Research, Genetics Laboratory, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
- Medical Genetics Laboratory, Emergency County Hospital of Targu Mures, 540136 Targu Mures, Romania
| | - Florin Tripon
- Department of Medical Genetics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (B.B.); (C.B.)
- Center for Advanced Medical and Pharmaceutical Research, Genetics Laboratory, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
- Medical Genetics Laboratory, Emergency County Hospital of Targu Mures, 540136 Targu Mures, Romania
| | - Erzsebet Lazar
- Department of Internal Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Claudia Bănescu
- Department of Medical Genetics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (B.B.); (C.B.)
- Center for Advanced Medical and Pharmaceutical Research, Genetics Laboratory, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
- Medical Genetics Laboratory, Emergency County Hospital of Targu Mures, 540136 Targu Mures, Romania
| |
Collapse
|
6
|
Moss P. Ibrutinib reversal of immune exhaustion in CLL. Blood 2024; 143:5-7. [PMID: 38175677 DOI: 10.1182/blood.2023022243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
|
7
|
Sánchez Suárez MDM, Martín Roldán A, Alarcón-Payer C, Rodríguez-Gil MÁ, Poquet-Jornet JE, Puerta Puerta JM, Jiménez Morales A. Treatment of Chronic Lymphocytic Leukemia in the Personalized Medicine Era. Pharmaceutics 2023; 16:55. [PMID: 38258066 PMCID: PMC10818903 DOI: 10.3390/pharmaceutics16010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/26/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Chronic lymphocytic leukemia is a lymphoproliferative disorder marked by the expansion of monoclonal, mature CD5+CD23+ B cells in peripheral blood, secondary lymphoid tissues, and bone marrow. The disease exhibits significant heterogeneity, with numerous somatic genetic alterations identified in the neoplastic clone, notably mutated TP53 and immunoglobulin heavy chain mutational statuses. Recent studies emphasize the pivotal roles of genetics and patient fragility in treatment decisions. This complexity underscores the need for a personalized approach, tailoring interventions to individual genetic profiles for heightened efficacy. The era of personalized treatment in CLL signifies a transformative shift, holding the potential for improved outcomes in the conquest of this intricate hematologic disorder. This review plays a role in elucidating the evolving CLL treatment landscape, encompassing all reported genetic factors. Through a comprehensive historical analysis, it provides insights into the evolution of CLL management. Beyond its retrospective nature, this review could be a valuable resource for clinicians, researchers, and stakeholders, offering a window into the latest advancements. In essence, it serves as a dynamic exploration of our current position and the promising prospects on the horizon.
Collapse
Affiliation(s)
- María Del Mar Sánchez Suárez
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| | - Alicia Martín Roldán
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| | - Carolina Alarcón-Payer
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| | - Miguel Ángel Rodríguez-Gil
- Unidad de Gestión Clínica Hematología y Hemoterapia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.Á.R.-G.); (J.M.P.P.)
| | | | - José Manuel Puerta Puerta
- Unidad de Gestión Clínica Hematología y Hemoterapia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.Á.R.-G.); (J.M.P.P.)
| | - Alberto Jiménez Morales
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| |
Collapse
|
8
|
Silva FS, Barros-Lima A, Souza-Barros M, Crespo-Neto JA, Santos VGR, Pereira DS, Alves-Hanna FS, Magalhães-Gama F, Faria JAQA, Costa AG. A dual-role for IL-10: From leukemogenesis to the tumor progression in acute lymphoblastic leukemia. Cytokine 2023; 171:156371. [PMID: 37725872 DOI: 10.1016/j.cyto.2023.156371] [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: 06/28/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer in the world, and accounts for 25% of all childhood cancers among children under 15 years of age. Longitudinal studies have shown that children with ALL are born with a deregulated immune response that, together with postnatal environmental exposures, favor the onset of the disease. In this context, IL-10, a key cytokine in the regulation of the immune response, presents itself as a paradoxical mediator, initially influencing the development of ALL through the regulation of inflammatory processes and later on the progression of malignancy, with the increase of this molecule in the leukemia microenvironment. According to the literature, this cytokine plays a critical role in the natural history of the disease and plays an important role in two different though complex scenarios. Thus, in this review, we explore the dual role of IL-10 in ALL, and describe its biological characteristics, immunological mechanisms and genetics, as well as its impact on the leukemia microenvironment and its clinical implications.
Collapse
Affiliation(s)
- Flavio Souza Silva
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Amanda Barros-Lima
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Mateus Souza-Barros
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Juniel Assis Crespo-Neto
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | | | - Daniele Sá Pereira
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Fabíola Silva Alves-Hanna
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Fábio Magalhães-Gama
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil
| | - Jerusa Araújo Quintão Arantes Faria
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil; Escola de Enfermagem de Manaus, UFAM, Manaus, Brazil.
| |
Collapse
|
9
|
Cerreto M, Foà R, Natoni A. The Role of the Microenvironment and Cell Adhesion Molecules in Chronic Lymphocytic Leukemia. Cancers (Basel) 2023; 15:5160. [PMID: 37958334 PMCID: PMC10647257 DOI: 10.3390/cancers15215160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy whose progression largely depends on the lymph node and bone marrow microenvironment. Indeed, CLL cells actively proliferate in specific regions of these anatomical compartments, known as proliferation centers, while being quiescent in the blood stream. Hence, CLL cell adhesion and migration into these protective niches are critical for CLL pathophysiology. CLL cells are lodged in their microenvironment through a series of molecular interactions that are mediated by cellular adhesion molecules and their counter receptors. The importance of these adhesion molecules in the clinic is demonstrated by the correlation between the expression levels of some of them, in particular CD49d, and the prognostic likelihood. Furthermore, novel therapeutic agents, such as ibrutinib, impair the functions of these adhesion molecules, leading to an egress of CLL cells from the lymph nodes and bone marrow into the circulation together with an inhibition of homing into these survival niches, thereby preventing disease progression. Several adhesion molecules have been shown to participate in CLL adhesion and migration. Their importance also stems from the observation that they are involved in promoting, directly or indirectly, survival signals that sustain CLL proliferation and limit the efficacy of standard and novel chemotherapeutic drugs, a process known as cell adhesion-mediated drug resistance. In this respect, many studies have elucidated the molecular mechanisms underlying cell adhesion-mediated drug resistance, which have highlighted different signaling pathways that may represent potential therapeutic targets. Here, we review the role of the microenvironment and the adhesion molecules that have been shown to be important in CLL and their impact on transendothelial migration and cell-mediated drug resistance. We also discuss how novel therapeutic compounds modulate the function of this important class of molecules.
Collapse
Affiliation(s)
| | | | - Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00100 Rome, Italy; (M.C.); (R.F.)
| |
Collapse
|
10
|
Lan X, Zebley CC, Youngblood B. Cellular and molecular waypoints along the path of T cell exhaustion. Sci Immunol 2023; 8:eadg3868. [PMID: 37656775 PMCID: PMC10618911 DOI: 10.1126/sciimmunol.adg3868] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/09/2023] [Indexed: 09/03/2023]
Abstract
Thirty years of foundational research investigating molecular and cellular mechanisms promoting T cell exhaustion are now enabling rational design of T cell-based therapies for the treatment of chronic infections and cancer. Once described as a static cell fate, it is now well appreciated that the developmental path toward exhaustion is composed of a heterogeneous pool of cells with varying degrees of effector potential that ultimately converge on a terminally differentiated state. Recent description of the developmental stages along the differentiation trajectory of T cell exhaustion has provided insight into past immunotherapeutic success and future opportunities. Here, we discuss the hallmarks of distinct developmental stages occurring along the path to T cell dysfunction and the impact of these discrete CD8+ T cell fates on cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Lan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Caitlin C. Zebley
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ben Youngblood
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
11
|
Maher N, Mouhssine S, Matti BF, Alwan AF, Gaidano G. Treatment Refractoriness in Chronic Lymphocytic Leukemia: Old and New Molecular Biomarkers. Int J Mol Sci 2023; 24:10374. [PMID: 37373521 PMCID: PMC10299596 DOI: 10.3390/ijms241210374] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults. Despite its indolent clinical course, therapy refractoriness and disease progression still represent an unmet clinical need. Before the advent of pathway inhibitors, chemoimmunotherapy (CIT) was the commonest option for CLL treatment and is still widely used in areas with limited access to pathway inhibitors. Several biomarkers of refractoriness to CIT have been highlighted, including the unmutated status of immunoglobulin heavy chain variable genes and genetic lesions of TP53, BIRC3 and NOTCH1. In order to overcome resistance to CIT, targeted pathway inhibitors have become the standard of care for the treatment of CLL, with practice-changing results obtained through the inhibitors of Bruton tyrosine kinase (BTK) and BCL2. However, several acquired genetic lesions causing resistance to covalent and noncovalent BTK inhibitors have been reported, including point mutations of both BTK (e.g., C481S and L528W) and PLCG2 (e.g., R665W). Multiple mechanisms are involved in resistance to the BCL2 inhibitor venetoclax, including point mutations that impair drug binding, the upregulation of BCL2-related anti-apoptotic family members, and microenvironmental alterations. Recently, immune checkpoint inhibitors and CAR-T cells have been tested for CLL treatment, obtaining conflicting results. Potential refractoriness biomarkers to immunotherapy were identified, including abnormal levels of circulating IL-10 and IL-6 and the reduced presence of CD27+CD45RO- CD8+ T cells.
Collapse
Affiliation(s)
- Nawar Maher
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| | - Samir Mouhssine
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| | - Bassam Francis Matti
- Department of Hematology and Bone Marrow Transplant, Hematology and Bone Marrow Transplant Center, Baghdad 00964, Iraq;
| | - Alaa Fadhil Alwan
- Department of Clinical Hematology, The National Center of Hematology, Mustansiriyah University, Baghdad 10015, Iraq;
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| |
Collapse
|
12
|
Coyne V, Mead HL, Mongini PKA, Barker BM. B Cell Chronic Lymphocytic Leukemia Development in Mice with Chronic Lung Exposure to Coccidioides Fungal Arthroconidia. Immunohorizons 2023; 7:333-352. [PMID: 37195872 PMCID: PMC10579974 DOI: 10.4049/immunohorizons.2300013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/24/2023] [Indexed: 05/19/2023] Open
Abstract
Links between repeated microbial infections and B cell chronic lymphocytic leukemia (B-CLL) have been proposed but not tested directly. This study examines how prolonged exposure to a human fungal pathogen impacts B-CLL development in Eµ-hTCL1-transgenic mice. Monthly lung exposure to inactivated Coccidioides arthroconidia, agents of Valley fever, altered leukemia development in a species-specific manner, with Coccidioides posadasii hastening B-CLL diagnosis/progression in a fraction of mice and Coccidioides immitis delaying aggressive B-CLL development, despite fostering more rapid monoclonal B cell lymphocytosis. Overall survival did not differ significantly between control and C. posadasii-treated cohorts but was significantly extended in C. immitis-exposed mice. In vivo doubling time analyses of pooled B-CLL showed no difference in growth rates of early and late leukemias. However, within C. immitis-treated mice, B-CLL manifests longer doubling times, as compared with B-CLL in control or C. posadasii-treated mice, and/or evidence of clonal contraction over time. Through linear regression, positive relationships were noted between circulating levels of CD5+/B220low B cells and hematopoietic cells previously linked to B-CLL growth, albeit in a cohort-specific manner. Neutrophils were positively linked to accelerated growth in mice exposed to either Coccidioides species, but not in control mice. Conversely, only C. posadasii-exposed and control cohorts displayed positive links between CD5+/B220low B cell frequency and abundance of M2 anti-inflammatory monocytes and T cells. The current study provides evidence that chronic lung exposure to fungal arthroconidia affects B-CLL development in a manner dependent on fungal genotype. Correlative studies suggest that fungal species differences in the modulation of nonleukemic hematopoietic cells are involved.
Collapse
Affiliation(s)
- Vanessa Coyne
- Pathogen Microbiome Institute, Northern Arizona University, Flagstaff, AZ
| | - Heather L. Mead
- Pathogen Microbiome Institute, Northern Arizona University, Flagstaff, AZ
| | | | - Bridget M. Barker
- Pathogen Microbiome Institute, Northern Arizona University, Flagstaff, AZ
| |
Collapse
|
13
|
Andreescu M, Berbec N, Tanase AD. Assessment of Impact of Human Leukocyte Antigen-Type and Cytokine-Type Responses on Outcomes after Targeted Therapy Currently Used to Treat Chronic Lymphocytic Leukemia. J Clin Med 2023; 12:jcm12072731. [PMID: 37048814 PMCID: PMC10094967 DOI: 10.3390/jcm12072731] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Tumor growth and metastasis are reliant on intricate interactions between the host immune system and various counter-regulatory immune escape mechanisms employed by the tumor. Tumors can resist immune surveillance by modifying the expression of human leukocyte antigen (HLA) molecules, which results in the impaired presentation of tumor-associated antigens, subsequently evading detection and destruction by the immune system. The management of chronic lymphocytic leukemia (CLL) is based on symptom severity and includes various types of targeted therapies, including rituximab, obinutuzumab, ibrutinib, acalabrutinib, zanubrutinib, idelalisib, and venetoclax. These therapies rely on the recognition of specific peptides presented by HLAs on the surface of tumor cells by T cells, leading to an immune response. HLA class I molecules are found in most human cell types and interact with T-cell receptors (TCRs) to activate T cells, which play a vital role in inducing adaptive immune responses. However, tumor cells may evade T-cell attack by downregulating HLA expression, limiting the efficacy of HLA-dependent immunotherapy. The prognosis of CLL largely depends on the presence or absence of genetic abnormalities, such as del(17p), TP53 point mutations, and IGHV somatic hypermutation status. These oral targeted therapies alone or in combination with anti-CD20 antibodies have replaced chemoimmunotherapy as the primary treatment for CLL. In this review, we summarize the current clinical evidence on the impact of HLA- and cytokine-type responses on outcomes after targeted therapies currently used to treat CLL.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Department of Clinical Sciences, Hematology, Faculty of Medicine, Titu Maiorescu University of Bucharest, 040051 Bucharest, Romania
- Department of Hematology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Nicoleta Berbec
- Department of Hematology, Coltea Clinical Hospital, 020125 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Bucharest, 040051 Bucharest, Romania
| | - Alina Daniela Tanase
- Faculty of Medicine, Carol Davila University of Bucharest, 040051 Bucharest, Romania
- Department of Hematology, Fundeni Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
14
|
Chen Y, Shao X, Yang H, Ren L, Cui Y, Zhang W, Macip S, Meng X. Interferon gamma regulates a complex pro-survival signal network in chronic lymphocytic leukemia. Eur J Haematol 2023; 110:435-443. [PMID: 36576398 DOI: 10.1111/ejh.13921] [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: 09/19/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND It is known that the microenvironmental cytokine interferon gamma (IFN-γ) provides a survival advantage for chronic lymphocytic leukemia (CLL) cells. However, the mechanisms involved in this effect have not been properly investigated. METHODS Herein, we conducted a comprehensive screening of the effects of IFN-γ on signaling pathways and gene expression profiles in CLL cells by using western blotting, real-time quantitative reverse transcription (RT-qPCR) and high-throughput RNA sequencing (RNA-seq). RESULTS We found that IFN-γ not only activated the pro-survival signal transducer and activator of transcription 3 (STAT3), but also activated the protein kinase B and extracellular signal-regulated kinase signaling pathways. RNA-seq analysis showed that IFN-γ stimulation changed the expression profiles of more than 500 genes, with 391 being up-regulated and 123 down-regulated. These genes are involved in numerous biological processes, including anti-apoptosis, cell migration, and proliferation. IFN-γ significantly up-regulated the expression of CD38, BCL6, CXCL9, BCL2A1, SCOS3, IL-10, HGF, EGFR, THBS-1, FN1, and MUC1, which encode proteins potentially associated with disease progression, worse prognosis or poor response to treatment. Blocking janus kinases1/2 (JAK1/2) or STAT3 signal by specific inhibitors affected the expression of most genes, suggesting a pivotal role of the JAK1/2-STAT3 pathway in IFN-γ pro-survival effects in CLL. CONCLUSIONS Our data demonstrate that IFN-γ regulates a complex pro-survival signal network in CLL through JAK1/2-STAT3, which provides a rational explanation for IFN-γ promoting CLL cells survival and drug resistance.
Collapse
Affiliation(s)
- Yixiang Chen
- School of Basic Medical Science, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Luoyang, China
| | - Xiaoya Shao
- School of Basic Medical Science, Henan University of Science and Technology, Luoyang, China
| | - Haiping Yang
- First Affiliated Hospital, Henan University of Science and Technology, Luoyang, China
| | - Leiying Ren
- School of Basic Medical Science, Henan University of Science and Technology, Luoyang, China
| | - Ying Cui
- School of Basic Medical Science, Henan University of Science and Technology, Luoyang, China
| | - Wenlu Zhang
- School of Basic Medical Science, Henan University of Science and Technology, Luoyang, China
| | - Salvador Macip
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Xueqiong Meng
- School of Basic Medical Science, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
15
|
Ringelstein-Harlev S, Fanadka M, Horowitz NA, Bettman NP, Katz T. In chronic lymphocytic leukemia, activation of the thrombopoietin receptor promotes T-cell inhibitory properties, contributing to immunosuppression. Eur J Haematol 2023; 110:371-378. [PMID: 36478591 DOI: 10.1111/ejh.13912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
In chronic lymphocytic leukemia (CLL), the immune system is skewed towards a suppressive milieu. Levels of thrombopoietin (TPO), promoting cellular immune regulatory activity in immune thrombocytopenic purpura, were shown to be elevated in CLL patients. This study explored TPO as a potential immunomodulator, supporting CLL progression. We evaluated CLL cell-induced expression of TPO receptor (TPO-R) on T-cells and effects of its activation on T-cell responses. CLL cell involvement in TPO generation was also assessed. Baseline TPO-R expression on CD4 + T-cells was found to be higher in CLL patients than in healthy controls (HC). Exposure of HC-T-cells to B-cells, especially to CLL-B-cells stimulated with B-cell activating molecules, resulted in enhanced TPO-R expression on T-cells. CLL-T-cell stimulation with TPO reduced their proliferation and expanded the regulatory T-cell (Treg) population. At baseline, phosphorylation of STAT5, known to impact the Treg phenotype, was elevated in CLL-T-cells relative to those of HC. Exposure to TPO further enhanced STAT5 phosphorylation in CLL-T-cells, possibly driving the observed Treg expansion. The CLL immune milieu is involved in promotion of inhibitory features in T-cells through increased TPO-R levels and TPO-induced intracellular signaling. TPO and its signaling pathway could potentially support immunosuppression in CLL, and may emerge as novel therapeutic targets.
Collapse
Affiliation(s)
- Shimrit Ringelstein-Harlev
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Mona Fanadka
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Noam P Bettman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Tami Katz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
16
|
Spaner DE, Luo TY, Wang G, Schreiber G, Harari D, Shi Y. Paradoxical activation of chronic lymphocytic leukemia cells by ruxolitinib in vitro and in vivo. Front Oncol 2023; 13:1043694. [PMID: 37114129 PMCID: PMC10126367 DOI: 10.3389/fonc.2023.1043694] [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: 09/13/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Chronic lymphocytic leukemia (CLL) is characterized by an aberrant cytokine network that can support tumor growth by triggering janus kinase (JAK)/STAT pathways. Targeting cytokine-signaling should then be a rational therapeutic strategy but the JAK inhibitor ruxolitinib failed to control and seemingly accelerated the disease in clinical trials. Methods The effect of ruxolitinib on primary human CLL cells was studied in vitro and in vivo. Results Ruxolitinib increased phosphorylation of IRAK4, an important toll-like receptor (TLR)- signaling intermediate, in circulating CLL cells in vitro. It also enhanced p38 and NFKB1 phosphorylation while lowering STAT3 phosphorylation in CLL cells activated with TLR-7/8 agonists and IL-2. Among the cytokines made by activated CLL cells, high levels of IL-10 contributed strongly to STAT3 phosphorylation and inhibited TLR7 activity. Ruxolitinib limited TLR-mediated IL10 transcription and markedly reduced IL-10 production in vitro. It also decreased blood levels of IL-10 while increasing TNFα along with phospho-p38 expression and gene sets associated with TLR-activation in CLL cells in vivo. The bruton's tyrosine kinase inhibitor ibrutinib decreased IL-10 production in vitro but, in contrast to ruxolitinib, blocked initial IL10 transcription induced by TLR-signaling in vitro, decreased TNFα production, and deactivates CLL cells in vivo. Discussion These findings suggest the possible benefits of inhibiting growth factors with JAK inhibitors in CLL are outweighed by negative effects on potential tumor suppressors such as IL-10 that allow unrestrained activation of NFκB by drivers such as TLRs. Specific inhibition of growth-promoting cytokines with blocking antibodies or infusing suppressive cytokines like IL-10 might be better strategies to manipulate cytokines in CLL.
Collapse
Affiliation(s)
- David E. Spaner
- Biology Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Hematology, Sunnybrook Odette Cancer Center, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- *Correspondence: David E. Spaner,
| | - Tina YuXuan Luo
- Biology Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Guizhi Wang
- Biology Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Harari
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yonghong Shi
- Biology Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
17
|
Paulus A, Malavasi F, Chanan-Khan A. CD38 as a multifaceted immunotherapeutic target in CLL. Leuk Lymphoma 2022; 63:2265-2275. [DOI: 10.1080/10428194.2022.2090551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Aneel Paulus
- Department of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Fabio Malavasi
- Dipartimento Scienze Mediche, Università di Torino, Torino, Italy
- Fondazione Ricerca Molinette ONLUS, Università di Torino, Torino, Italy
| | - Asher Chanan-Khan
- Department of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
18
|
Collard JP, McKenna MK, Noothi SK, Alhakeem SS, Rivas JR, Rangnekar VM, Muthusamy N, Bondada S. Role of the splenic microenvironment in chronic lymphocytic leukemia development in Eµ-TCL1 transgenic mice. Leuk Lymphoma 2022; 63:1810-1822. [DOI: 10.1080/10428194.2022.2045596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- James P. Collard
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Mary K. McKenna
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Sunil K. Noothi
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Sara S. Alhakeem
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jacqueline R. Rivas
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Vivek M. Rangnekar
- Department of Radiation Medicine and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Natarajan Muthusamy
- Division of Hematology, James Cancer Center, Ohio State University, Columbus, OH, USA
| | - Subbarao Bondada
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
19
|
Allard D, Chrobak P, Bareche Y, Allard B, Tessier P, Bergeron MA, Johnson NA, Stagg J. CD73 Promotes Chronic Lymphocytic Leukemia. Cancers (Basel) 2022; 14:cancers14133130. [PMID: 35804900 PMCID: PMC9264813 DOI: 10.3390/cancers14133130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Many patients with chronic lymphocytic leukemia (CLL) still fail current therapies. CD73 is a novel therapeutic target for solid tumors, but its role in CLL remains unclear. The aim of our study was to investigate the therapeutic potential of targeting CD73 in CLL. Using genetically engineered mice, our study reports a pro-leukemic role for CD73 in an autochthonous mouse model of CLL. Furthermore, we observed an association between PD-L1 expression on CLL cells and adenosine signaling according to sex. Our findings provide a rationale for targeting CD73 in CLL in combination with anti-PD-1/PD-L1 immunotherapies and suggest that sex may contribute to responses to adenosine-targeting agents. Abstract The ecto-nucleotidase CD73 is an important immune checkpoint in tumor immunity that cooperates with CD39 to hydrolyze pro-inflammatory extracellular ATP into immunosuppressive adenosine. While the role of CD73 in immune evasion of solid cancers is well established, its role in leukemia remains unclear. To investigate the role of CD73 in the pathogenesis of chronic lymphocytic leukemia (CLL), Eµ-TCL1 transgenic mice that spontaneously develop CLL were crossed with CD73−/− mice. Disease progression in peripheral blood and spleen, and CLL markers were evaluated by flow cytometry and survival was compared to CD73-proficient Eµ-TCL1 transgenic mice. We observed that CD73 deficiency significantly delayed CLL progression and prolonged survival in Eµ-TCL1 transgenic mice, and was associated with increased accumulation of IFN-γ+ T cells and effector-memory CD8+ T cells. Neutralizing IFN-γ abrogated the survival advantage of CD73-deficient Eµ-TCL1 mice. Intriguingly, the beneficial effects of CD73 deletion were restricted to male mice. In females, CD73 deficiency was uniquely associated with the upregulation of CD39 in normal lymphocytes and sustained high PD-L1 expression on CLL cells. In vitro studies revealed that adenosine signaling via the A2a receptor enhanced PD-L1 expression on Eµ-TCL1-derived CLL cells, and a genomic analysis of human CLL samples found that PD-L1 correlated with adenosine signaling. Our study, thus, identified CD73 as a pro-leukemic immune checkpoint in CLL and uncovered a previously unknown sex bias for the CD73-adenosine pathway.
Collapse
Affiliation(s)
- David Allard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
- Faculté de Pharmacie, l’Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Pavel Chrobak
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Yacine Bareche
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
- Faculté de Pharmacie, l’Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Priscilla Tessier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Marjorie A. Bergeron
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
- Faculté de Pharmacie, l’Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Nathalie A. Johnson
- Department of Medicine, Jewish General Hospital, Montréal, QC H3T 1E2, Canada;
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada; (D.A.); (P.C.); (Y.B.); (B.A.); (P.T.); (M.A.B.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
- Faculté de Pharmacie, l’Université de Montréal, Montréal, QC H3T 1J4, Canada
- Correspondence:
| |
Collapse
|
20
|
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
|
21
|
Regulation of S100As Expression by Inflammatory Cytokines in Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:ijms23136952. [PMID: 35805957 PMCID: PMC9267105 DOI: 10.3390/ijms23136952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The calcium-binding proteins S100A4, S100A8, and S100A9 are upregulated in chronic lymphocytic leukemia (CLL), while the S100A9 promotes NF-κB activity during disease progression. The S100-protein family has been involved in several malignancies as mediators of inflammation and proliferation. The hypothesis of our study is that S100A proteins are mediators in signaling pathways associated with inflammation-induced proliferation, such as NF-κB, PI3K/AKT, and JAK/STAT. The mononuclear cells (MNCs) of CLL were treated with proinflammatory IL-6, anti-inflammatory IL-10 cytokines, inhibitors of JAK1/2, NF-κB, and PI3K signaling pathways, to evaluate S100A4, S100A8, S100A9, and S100A12 expression as well as NF-κB activation by qRT-PCR, immunocytochemistry, and immunoblotting. The quantity of S100A4, S100A8, and S100A9 positive cells (p < 0.05) and their protein expression (p < 0.01) were significantly decreased in MNCs of CLL patients compared to healthy controls. The S100A levels were generally increased in CD19+ cells compared to MNCs of CLL. The S100A4 gene expression was significantly stimulated (p < 0.05) by the inhibition of the PI3K/AKT signaling pathway in MNCs. IL-6 stimulated S100A4 and S100A8 protein expression, prevented by the NF-κB and JAK1/2 inhibitors. In contrast, IL-10 reduced S100A8, S100A9, and S100A12 protein expressions in MNCs of CLL. Moreover, IL-10 inhibited activation of NF-κB signaling (4-fold, p < 0.05). In conclusion, inflammation stimulated the S100A protein expression mediated via the proliferation-related signaling and balanced by the cytokines in CLL.
Collapse
|
22
|
The Tumor Microenvironment-Dependent Transcription Factors AHR and HIF-1α Are Dispensable for Leukemogenesis in the Eµ-TCL1 Mouse Model of Chronic Lymphocytic Leukemia. Cancers (Basel) 2021; 13:cancers13184518. [PMID: 34572746 PMCID: PMC8466120 DOI: 10.3390/cancers13184518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/24/2023] Open
Abstract
Simple Summary Chronic lymphocytic leukemia (CLL) is the most common leukemia in Western countries, mostly affecting the elderly. The survival of leukemic cells depends on multiple soluble factors and on the stimulation of the BCR signaling pathway. Microenvironment-dependent transcription factors also contribute to CLL biology. Here, we generated new transgenic murine conditional knock-out models of CLL to study the role of the two transcription factors HIF-1α and AHR. Unexpectedly, we observed that both factors are dispensable for leukemia development in these models. Abstract Chronic lymphocytic leukemia (CLL) is the most frequent leukemia in the elderly and is characterized by the accumulation of mature B lymphocytes in peripheral blood and primary lymphoid organs. In order to proliferate, leukemic cells are highly dependent on complex interactions with their microenvironment in proliferative niches. Not only soluble factors and BCR stimulation are important for their survival and proliferation, but also the activation of transcription factors through different signaling pathways. The aryl hydrocarbon receptor (AHR) and hypoxia-inducible factor (HIF)-1α are two transcription factors crucial for cancer development, whose activities are dependent on tumor microenvironment conditions, such as the presence of metabolites from the tryptophan pathway and hypoxia, respectively. In this study, we addressed the potential role of AHR and HIF-1α in chronic lymphocytic leukemia (CLL) development in vivo. To this end, we crossed the CLL mouse model Eµ-TCL1 with the corresponding transcription factor-conditional knock-out mice to delete one or both transcription factors in CD19+ B cells only. Despite AHR and HIF-1α being activated in CLL cells, deletion of either or both of them had no impact on CLL progression or survival in vivo, suggesting that these transcription factors are not crucial for leukemogenesis in CLL.
Collapse
|
23
|
Svanberg R, Janum S, Patten PEM, Ramsay AG, Niemann CU. Targeting the tumor microenvironment in chronic lymphocytic leukemia. Haematologica 2021; 106:2312-2324. [PMID: 33882636 PMCID: PMC8409023 DOI: 10.3324/haematol.2020.268037] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
The tumor microenvironment (TME) plays an essential role in the development, growth, and survival of the malignant B-cell clone in chronic lymphocytic leukemia (CLL). Within the proliferation niches of lymph nodes, bone marrow, and secondary lymphoid organs, a variety of phenotypically and functionally altered cell types, including T cells, natural killer cells, monocytes/macrophages, endothelial and mesenchymal stroma cells, provide crucial survival signals, along with CLL-cellinduced suppression of antitumor immune responses. The B-cell receptor pathway plays a pivotal role in mediating the interaction between CLL cells and the TME. However, an increasing number of additional components of the multifactorial TME are being discovered. Although the majority of therapeutic strategies employed in CLL hitherto have focused on targeting the leukemic cells, emerging evidence implies that modulation of microenvironmental cells and CLL-TME interactions by novel therapeutic agents significantly affect their clinical efficacy. Thus, improving our understanding of CLL-TME interactions and how they are affected by current therapeutic agents may improve and guide treatment strategies. Identification of novel TME interactions may also pave the road for the development of novel therapeutic strategies targeting the TME. In this review, we summarize current evidence on the effects of therapeutic agents on cells and interactions within the TME. With a growing demand for improved and personalized treatment options in CLL, this review aims at inspiring future exploration of smart drug combination strategies, translational studies, and novel therapeutic targets in clinical trials.
Collapse
Affiliation(s)
| | - Sine Janum
- Department of Clinical Haemato-oncology, Bartholomew's Hospital, Barts Health Trust, London
| | - Piers E M Patten
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London
| | - Alan G Ramsay
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London
| | | |
Collapse
|
24
|
TCL1A, B Cell Regulation and Tolerance in Renal Transplantation. Cells 2021; 10:cells10061367. [PMID: 34206047 PMCID: PMC8230170 DOI: 10.3390/cells10061367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022] Open
Abstract
Despite much progress in the management of kidney transplantation, the need for life-long immunosuppressive therapies remains a major issue representing many risks for patients. Operational tolerance, defined as allograft acceptance without immunosuppression, has logically been subject to many investigations with the aim of a better understanding of post-transplantation mechanisms and potentially how it would be induced in patients. Among proposed biomarkers, T-cell Leukemia/Lymphoma protein 1A (TCL1A) has been observed as overexpressed in the peripheral blood of operational tolerant patients in several studies. TCL1A expression is restricted to early B cells, also increased in the blood of tolerant patients, and showing regulatory properties, notably through IL-10 secretion for some subsets. TCL1A has first been identified as an oncogene, overexpression of which is associated to the development of T and B cell cancer. TCL1A acts as a coactivator of the serine threonine kinase Akt and through other interactions favoring cell survival, growth, and proliferation. It has also been identified as interacting with others major actors involved in B cells differentiation and regulation, including IL-10 production. Herein, we reviewed known interactions and functions of TCL1A in B cells which could involve its potential role in the set up and maintenance of renal allograft tolerance.
Collapse
|
25
|
Jiménez I, Tazón-Vega B, Abrisqueta P, Nieto JC, Bobillo S, Palacio-García C, Carabia J, Valdés-Mas R, Munuera M, Puigdefàbregas L, Parra G, Esteve-Codina A, Franco-Jarava C, Iacoboni G, Terol MJ, García-Marco JA, Crespo M, Bosch F. Immunological and genetic kinetics from diagnosis to clinical progression in chronic lymphocytic leukemia. Biomark Res 2021; 9:37. [PMID: 34016160 PMCID: PMC8138982 DOI: 10.1186/s40364-021-00290-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mechanisms driving the progression of chronic lymphocytic leukemia (CLL) from its early stages are not fully understood. The acquisition of molecular changes at the time of progression has been observed in a small fraction of patients, suggesting that CLL progression is not mainly driven by dynamic clonal evolution. In order to shed light on mechanisms that lead to CLL progression, we investigated longitudinal changes in both the genetic and immunological scenarios. METHODS We performed genetic and immunological longitudinal analysis using paired primary samples from untreated CLL patients that underwent clinical progression (sampling at diagnosis and progression) and from patients with stable disease (sampling at diagnosis and at long-term asymptomatic follow-up). RESULTS Molecular analysis showed limited and non-recurrent molecular changes at progression, indicating that clonal evolution is not the main driver of clinical progression. Our analysis of the immune kinetics found an increasingly dysfunctional CD8+ T cell compartment in progressing patients that was not observed in those patients that remained asymptomatic. Specifically, terminally exhausted effector CD8+ T cells (T-betdim/-EomeshiPD1hi) accumulated, while the the co-expression of inhibitory receptors (PD1, CD244 and CD160) increased, along with an altered gene expression profile in T cells only in those patients that progressed. In addition, malignant cells from patients at clinical progression showed enhanced capacity to induce exhaustion-related markers in CD8+ T cells ex vivo mainly through a mechanism dependent on soluble factors including IL-10. CONCLUSIONS Altogether, we demonstrate that the interaction with the immune microenvironment plays a key role in clinical progression in CLL, thereby providing a rationale for the use of early immunotherapeutic intervention.
Collapse
Affiliation(s)
- Isabel Jiménez
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Bárbara Tazón-Vega
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Pau Abrisqueta
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Juan C Nieto
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Sabela Bobillo
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Carles Palacio-García
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Júlia Carabia
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | | | - Magdalena Munuera
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Lluís Puigdefàbregas
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Genís Parra
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08003, Barcelona, Spain.,Universitat Pompeu Fabra, 08002, Barcelona, Spain
| | - Anna Esteve-Codina
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08003, Barcelona, Spain.,Universitat Pompeu Fabra, 08002, Barcelona, Spain
| | - Clara Franco-Jarava
- Servei d'Immunologia, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Gloria Iacoboni
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - María José Terol
- Department of Hematology, Clínic University Hospital, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | | | - Marta Crespo
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Francesc Bosch
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain. .,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
26
|
Bozorgmehr N, Okoye I, Oyegbami O, Xu L, Fontaine A, Cox-Kennett N, Larratt LM, Hnatiuk M, Fagarasanu A, Brandwein J, Peters AC, Elahi S. Expanded antigen-experienced CD160 +CD8 +effector T cells exhibit impaired effector functions in chronic lymphocytic leukemia. J Immunother Cancer 2021; 9:jitc-2020-002189. [PMID: 33931471 PMCID: PMC8098955 DOI: 10.1136/jitc-2020-002189] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background T cell exhaustion compromises antitumor immunity, and a sustained elevation of co-inhibitory receptors is a hallmark of T cell exhaustion in solid tumors. Similarly, upregulation of co-inhibitory receptors has been reported in T cells in hematological cancers such as chronic lymphocytic leukemia (CLL). However, the role of CD160, a glycosylphosphatidylinositol-anchored protein, as one of these co-inhibitory receptors has been contradictory in T cell function. Therefore, we decided to elucidate how CD160 expression and/or co-expression with other co-inhibitory receptors influence T cell effector functions in patients with CLL. Methods We studied 56 patients with CLL and 25 age-matched and sex-matched healthy controls in this study. The expression of different co-inhibitory receptors was analyzed in T cells obtained from the peripheral blood or the bone marrow. Also, we quantified the properties of extracellular vesicles (EVs) in the plasma of patients with CLL versus healthy controls. Finally, we measured 29 different cytokines, chemokines or other biomarkers in the plasma specimens of patients with CLL and healthy controls. Results We found that CD160 was the most upregulated co-inhibitory receptor in patients with CLL. Its expression was associated with an exhausted T cell phenotype. CD160+CD8+ T cells were highly antigen-experienced/effector T cells, while CD160+CD4+ T cells were more heterogeneous. In particular, we identified EVs as a source of CD160 in the plasma of patients with CLL that can be taken up by T cells. Moreover, we observed a dominantly proinflammatory cytokine profile in the plasma of patients with CLL. In particular, interleukin-16 (IL-16) was highly elevated and correlated with the advanced clinical stage (Rai). Furthermore, we observed that the incubation of T cells with IL-16 results in the upregulation of CD160. Conclusions Our study provides a novel insight into the influence of CD160 expression/co-expression with other co-inhibitory receptors in T cell effector functions in patients with CLL. Besides, IL-16-mediated upregulation of CD160 expression in T cells highlights the importance of IL-16/CD160 as potential immunotherapy targets in patients with CLL. Therefore, our findings propose a significant role for CD160 in T cell exhaustion in patients with CLL.
Collapse
Affiliation(s)
- Najmeh Bozorgmehr
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Isobel Okoye
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Olaide Oyegbami
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lai Xu
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amelie Fontaine
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nanette Cox-Kennett
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Loree M Larratt
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Hnatiuk
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andrei Fagarasanu
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph Brandwein
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anthea C Peters
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shokrollah Elahi
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada .,Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Baptista MJ, Baskar S, Gaglione EM, Keyvanfar K, Ahn IE, Wiestner A, Sun C. Select Antitumor Cytotoxic CD8 + T Clonotypes Expand in Patients with Chronic Lymphocytic Leukemia Treated with Ibrutinib. Clin Cancer Res 2021; 27:4624-4633. [PMID: 33875521 DOI: 10.1158/1078-0432.ccr-20-4894] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE In chronic lymphocytic leukemia (CLL), the T-cell receptor (TCR) repertoire is skewed and tumor-derived antigens are hypothesized as drivers of oligoclonal expansion. Ibrutinib, a standard treatment for CLL, inhibits not only Bruton tyrosine kinase of the B-cell receptor signaling pathway, but also IL2-inducible kinase of the TCR signaling pathway. T-cell polarization and activation are affected by ibrutinib, but it is unknown whether T cells contribute to clinical response. EXPERIMENTAL DESIGN High-throughput TCRβ sequencing was performed in 77 longitudinal samples from 26 patients with CLL treated with ibrutinib. TCRβ usage in CD4+ and CD8+ T cells and granzyme B expression were assessed by flow cytometric analysis. Antitumor cytotoxicity of T cells expanded with autologous CLL cells or with antigen-independent anti-CD3/CD28/CD137 beads was tested. RESULTS The clonality of the TCR repertoire increased at the time of response. With extended treatment, TCR clonality remained stable in patients with sustained remission and decreased in patients with disease progression. Expanded clonotypes were rarely shared between patients, indicating specificity for private antigens. Flow cytometry demonstrated a predominance of CD8+ cells among expanded clonotypes. Importantly, bulk T cells from responding patients were cytotoxic against autologous CLL cells in vitro and selective depletion of major expanded clonotypes reduced CLL cell killing. CONCLUSIONS In patients with CLL, established T-cell responses directed against tumor are suppressed by disease and reactivated by ibrutinib.See related commentary by Zent, p. 4465.
Collapse
Affiliation(s)
- Maria Joao Baptista
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.,Lymphoid Neoplasms, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | | | - Erika M Gaglione
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Keyvan Keyvanfar
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Inhye E Ahn
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| | - Clare Sun
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
28
|
Immunomodulatory effects of galectin-1 in patients with chronic lymphocytic leukemia. Cent Eur J Immunol 2021; 46:54-62. [PMID: 33897284 PMCID: PMC8056350 DOI: 10.5114/ceji.2021.105246] [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: 01/14/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Galectin-1 (Gal-1) has been implicated in the progression of chronic lymphocytic leukemia (CLL) but also the development of immunodeficiency, which commonly accompany this malignancy. In this in vitro study, we investigated the effects of Gal-1 inhibition in the sera of immunocompromised CLL patients on immunomodulating properties of dendritic cells (DCs). DCs derived from peripheral blood mononuclear cells were treated with a healthy serum, CLL serum as well as the combination of CLL serum and Gal-1 inhibitor (OTX008). Following the treatment, the expression levels of DC maturation markers (CD80, CD83, CD86 and IDO-1) were determined as well as their cytokine profile and the ability to polarize the immune response in co-cultures with CD4+ T cells. After treatment with CLL serum, an increase in interleukin (IL)-10 production was observed in both DC cultures and co-cultures with CD4+ T cells. OTX008 caused a reduction in IL-10 production as well as IL-2, but no significant alteration in the expression of DC maturation markers or T regulatory cell (Treg) frequency was observed. The results of our study suggest that Gal-1 from CLL serum give rise to a specific IL-10+ CD4+ T cell phenotype, other than Treg, that could mediate immunodeficiency development in CLL patients.
Collapse
|
29
|
BTLA/HVEM Axis Induces NK Cell Immunosuppression and Poor Outcome in Chronic Lymphocytic Leukemia. Cancers (Basel) 2021; 13:cancers13081766. [PMID: 33917094 PMCID: PMC8067870 DOI: 10.3390/cancers13081766] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Chronic lymphocytic leukemia (CLL) represents the most frequent B cell malignancy in Western countries and still remains as an incurable disease. Despite recent advances in targeted therapies including ibrutinib, idelalisib or venetoclax, resistance mechanisms have been described and patients develop a progressive immunosuppression. Since immune checkpoint blockade has demonstrated to reinvigorate T and NK cell-mediated anti-tumor responses, the aim of this work was to elucidate whether this immunosuppression relies, at least in part, in BTLA/HVEM axis in patients with CLL. Our results demonstrate that BTLA and HVEM expression is deeply dysregulated on leukemic and NK cells and correlates with poor outcome. Moreover, soluble BTLA levels correlated with adverse cytogenetics and shorter time to treatment. BTLA blockade restored, at least in part, NK cell-mediated responses in patients with CLL. Altogether, our results provide the rationale to further investigate the role of BTLA/HVEM axis in the pathogenesis of CLL. Abstract Chronic lymphocytic leukemia (CLL) is characterized by progressive immunosuppression and diminished cancer immunosurveillance. Immune checkpoint blockade (ICB)-based therapies, a major breakthrough against cancer, have emerged as a powerful tool to reinvigorate antitumor responses. Herein, we analyzed the role of the novel inhibitory checkpoint BTLA and its ligand, HVEM, in the regulation of leukemic and natural killer (NK) cells in CLL. Flow cytometry analyses showed that BTLA expression is upregulated on leukemic cells and NK cells from patients with CLL, whereas HVEM is downregulated only in leukemic cells, especially in patients with advanced Rai-Binet stage. In silico analysis revealed that increased HVEM, but not BTLA, mRNA expression in leukemic cells correlated with diminished overall survival. Further, soluble BTLA (sBTLA) was found to be increased in the sera of patients with CLL and highly correlated with poor prognostic markers and shorter time to treatment. BTLA blockade with an anti-BTLA monoclonal antibody depleted leukemic cells and boosted NK cell-mediated responses ex vivo by increasing their IFN-γ production, cytotoxic capability, and antibody-dependent cytotoxicity (ADCC). In agreement with an inhibitory role of BTLA in NK cells, surface BTLA expression on NK cells was associated with poor outcome in patients with CLL. Overall, this study is the first to bring to light a role of BTLA/HVEM in the suppression of NK cell-mediated immune responses in CLL and its impact on patient’s prognosis, suggesting that BTLA/HVEM axis may be a potential therapeutic target in this disease.
Collapse
|
30
|
Apollonio B, Ioannou N, Papazoglou D, Ramsay AG. Understanding the Immune-Stroma Microenvironment in B Cell Malignancies for Effective Immunotherapy. Front Oncol 2021; 11:626818. [PMID: 33842331 PMCID: PMC8027510 DOI: 10.3389/fonc.2021.626818] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022] Open
Abstract
Cancers, including lymphomas, develop in complex tissue environments where malignant cells actively promote the creation of a pro-tumoral niche that suppresses effective anti-tumor effector T cell responses. Research is revealing that the tumor microenvironment (TME) differs between different types of lymphoma, covering inflamed environments, as exemplified by Hodgkin lymphoma, to non-inflamed TMEs as seen in chronic lymphocytic leukemia (CLL) or diffuse-large B-cell lymphoma (DLBCL). In this review we consider how T cells and interferon-driven inflammatory signaling contribute to the regulation of anti-tumor immune responses, as well as sensitivity to anti-PD-1 immune checkpoint blockade immunotherapy. We discuss tumor intrinsic and extrinsic mechanisms critical to anti-tumor immune responses, as well as sensitivity to immunotherapies, before adding an additional layer of complexity within the TME: the immunoregulatory role of non-hematopoietic stromal cells that co-evolve with tumors. Studying the intricate interactions between the immune-stroma lymphoma TME should help to design next-generation immunotherapies and combination treatment strategies to overcome complex TME-driven immune suppression.
Collapse
Affiliation(s)
- Benedetta Apollonio
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Nikolaos Ioannou
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Despoina Papazoglou
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Alan G Ramsay
- Faculty of Life Sciences & Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
31
|
Interleukin-10 suppression enhances T-cell antitumor immunity and responses to checkpoint blockade in chronic lymphocytic leukemia. Leukemia 2021; 35:3188-3200. [PMID: 33731852 PMCID: PMC8446094 DOI: 10.1038/s41375-021-01217-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
T-cell dysfunction is a hallmark of B-cell Chronic Lymphocytic Leukemia (CLL), where CLL cells downregulate T-cell responses through regulatory molecules including programmed death ligand-1 (PD-L1) and Interleukin-10 (IL-10). Immune checkpoint blockade (ICB) aims to restore T-cell function by preventing the ligation of inhibitory receptors like PD-1. However, most CLL patients do not respond well to this therapy. Thus, we investigated whether IL-10 suppression could enhance antitumor T-cell activity and responses to ICB. Since CLL IL-10 expression depends on Sp1, we utilized a novel, better tolerated analogue of the Sp1 inhibitor mithramycin (MTMox32E) to suppress CLL IL-10. MTMox32E treatment inhibited mouse and human CLL IL-10 production and maintained T-cell effector function in vitro. In the Eμ-Tcl1 mouse model, treatment reduced plasma IL-10 and CLL burden and increased CD8+ T-cell proliferation, effector and memory cell prevalence, and interferon-γ production. When combined with ICB, suppression of IL-10 improved responses to anti-PD-L1 as shown by a 4.5-fold decrease in CLL cell burden compared to anti-PD-L1 alone. Combination therapy also produced more interferon-γ+, cytotoxic effector KLRG1+, and memory CD8+ T-cells, and fewer exhausted T-cells. Since current therapies for CLL do not target IL-10, this provides a novel strategy to improve immunotherapies.
Collapse
|
32
|
Matthijssens F, Sharma ND, Nysus M, Nickl CK, Kang H, Perez DR, Lintermans B, Van Loocke W, Roels J, Peirs S, Demoen L, Pieters T, Reunes L, Lammens T, De Moerloose B, Van Nieuwerburgh F, Deforce DL, Cheung LC, Kotecha RS, Risseeuw MD, Van Calenbergh S, Takarada T, Yoneda Y, van Delft FW, Lock RB, Merkley SD, Chigaev A, Sklar LA, Mullighan CG, Loh ML, Winter SS, Hunger SP, Goossens S, Castillo EF, Ornatowski W, Van Vlierberghe P, Matlawska-Wasowska K. RUNX2 regulates leukemic cell metabolism and chemotaxis in high-risk T cell acute lymphoblastic leukemia. J Clin Invest 2021; 131:141566. [PMID: 33555272 DOI: 10.1172/jci141566] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy with inferior outcome compared with that of B cell ALL. Here, we show that Runt-related transcription factor 2 (RUNX2) was upregulated in high-risk T-ALL with KMT2A rearrangements (KMT2A-R) or an immature immunophenotype. In KMT2A-R cells, we identified RUNX2 as a direct target of the KMT2A chimeras, where it reciprocally bound the KMT2A promoter, establishing a regulatory feed-forward mechanism. Notably, RUNX2 was required for survival of immature and KMT2A-R T-ALL cells in vitro and in vivo. We report direct transcriptional regulation of CXCR4 signaling by RUNX2, thereby promoting chemotaxis, adhesion, and homing to medullary and extramedullary sites. RUNX2 enabled these energy-demanding processes by increasing metabolic activity in T-ALL cells through positive regulation of both glycolysis and oxidative phosphorylation. Concurrently, RUNX2 upregulation increased mitochondrial dynamics and biogenesis in T-ALL cells. Finally, as a proof of concept, we demonstrate that immature and KMT2A-R T-ALL cells were vulnerable to pharmacological targeting of the interaction between RUNX2 and its cofactor CBFβ. In conclusion, we show that RUNX2 acts as a dependency factor in high-risk subtypes of human T-ALL through concomitant regulation of tumor metabolism and leukemic cell migration.
Collapse
Affiliation(s)
- Filip Matthijssens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Nitesh D Sharma
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Monique Nysus
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Christian K Nickl
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Huining Kang
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA.,Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Dominique R Perez
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA.,University of New Mexico Center for Molecular Discovery, Albuquerque, New Mexico, USA
| | - Beatrice Lintermans
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Wouter Van Loocke
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Juliette Roels
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Sofie Peirs
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lisa Demoen
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Tim Pieters
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lindy Reunes
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Tim Lammens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | | | - Dieter L Deforce
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Laurence C Cheung
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
| | - Rishi S Kotecha
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
| | - Martijn Dp Risseeuw
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Laboratory for Medicinal Chemistry, Ghent University, Ghent, Belgium
| | - Serge Van Calenbergh
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Laboratory for Medicinal Chemistry, Ghent University, Ghent, Belgium
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yukio Yoneda
- Department of Pharmacology, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Frederik W van Delft
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Richard B Lock
- Children's Cancer Institute, School of Women's and Children's Health, Lowy Cancer Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Seth D Merkley
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Alexandre Chigaev
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA.,University of New Mexico Center for Molecular Discovery, Albuquerque, New Mexico, USA
| | - Larry A Sklar
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA.,University of New Mexico Center for Molecular Discovery, Albuquerque, New Mexico, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital, UCSF, San Francisco, California, USA
| | - Stuart S Winter
- Cancer and Blood Disorders Program, Children's Minnesota, Minneapolis, Minnesota, USA
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Goossens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Eliseo F Castillo
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | | | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ksenia Matlawska-Wasowska
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
33
|
Hematopoietic versus Solid Cancers and T Cell Dysfunction: Looking for Similarities and Distinctions. Cancers (Basel) 2021; 13:cancers13020284. [PMID: 33466674 PMCID: PMC7828769 DOI: 10.3390/cancers13020284] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Dysfunction of the immune T cell compartment occurs in many hematopoietic as well as solid cancers and hampers successful application of new immunotherapeutic approaches. A complete understanding of T cell dysfunction might improve the outcome of such therapies, but an overview in the various cancers is still lacking. We aim to map areas of similarities and differences in solid versus hematopoietic malignancies, providing a high-level rather than a detailed perspective on T cell dysfunction in those tumors. Abstract Cancer cells escape, suppress and exploit the host immune system to sustain themselves, and the tumor microenvironment (TME) actively dampens T cell function by various mechanisms. Over the last years, new immunotherapeutic approaches, such as adoptive chimeric antigen receptor (CAR) T cell therapy and immune checkpoint inhibitors, have been successfully applied for refractory malignancies that could only be treated in a palliative manner previously. Engaging the anti-tumor activity of the immune system, including CAR T cell therapy to target the CD19 B cell antigen, proved to be effective in acute lymphocytic leukemia. In low-grade hematopoietic B cell malignancies, such as chronic lymphocytic leukemia, clinical outcomes have been tempered by cancer-induced T cell dysfunction characterized in part by a state of metabolic lethargy. In multiple myeloma, novel antigens such as BCMA and CD38 are being explored for CAR T cells. In solid cancers, T cell-based immunotherapies have been applied successfully to melanoma and lung cancers, whereas application in e.g., breast cancer lags behind and is modestly effective as yet. The main hurdles for CAR T cell immunotherapy in solid tumors are the lack of suitable antigens, anatomical inaccessibility, and T cell anergy due to immunosuppressive TME. Given the wide range of success and failure of immunotherapies in various cancer types, it is crucial to comprehend the underlying similarities and distinctions in T cell dysfunction. Hence, this review aims at comparing selected, distinct B cell-derived versus solid cancer types and at describing means by which malignant cells and TME might dampen T cell anti-tumor activity, with special focus on immunometabolism. Drawing a meaningful parallel between the efficacy of immunotherapy and the extent of T cell dysfunction will shed light on areas where we can improve immune function to battle cancer.
Collapse
|
34
|
Tonon S, Martinis E, Pucillo CEM, Mion F. il-10 Gene Locus DNA Methylation in Regulatory B Cells. Methods Mol Biol 2021; 2270:323-339. [PMID: 33479907 DOI: 10.1007/978-1-0716-1237-8_18] [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] [Indexed: 06/12/2023]
Abstract
Epigenetic studies are becoming increasingly common in the immunology field thanks to the support of cutting edge technology and to their potential of providing a large amount of data at the single cell level. Moreover, epigenetic modifications were shown to play a role in autoimmune/inflammatory disorders, paving the way for the possibility of using the results of epigenetic studies for therapeutic purposes. In recent years, epigenetic marks such as DNA methylation, histone modifications and nucleosome positioning were shown to regulate B cell fate and function during an immune response, but very little has been done in the context of one of the most recently discovered B cell subsets, that is regulatory B cells. Although no consensus has yet been found on the identity of these immunosuppressive B cells, the role of the IL-10 cytokine is consolidated, both in the murine and human setting. In this chapter we will focus on the analysis of the methylation profile of a gene of interest and we will specifically describe cloning and pyrosequencing bisulphite sequencing PCR (BSP). Given the specific context, we will provide tips and tricks for the analysis of the il-10 gene locus. Nonetheless, the methods presented are valid for the study of any gene of interest.
Collapse
Affiliation(s)
- Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy.
| | | | | | - Francesca Mion
- Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
35
|
Molecular Remission Using Low-Dose Immunotherapy with Minimal Toxicities for Poor Prognosis IGHV- Unmutated Chronic Lymphocytic Leukemia. Cells 2020; 10:cells10010010. [PMID: 33375215 PMCID: PMC7822209 DOI: 10.3390/cells10010010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) accounts for 10% of hematologic malignancies. CLL is a malignancy of CD5+ B cells and it is characterized by the accumulation of small, mature-appearing neoplastic lymphocytes in the blood, bone marrow, and secondary lymphoid tissues. In the present case, a middle-aged female patient with poor prognosis unmutated IGHV CLL achieved cytogenetic and molecular remission with minimal adverse events following six cycles of low dose recombinant human IL-2 (rIL-2) in combination with low dose targeted venetoclax. Personalized low dose rIL-2 in combination with either lenalidomide or venetoclax mediates natural killer stimulation and is an effective non-toxic immunotherapy administered in the outpatient setting for poor prognosis CLL.
Collapse
|
36
|
Mesaros O, Jimbu L, Neaga A, Popescu C, Berceanu I, Tomuleasa C, Fetica B, Zdrenghea M. Macrophage Polarization in Chronic Lymphocytic Leukemia: Nurse-Like Cells Are the Caretakers of Leukemic Cells. Biomedicines 2020; 8:E516. [PMID: 33228048 PMCID: PMC7699370 DOI: 10.3390/biomedicines8110516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Macrophages are terminally differentiated innate immune cells. Through their activation, they can be polarized towards the pro-inflammatory M1 type or the wound healing-associated, anti-inflammatory M2 type macrophages. In the tumor microenvironment (TME), M2 is the dominant phenotype and these cells are referred to as tumor-associated macrophages (TAMs). TAMs secrete cytokines and chemokines, exerting an antiapoptotic, proliferative and pro-metastatic effect on the tumor cells. TAMs can be found in many cancers, including chronic lymphocytic leukemia (CLL), where they are called nurse-like cells (NLCs). Despite the generally indolent behavior of CLL, the proportion of treatment-refractory patients is significant. As with the majority of cancers, despite significant recent progress, CLL pathogenesis is poorly understood. The emerging role of the TME in nurturing the neoplastic process warrants the investigation of macrophages as a significant pathogenetic element of tumors. In this paper, we review the current knowledge on the role of stromal macrophages in CLL.
Collapse
Affiliation(s)
- Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Alexandra Neaga
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
| | - Cristian Popescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Infectious Diseases, County Emergency Hospital Alba Iulia, 20 Decebal str., 510093 Alba-Iulia, Romania
| | - Iulia Berceanu
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Bogdan Fetica
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| |
Collapse
|
37
|
Gargiulo E, Morande PE, Largeot A, Moussay E, Paggetti J. Diagnostic and Therapeutic Potential of Extracellular Vesicles in B-Cell Malignancies. Front Oncol 2020; 10:580874. [PMID: 33117718 PMCID: PMC7550802 DOI: 10.3389/fonc.2020.580874] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EV), comprising microvesicles and exosomes, are particles released by every cell of an organism, found in all biological fluids, and commonly involved in cell-to-cell communication through the transfer of cargo materials such as miRNA, proteins, and immune-related ligands (e.g., FasL and PD-L1). An important characteristic of EV is that their composition, abundance, and roles are tightly related to the parental cells. This translates into a higher release of characteristic pro-tumor EV by cancer cells that leads to harming signals toward healthy microenvironment cells. In line with this, the key role of tumor-derived EV in cancer progression was demonstrated in multiple studies and is considered a hot topic in the field of oncology. Given their characteristics, tumor-derived EV carry important information concerning the state of tumor cells. This can be used to follow the outset, development, and progression of the neoplasia and to evaluate the design of appropriate therapeutic strategies. In keeping with this, the present brief review will focus on B-cell malignancies and how EV can be used as potential biomarkers to follow disease progression and stage. Furthermore, we will explore several proposed strategies aimed at using biologically engineered EV for treatment (e.g., drug delivery mechanisms) as well as for impairing the biogenesis, release, and internalization of cancer-derived EV, with the final objective to disrupt tumor–microenvironment communication.
Collapse
Affiliation(s)
- Ernesto Gargiulo
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Pablo Elías Morande
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Anne Largeot
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Etienne Moussay
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Jérôme Paggetti
- Tumor-Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
38
|
Bessler H, Leibovitch CM, Djaldetti M. Cytokine Release Ensuing Interaction Between Human Peripheral Blood Mononuclears and Epstein-Barr Virus Transformed B-CLL Cell Line. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2020; 3:99-104. [PMID: 35663257 PMCID: PMC9165579 DOI: 10.36401/jipo-19-33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/14/2020] [Indexed: 06/15/2023]
Abstract
INTRODUCTION B-cell chronic lymphocytic leukemia (B-CLL) is a common form of leukemia affecting mostly elderly individuals. The course of the disease is usually unremarkable, but because it may proceed with impaired immune defense, B-CLL might be complicated with infections and even death. The leukemic microenvironment containing a number of immune cells, mainly lymphocytes and macrophages capable to produce various molecules including inflammatory cytokines, plays an important role in the development and outcome of the disease. We studied the capacity of Epstein-Barr virus (EBV)-transformed B-cell chronic lymphocytic leukemia (B-CLL) cell line (EHEB) cells, an EBV-transformed line established from a B-CLL patient, to affect the production of inflammatory cytokines by human peripheral blood mononuclear cells (PBMC). METHODS PBMC isolated from peripheral blood of healthy donors were incubated either with EHEB cells or with their supernatants and the production of the following cytokines: tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, interferon (IFN)-γ, IL-1ra, and IL-10 were detected using the enzyme-linked immunosorbent assay method. RESULTS Direct contact of PBMC incubated with EHEB cells induced a marked increase of TNFα, IL-1β, IL-6, IFNγ, and IL-10 release by the immune cells. Yet, incubation of PBMC with EHEB cells' supernatant resulted in a mild production of the same cytokines. CONCLUSIONS The noticeable increased production of inflammatory cytokines by PBMC following direct contact with EHEB cells and to a lesser degree with their supernatants implies the existence of an immune dialogue between these two types of cells. The results support the concept that not only leukemic cells, but also peripheral blood mononuclears could serve as a therapeutic target for B-CLL.
Collapse
Affiliation(s)
- Hanna Bessler
- Laboratory for Immunology and Hematology Research, Rabin Medical Center, Hasharon Hospital, Petah-Tiqva, the Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| | - Chiya Moshe Leibovitch
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tiqva, the Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| | - Meir Djaldetti
- Laboratory for Immunology and Hematology Research, Rabin Medical Center, Hasharon Hospital, Petah-Tiqva, the Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| |
Collapse
|
39
|
IL-10-producing regulatory B cells and plasmocytes: Molecular mechanisms and disease relevance. Semin Immunol 2019; 44:101323. [DOI: 10.1016/j.smim.2019.101323] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022]
|
40
|
Tonon S, Mion F, Dong J, Chang HD, Dalla E, Scapini P, Perruolo G, Zanello A, Dugo M, Cassatella MA, Colombo MP, Radbruch A, Tripodo C, Pucillo CE. IL-10-producing B cells are characterized by a specific methylation signature. Eur J Immunol 2019; 49:1213-1225. [PMID: 31034584 DOI: 10.1002/eji.201848025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/13/2019] [Accepted: 04/26/2019] [Indexed: 01/13/2023]
Abstract
Among the family of regulatory B cells, the subset able to produce interleukin-10 (IL-10) is the most studied, yet its biology is still a matter of investigation. The DNA methylation profiling of the il-10 gene locus revealed a novel epigenetic signature characterizing murine B cells ready to respond through IL-10 synthesis: a demethylated region located 4.5 kb from the transcription starting site (TSS), that we named early IL10 regulatory region (eIL10rr). This feature allows to distinguish B cells that are immediately prone and developmentally committed to IL-10 production from those that require a persistent stimulation to exert an IL-10-mediated regulatory function. These late IL-10 producers are instead characterized by a delayed IL10 regulatory region (dIL10rr), a partially demethylated DNA portion located 9 kb upstream from the TSS. A demethylated region was also found in human IL-10-producing B cells and, very interestingly, in some B-cell malignancies, such as chronic lymphocytic leukemia and mantle cell lymphoma, characterized by an immunosuppressive microenvironment. Our findings define murine and human regulatory B cells as an epigenetically controlled functional state of mature B cell subsets and open a new perspective on IL-10 regulation in B cells in homeostasis and disease.
Collapse
Affiliation(s)
- Silvia Tonon
- Department of Medical Area, University of Udine, Udine, Italy
| | - Francesca Mion
- Department of Medical Area, University of Udine, Udine, Italy
| | - Jun Dong
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Hyun-Dong Chang
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Emiliano Dalla
- Department of Medical Area, University of Udine, Udine, Italy
| | - Patrizia Scapini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Giuseppe Perruolo
- Department of Translational Medical Science, University of Napoli ''Federico II'', Napoli, Italy
| | - Andrea Zanello
- Department of Medical Area, University of Udine, Udine, Italy
| | - Matteo Dugo
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Andreas Radbruch
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Carlo E Pucillo
- Department of Medical Area, University of Udine, Udine, Italy
| |
Collapse
|
41
|
Rejection of adoptively transferred Eµ-TCL1 chronic lymphocytic leukemia cells in C57BL/6 substrains or knockout mouse lines. Leukemia 2019; 33:1514-1539. [DOI: 10.1038/s41375-018-0332-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/10/2018] [Accepted: 10/29/2018] [Indexed: 01/14/2023]
|
42
|
Takács F, Tolnai-Kriston C, Hernádfői M, Szabó O, Szalóki G, Szepesi Á, Czeti Á, Matolcsy A, Barna G. The Effect of CD86 Expression on the Proliferation and the Survival of CLL Cells. Pathol Oncol Res 2018; 25:647-652. [PMID: 30406401 DOI: 10.1007/s12253-018-0512-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 12/26/2022]
Abstract
Micro-environment plays important role in the pathogenesis of CLL by providing protective niche for CLL cells. Several molecules play important role in communication between CLL cells and immune cells like CD86.Some of the data suggest that CLL patients with high CD86 level need earlier treatments and cells with higher CD86 expression has higher proliferation rate but the role of CD86 in the survival and proliferation of CLL cells is unclear. We investigated the effect of CD86 expression to CLL cells in 50 peripheral blood and 15 lymph node biopsy samples from CLL patients. Our results showed that the expressions of CD86 increased significantly after 7 day culturing in medium, or in the presence of bone marrow stromal cells (BMSCs). We found positive correlation between CD86 and CD23 expression (p < 0.05), but no correlation with other markers. Furthermore, no correlation were found between the CD86 expression and the proliferation of CLL cells. Analysis of clinical data showed that cases with high CD86 expression had lower level of serum lymphocyte count (p < 0.04) at the time of the diagnosis. CD86 shows multiple appearances in the lymph nodes containing pseudofollicules, but no correlation was found between CD86 positivity, and Ki67 positivity. Our results suggest that the use of CD86 molecule as a proliferation marker for CLL is highly questionable. However, the CD86 molecule may interfere with the immune system of patients with CLL by activating and depleting immune functions. That can be the reason why CD86 positivity may mean worse prognosis.
Collapse
Affiliation(s)
- Ferenc Takács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Csilla Tolnai-Kriston
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Márk Hernádfői
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Orsolya Szabó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Gábor Szalóki
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Ágota Szepesi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Ágnes Czeti
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - András Matolcsy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Gábor Barna
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary.
| |
Collapse
|