1
|
Biernacki MA, Bleakley M. Clinical trials, challenges, and changes in TCR-based therapeutics for hematologic malignancies. Expert Rev Hematol 2025; 18:21-31. [PMID: 39667756 DOI: 10.1080/17474086.2024.2441962] [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: 03/15/2024] [Revised: 08/14/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION T cells engineered to express antigen-specific T cell receptors (TCR; TCR-T) are a promising class of immunotherapeutic for patients with hematologic malignancies. Like chimeric antigen receptor-engineered T cells (CAR-T), TCR-T are cell products with defined specificity and composition. Unlike CAR-T, TCR-T can recognize targets arising both from intracellular and cell surface proteins and leverage the sensitivity of natural TCR signaling machinery. A growing number of TCR-T targeting various antigens in different hematologic malignancies are in early-phase clinical trials, and more are in preclinical development. AREAS COVERED This review covers results from early-phase TCR-T clinical trials for hematologic malignancies. Challenges in the field are reviewed, including identifying optimal targets, engaging CD4+ help for CD8+ T cells, and overcoming tumor-induced suppression; recent innovations to overcome these challenges are also highlighted. EXPERT OPINION In the future, TCR-T's promise for hematologic malignancies will be borne out in later-phase clinical trials and approvals for clinical use. Improved antigen discovery methods will help build the toolbox of targets needed for broadly applicable TCR-T. Rationally designed TCR-T modifications including incorporation of accessory receptors and gene editing will enhance TCR-T function. New hybrid receptors combining features of TCR and CAR will enter the clinic.
Collapse
Affiliation(s)
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Gardano L, Ferreira J, Le Roy C, Ledoux D, Varin-Blank N. The survival grip-how cell adhesion promotes tumor maintenance within the microenvironment. FEBS Lett 2024. [PMID: 39704141 DOI: 10.1002/1873-3468.15074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Cell adhesion is warranted by proteins that are crucial for the maintenance of tissue integrity and homeostasis. Most of these proteins behave as receptors to link adhesion to the control of cell survival and their expression or regulation are often altered in cancers. B-cell malignancies do not evade this principle as they are sustained in relapsed niches by interacting with the microenvironment that includes cells and their secreted factors. Focusing on chronic lymphocytic leukemia and mantle cell lymphoma, this Review delves with the molecules involved in the dialog between the adhesion platforms and signaling pathways known to regulate both cell adhesion and survival. Current therapeutic strategies disrupt adhesive structures and compromise the microenvironment support to tumor cells, rendering them sensitive to immune recognition. The development of organ-on-chip and 3D culture systems, such as spheroids, have revealed the importance of mechanical cues in regulating signaling pathways to organize cell adhesion and survival. All these elements contribute to the elaboration of the crosstalk of lymphoma cells with the microenvironment and the education processes that allow the establishment of the supportive niche.
Collapse
Affiliation(s)
- Laura Gardano
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Jordan Ferreira
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Christine Le Roy
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Dominique Ledoux
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| | - Nadine Varin-Blank
- INSERM U978, Bobigny, France
- UFR SMBH Universite Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
3
|
Ikhlef L, Ratti N, Durand S, Formento R, Daverat H, Boutaud M, Guillou C, Dmytruk N, Gachard N, Cosette P, Jauberteau MO, Gallet PF. Extracellular vesicles from type-2 macrophages increase the survival of chronic lymphocytic leukemia cells ex vivo. Cancer Gene Ther 2024; 31:1164-1176. [PMID: 38918490 PMCID: PMC11327105 DOI: 10.1038/s41417-024-00802-7] [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: 11/17/2023] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
The resistance of Chronic Lymphocytic Leukemia (CLL) B-cells to cell death is mainly attributed to interactions within their microenvironment, where they interact with various types of cells. Within this microenvironment, CLL-B-cells produce and bind cytokines, growth factors, and extracellular vesicles (EVs). In the present study, EVs purified from nurse-like cells and M2-polarized THP1 cell (M2-THP1) cultures were added to CLL-B-cells cultures. EVs were rapidly internalized by B-cells, leading to a decrease in apoptosis (P = 0.0162 and 0.0469, respectively) and an increased proliferation (P = 0.0335 and 0.0109). Additionally, they induced an increase in the resistance of CLL-B-cells to Ibrutinib, the Bruton kinase inhibitor in vitro (P = 0.0344). A transcriptomic analysis showed an increase in the expression of anti-apoptotic gene BCL-2 (P = 0.0286) but not MCL-1 and an increase in the expression of proliferation-inducing gene APRIL (P = 0.0286) following treatment with EVs. Meanwhile, an analysis of apoptotic protein markers revealed increased amounts of IGFBP-2 (P = 0.0338), CD40 (P = 0.0338), p53 (P = 0.0219) and BCL-2 (P = 0.0338). Finally, exploration of EVs protein content by mass spectrometry revealed they carry various proteins involved in known oncogenic pathways and the RNAseq analysis of CLL-B-cells treated or not with NLCs EVs show various differentially expressed genes.
Collapse
Affiliation(s)
- Léa Ikhlef
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Nina Ratti
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | | | - Rémy Formento
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Héloïse Daverat
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Marie Boutaud
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Clément Guillou
- PISSARO Proteomics Platform, Mont-Saint-Aignan Campus, Mont-Saint-Aignan, France
| | - Natalya Dmytruk
- Department of Clinical Hematology, University Hospital of Limoges, Limoges, France
| | - Nathalie Gachard
- Hematology laboratory, UMR CNRS7276/ INSERM 1262, University Hospital of Limoges, Limoges, France
| | - Pascal Cosette
- Polymers, Biopolymers, Surface Laboratory, UMR 6270 CNRS, Normandie University, UNIROUEN, INSA Rouen, Mont-Saint-Aignan, France
- HeRacLeS-PISSARO, INSERM US 51, CNRS UAR 2026, Normandie University, Mont-Saint-Aignan, France
| | - Marie-Odile Jauberteau
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
- Immunology laboratory, University Hospital of Limoges, Limoges, France
| | | |
Collapse
|
4
|
Manivannan MS, Yang X, Patel N, Peters A, Johnston JB, Gibson SB. Lysosome-Disrupting Agents in Combination with Venetoclax Increase Apoptotic Response in Primary Chronic Lymphocytic Leukemia (CLL) Cells Mediated by Lysosomal Cathepsin D Release and Inhibition of Autophagy. Cells 2024; 13:1041. [PMID: 38920669 PMCID: PMC11202145 DOI: 10.3390/cells13121041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Venetoclax and obinutuzumab are becoming frontline therapies for chronic lymphocytic leukemia (CLL) patients. Unfortunately, drug resistance still occurs, and the combination could be immunosuppressive. Lysosomes have previously been identified as a target for obinutuzumab cytotoxicity in CLL cells, but the mechanism remains unclear. In addition, studies have shown that lysosomotropic agents can cause synergistic cell death in vitro when combined with the BTK inhibitor, ibrutinib, in primary CLL cells. This indicates that targeting lysosomes could be a treatment strategy for CLL. In this study, we have shown that obinutuzumab induces lysosome membrane permeabilization (LMP) and cathepsin D release in CLL cells. Inhibition of cathepsins reduced obinutuzumab-induced cell death in CLL cells. We further determined that the lysosomotropic agent siramesine in combination with venetoclax increased cell death in primary CLL cells through an increase in reactive oxygen species (ROS) and cathepsin release. Siramesine treatment also induced synergistic cytotoxicity when combined with venetoclax. Microenvironmental factors IL4 and CD40L or incubation with HS-5 stromal cells failed to significantly protect CLL cells from siramesine- and venetoclax-induced apoptosis. We also found that siramesine treatment inhibited autophagy through reduced autolysosomes. Finally, the autophagy inhibitor chloroquine failed to further increase siramesine-induced cell death. Taken together, lysosome-targeting drugs could be an effective strategy in combination with venetoclax to overcome drug resistance in CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Sulfonamides/pharmacology
- Lysosomes/metabolism
- Lysosomes/drug effects
- Apoptosis/drug effects
- Autophagy/drug effects
- Cathepsin D/metabolism
- Reactive Oxygen Species/metabolism
- Drug Synergism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Madhumita S. Manivannan
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Xiaoyan Yang
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Nirav Patel
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
| | - Anthea Peters
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
- Cross Cancer Institute, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| | - James B. Johnston
- CancerCare Manitoba Research Institute, Hematologist/Oncologist, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Spencer B. Gibson
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada; (M.S.M.); (X.Y.); (N.P.); (A.P.)
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, USA
| |
Collapse
|
5
|
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
|
6
|
Sbrana FV, Fiordi B, Bordini J, Belloni D, Barbaglio F, Russo L, Scarfò L, Ghia P, Scielzo C. PYK2 is overexpressed in chronic lymphocytic leukaemia: A potential new therapeutic target. J Cell Mol Med 2023; 27:576-586. [PMID: 36747338 PMCID: PMC9930416 DOI: 10.1111/jcmm.17688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/08/2023] Open
Abstract
Chronic Lymphocytic Leukaemia (CLL) is the most common adult B-cell leukaemia and despite improvement in patients' outcome, following the use of targeted therapies, it remains incurable. CLL supportive microenvironment plays a key role in both CLL progression and drug resistance through signals that can be sensed by the main components of the focal adhesion complex, such as FAK and PYK2 kinases. Dysregulations of both kinases have been observed in several metastatic cancers, but their role in haematological malignancies is still poorly defined. We characterized FAK and PYK2 expression and observed that PYK2 expression is higher in leukaemic B cells and its overexpression significantly correlates with their malignant transformation. When targeting both FAK and PYK2 with the specific inhibitor defactinib, we observed a dose-response effect on CLL cells viability and survival. In vivo treatment of a CLL mouse model showed a decrease of the leukaemic clone in all the lymphoid organs along with a significant reduction of macrophages and of the spleen weight and size. Our results first define a possible prognostic value for PYK2 in CLL, and show that both FAK and PYK2 might become putative targets for both CLL and its microenvironment (e.g. macrophages), thus paving the way to an innovative therapeutic strategy.
Collapse
Affiliation(s)
- Francesca Vittoria Sbrana
- Malignant B cells biology and 3D modelling Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Benedetta Fiordi
- Malignant B cells biology and 3D modelling Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
- School of MedicineUniversità Vita‐Salute San RaffaeleMilanItaly
| | - Jessica Bordini
- B‐cell neoplasia Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Daniela Belloni
- B‐cell neoplasia Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Federica Barbaglio
- Malignant B cells biology and 3D modelling Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Luca Russo
- Malignant B cells biology and 3D modelling Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Lydia Scarfò
- School of MedicineUniversità Vita‐Salute San RaffaeleMilanItaly
- B‐cell neoplasia Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Paolo Ghia
- School of MedicineUniversità Vita‐Salute San RaffaeleMilanItaly
- B‐cell neoplasia Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| | - Cristina Scielzo
- Malignant B cells biology and 3D modelling Unit, Division of Experimental OncologyIRCCS Ospedale San RaffaeleMilanItaly
| |
Collapse
|
7
|
Braham MV, van Binnendijk RS, Buisman AMM, Mebius RE, de Wit J, van Els CA. A synthetic human 3D in vitro lymphoid model enhancing B-cell survival and functional differentiation. iScience 2022; 26:105741. [PMID: 36590159 PMCID: PMC9794978 DOI: 10.1016/j.isci.2022.105741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 10/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
To investigate B-cell differentiation and maturation occurring in the germinal center (GC) using in vitro culture systems, key factors and interactions of the GC reaction need to be accurately simulated. This study aims at improving in vitro GC simulation using 3D culture techniques. Human B-cells were incorporated into PEG-4MAL hydrogels, to create a synthetic extracellular matrix, supported by CD40L cells, human tonsil-derived lymphoid stromal cells, and cytokines. The differentiation and antibody production of CD19+B-cells was best supported in a 5.0%-PEG-4MAL, 2.0 mM-RGD-peptide composition. The 3D culture significantly increased plasmablast and plasma cell numbers as well as antibody production, with less B-cell death compared to 2D cultures. Class switching of naive CD19+IgD+B-cells toward IgG+ and IgA+B-cells was observed. The formation of large B-cell clusters indicates the formation of GC-like structures. In conclusion, a well-characterized and controllable hydrogel-based human 3D lymphoid model is presented that supports enhanced B-cell survival, proliferation, differentiation, and antibody production.
Collapse
Affiliation(s)
- Maaike V.J. Braham
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Rob S. van Binnendijk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Anne-Marie M. Buisman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Reina E. Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands,Corresponding author
| | - Cécile A.C.M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands,Corresponding author
| |
Collapse
|
8
|
Mehrpouri M. The contributory roles of the CXCL12/CXCR4/CXCR7 axis in normal and malignant hematopoiesis: A possible therapeutic target in hematologic malignancies. Eur J Pharmacol 2022; 920:174831. [PMID: 35183534 DOI: 10.1016/j.ejphar.2022.174831] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/03/2022]
|
9
|
Handl S, von Heydebrand F, Voelkl S, Oostendorp RAJ, Wilke J, Kremer AN, Mackensen A, Lutzny-Geier G. Immune modulatory effects of Idelalisib in stromal cells of chronic lymphocytic leukemia. Leuk Lymphoma 2021; 62:2679-2689. [PMID: 33999745 DOI: 10.1080/10428194.2021.1927019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Molecular targets of tyrosine kinase inhibitors are not restricted to the B-cell compartment but also regulate functions in the tumor microenvironment. Increasing evidence suggests that B-cell receptor-associated kinases like protein kinase C (PKC)-β is essential for the formation of a microenvironment supporting leukemic growth. Here we describe the effect of Idelalisib on the PKCβ/NF-κB and Notch pathway in stromal cells upon contact to primary chronic lymphocytic leukemia cells (CLL). There is no Idelalisib-dependent regulation of the Notch expression in stromal cells, whereas Idelalisib induces PKCβ expression and activates the canonical NF-κB pathway. Idelalisib deregulates important immune-modulatory proteins in activated stromal cells, which might provoke the patient's side effects. Additionally, we established a 3D-stroma/leukemia model, that can give us a more defined look into the communication between tumor and stromal cells than standard cell cultures. This opens up the possibility to improve therapies, especially in the context of minimal-residual disease.
Collapse
Affiliation(s)
- Sarah Handl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Franziska von Heydebrand
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Simon Voelkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert A J Oostendorp
- Clinic and Polyclinic for Internal Medicine III: Hematology and Oncology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Jochen Wilke
- Practice for Oncology and Hematology, Fürth, Germany
| | - Anita N Kremer
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gloria Lutzny-Geier
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
10
|
Arruga F, Serra S, Vitale N, Guerra G, Papait A, Baffour Gyau B, Tito F, Efremov D, Vaisitti T, Deaglio S. Targeting of the A2A adenosine receptor counteracts immunosuppression in vivo in a mouse model of chronic lymphocytic leukemia. Haematologica 2021; 106:1343-1353. [PMID: 32299906 PMCID: PMC8094100 DOI: 10.3324/haematol.2019.242016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Indexed: 11/09/2022] Open
Abstract
Tumor immunosuppression is a major cause for treatment failure and disease relapse, both in solid tumors and leukemia. Local hypoxia is among the conditions that cause immunosuppression, acting at least in part through the upregulation of extracellular adenosine levels, which potently suppress T cell responses and skew macrophages towards an M2 phenotype. Hence, there is intense investigation to identify drugs that target this axis. By using the TCL1 adoptive transfer CLL mouse model, we show that adenosine production and signaling are upregulated in the hypoxic lymphoid niches, where intense colonization of leukemic cells occurs. This leads to a progressive remodeling of the immune system towards tolerance, with expansion of T regulatory cells (Tregs), loss of CD8+ T cell cytotoxicity and differentiation of murine macrophages towards the patrolling (M2-like) subset. In vivo administration of SCH58261, an inhibitor the A2A adenosine receptor, re-awakens T cell responses, while limiting Tregs expansion, and re-polarizes monocytes towards the inflammatory (M1-like) phenotype. These results show for the first time the in vivo contribution of adenosine signaling to immune tolerance in CLL, and the translational implication of drugs interrupting this pathway. Although the effects of SCH58261 on leukemic cells are limited, interfering with adenosine signaling may represent an appealing strategy for combination-based therapeutic approaches.
Collapse
Affiliation(s)
- Francesca Arruga
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Sara Serra
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Nicoletta Vitale
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulia Guerra
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Andrea Papait
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Benjamin Baffour Gyau
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Francesco Tito
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Tiziana Vaisitti
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Lab of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
11
|
Co-expression network-based identification of biomarkers correlated with the lymph node metastasis of patients with head and neck squamous cell carcinoma. Biosci Rep 2021; 40:222104. [PMID: 32076707 PMCID: PMC7033310 DOI: 10.1042/bsr20194067] [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: 11/29/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 01/07/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is ranked as one of the most frequent malignancies worldwide with a high risk of lymph node metastasis, which serves as a main reason for cancer deaths. Identification of the potential biomarkers for lymph node metastasis in HNSCC patients may contribute to personalized treatment and better therapeutic effect. In the present study, GSE30788 microarray data and corresponding clinical parameters were downloaded from Gene Expression Omnibus (GEO) and Weighted Gene Co-expression Network Analysis (WGCNA) was performed to investigate significant modules associated with clinical traits. As a result, the genes in the blue module were determined as candidate genes related with HNSCC lymph node metastasis and ten hub genes were selected from the PPI network. Further analysis validated the close associations of hub gene expression with lymph node metastasis of HNSCC patients. Furthermore, survival analysis suggested the level of Loricrin (LOR) was statistically significantly associated with the disease-free survival of HNSCC patients, indicating the potential of utilizing it as prognosis predictor. Overall, our study conducted a co-expression network-based analysis to investigate significant genes underlying HNSCC metastasis, providing promising biomarkers and therapeutic targets.
Collapse
|
12
|
Svozilová H, Plichta Z, Proks V, Studená R, Baloun J, Doubek M, Pospíšilová Š, Horák D. RGDS-Modified Superporous Poly(2-Hydroxyethyl Methacrylate)-Based Scaffolds as 3D In Vitro Leukemia Model. Int J Mol Sci 2021; 22:ijms22052376. [PMID: 33673496 PMCID: PMC7956824 DOI: 10.3390/ijms22052376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/05/2023] Open
Abstract
Superporous poly(2-hydroxyethyl methacrylate-co-2-aminoethyl methacrylate) (P(HEMA-AEMA)) hydrogel scaffolds are designed for in vitro 3D culturing of leukemic B cells. Hydrogel porosity, which influences cell functions and growth, is introduced by adding ammonium oxalate needle-like crystals in the polymerization mixture. To improve cell vitality, cell-adhesive Arg-Gly-Asp-Ser (RGDS) peptide is immobilized on the N-(γ-maleimidobutyryloxy)succinimide-activated P(HEMA-AEMA) hydrogels via reaction of SH with maleimide groups. This modification is especially suitable for the survival of primary chronic lymphocytic leukemia cells (B-CLLs) in 3D cell culture. No other tested stimuli (interleukin-4, CD40 ligand, or shaking) can further improve B-CLL survival or metabolic activity. Both unmodified and RGDS-modified P(HEMA-AEMA) scaffolds serve as a long-term (70 days) 3D culture platforms for HS-5 and M2-10B4 bone marrow stromal cell lines and MEC-1 and HG-3 B-CLL cell lines, although the adherent cells retain their physiological morphologies, preferably on RGDS-modified hydrogels. Moreover, the porosity of hydrogels allows direct cell lysis, followed by efficient DNA isolation from the 3D-cultured cells. P(HEMA-AEMA)-RGDS thus serves as a suitable 3D in vitro leukemia model that enables molecular and metabolic assays and allows imaging of cell morphology, interactions, and migration by confocal microscopy. Such applications can prospectively assist in testing of drugs to treat this frequently recurring or refractory cancer.
Collapse
Affiliation(s)
- Hana Svozilová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
- Department of Internal Medicine—Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Zdeněk Plichta
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic; (Z.P.); (V.P.)
| | - Vladimír Proks
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic; (Z.P.); (V.P.)
| | - Radana Studená
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
| | - Jiří Baloun
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
| | - Michael Doubek
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
- Department of Internal Medicine—Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Šárka Pospíšilová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; (H.S.); (R.S.); (J.B.); (M.D.); (Š.P.)
- Department of Internal Medicine—Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic; (Z.P.); (V.P.)
- Correspondence: ; Tel.: +420-296-809-260
| |
Collapse
|
13
|
von Heydebrand F, Fuchs M, Kunz M, Voelkl S, Kremer AN, Oostendorp RAJ, Wilke J, Leitges M, Egle A, Mackensen A, Lutzny-Geier G. Protein kinase C-β-dependent changes in the glucose metabolism of bone marrow stromal cells of chronic lymphocytic leukemia. STEM CELLS (DAYTON, OHIO) 2021; 39:819-830. [PMID: 33539629 DOI: 10.1002/stem.3352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/15/2021] [Indexed: 11/10/2022]
Abstract
Survival of chronic lymphocytic leukemia (CLL) cells critically depends on the support of an adapted and therefore appropriate tumor microenvironment. Increasing evidence suggests that B-cell receptor-associated kinases such as protein kinase C-β (PKCβ) or Lyn kinase are essential for the formation of a microenvironment supporting leukemic growth. Here, we describe the impact of PKCβ on the glucose metabolism in bone marrow stromal cells (BMSC) upon CLL contact. BMSC get activated by CLL contact expressing stromal PKCβ that diminishes mitochondrial stress and apoptosis in CLL cells by stimulating glucose uptake. In BMSC, the upregulation of PKCβ results in increased mitochondrial depolarization and leads to a metabolic switch toward oxidative phosphorylation. In addition, PKCβ-deficient BMSC regulates the expression of Hnf1 promoting stromal insulin signaling after CLL contact. Our data suggest that targeting PKCβ and the glucose metabolism of the leukemic niche could be a potential therapeutic strategy to overcome stroma-mediated drug resistance.
Collapse
Affiliation(s)
- Franziska von Heydebrand
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maximilian Fuchs
- Department of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Meik Kunz
- Department of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Simon Voelkl
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anita N Kremer
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert A J Oostendorp
- Clinic and Polyclinic for Internal Medicine III: Hematology and Oncology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Jochen Wilke
- Practice for Oncology and Hematology, Fürth, Germany
| | - Michael Leitges
- Faculty of Medicine, Division of BioMedical Sciences, Craig L. Dobbin Genetics Research Centre, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Alexander Egle
- IIIrd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) with Laboratory of Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | - Andreas Mackensen
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gloria Lutzny-Geier
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
14
|
Zarobkiewicz M, Kowalska W, Chocholska S, Tomczak W, Szymańska A, Morawska I, Wojciechowska A, Bojarska-Junak A. High M-MDSC Percentage as a Negative Prognostic Factor in Chronic Lymphocytic Leukaemia. Cancers (Basel) 2020; 12:2614. [PMID: 32937740 PMCID: PMC7563618 DOI: 10.3390/cancers12092614] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/29/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
In the current study, we analysed the role and prognostic value of myeloid-derived suppressor cells (MDSC) in chronic lymphocytic leukaemia (CLL). The frequency of circulating monocytic MDSC (M-MDSC; defined as CD14+CD11b+CD15-HLA-DR-/low cells) was assessed in correlation with clinical and laboratory parameters characterising the disease activity and patient immune status. Samples of peripheral blood from untreated CLL patients and healthy volunteers were stained with monoclonal antibodies for flow cytometry analysis. CLL patients with M-MDSC percentages above 9.35% (according to the receiver operating characteristic (ROC) analysis) had a shorter time-to-treatment and shorter survival time than the group with a lower percentage of M-MDSC. The M-MDSC percentage was higher in patients with adverse prognostic factors (i.e., 17p and 11q deletion and CD38 and ZAP-70 expression). A high M-MDSC percentage was linked to significantly lower expression of the CD3ζ in T cells. Furthermore, an analysis of immune regulatory molecules (arginase 1 (ARG1), nitric oxide synthase (NOS2), indoleamine 2,3-dioxygenase (IDO), transforming growth factor beta (TGF-β), and interleukin (IL)-10) was performed. By the means of flow cytometry and RT-qPCR, we showed an overexpression of three of them in M-MDSC of CLL patients. M-MDSC cells seem to be an important factor in the immunosuppressive microenvironment of CLL and seem to be a good and novel prognostic factor.
Collapse
Affiliation(s)
- Michał Zarobkiewicz
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (W.K.); (I.M.)
| | - Wioleta Kowalska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (W.K.); (I.M.)
| | - Sylwia Chocholska
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-080 Lublin, Poland; (S.C.); (W.T.)
| | - Waldemar Tomczak
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-080 Lublin, Poland; (S.C.); (W.T.)
| | - Agata Szymańska
- Department of Clinical Transplantology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Izabela Morawska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (W.K.); (I.M.)
| | | | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (W.K.); (I.M.)
| |
Collapse
|
15
|
Rendeiro AF, Krausgruber T, Fortelny N, Zhao F, Penz T, Farlik M, Schuster LC, Nemc A, Tasnády S, Réti M, Mátrai Z, Alpár D, Bödör C, Schmidl C, Bock C. Chromatin mapping and single-cell immune profiling define the temporal dynamics of ibrutinib response in CLL. Nat Commun 2020; 11:577. [PMID: 31996669 PMCID: PMC6989523 DOI: 10.1038/s41467-019-14081-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/11/2019] [Indexed: 01/10/2023] Open
Abstract
The Bruton tyrosine kinase (BTK) inhibitor ibrutinib provides effective treatment for patients with chronic lymphocytic leukemia (CLL), despite extensive heterogeneity in this disease. To define the underlining regulatory dynamics, we analyze high-resolution time courses of ibrutinib treatment in patients with CLL, combining immune-phenotyping, single-cell transcriptome profiling, and chromatin mapping. We identify a consistent regulatory program starting with a sharp decrease of NF-κB binding in CLL cells, which is followed by reduced activity of lineage-defining transcription factors, erosion of CLL cell identity, and acquisition of a quiescence-like gene signature. We observe patient-to-patient variation in the speed of execution of this program, which we exploit to predict patient-specific dynamics in the response to ibrutinib based on the pre-treatment patient samples. In aggregate, our study describes time-dependent cellular, molecular, and regulatory effects for therapeutic inhibition of B cell receptor signaling in CLL, and it establishes a broadly applicable method for epigenome/transcriptome-based treatment monitoring. Ibrutinib, a Bruton tyrosine kinase inhibitor, provides effective treatment for chronic lymphocytic leukemia (CLL). Here, the authors describe time-dependent molecular changes to malignant cells and to the immune system in patients undergoing ibrutinib therapy, with can be used for therapy monitoring.
Collapse
Affiliation(s)
- André F Rendeiro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nikolaus Fortelny
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Fangwen Zhao
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Linda C Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Amelie Nemc
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Szabolcs Tasnády
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Marienn Réti
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Zoltán Mátrai
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Donát Alpár
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Christian Schmidl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria. .,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria. .,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
16
|
Borgovan T, Crawford L, Nwizu C, Quesenberry P. Stem cells and extracellular vesicles: biological regulators of physiology and disease. Am J Physiol Cell Physiol 2019; 317:C155-C166. [PMID: 30917031 DOI: 10.1152/ajpcell.00017.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many different subpopulations of subcellular extracellular vesicles (EVs) have been described. EVs are released from all cell types and have been shown to regulate normal physiological homeostasis, as well as pathological states by influencing cell proliferation, differentiation, organ homing, injury and recovery, as well as disease progression. In this review, we focus on the bidirectional actions of vesicles from normal and diseased cells on normal or leukemic target cells; and on the leukemic microenvironment as a whole. EVs from human bone marrow mesenchymal stem cells (MSC) can have a healing effect, reversing the malignant phenotype in prostate and colorectal cancer, as well as mitigating radiation damage to marrow. The role of EVs in leukemia and their bimodal cross talk with the encompassing microenvironment remains to be fully characterized. This may provide insight for clinical advances via the application of EVs as potential therapy and the employment of statistical and machine learning models to capture the pleiotropic effects EVs endow to a dynamic microenvironment, possibly allowing for precise therapeutic intervention.
Collapse
Affiliation(s)
- Theodor Borgovan
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University , Providence, Rhode Island
| | - Lorin Crawford
- School of Public Health, Center for Computational Molecular Biology, Brown University , Providence, Rhode Island
| | - Chibuikem Nwizu
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University , Providence, Rhode Island
| | - Peter Quesenberry
- Division of Hematology and Oncology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University , Providence, Rhode Island
| |
Collapse
|
17
|
Steurer M, Montillo M, Scarfò L, Mauro FR, Andel J, Wildner S, Trentin L, Janssens A, Burgstaller S, Frömming A, Dümmler T, Riecke K, Baumann M, Beyer D, Vauléon S, Ghia P, Foà R, Caligaris-Cappio F, Gobbi M. Olaptesed pegol (NOX-A12) with bendamustine and rituximab: a phase IIa study in patients with relapsed/refractory chronic lymphocytic leukemia. Haematologica 2019; 104:2053-2060. [PMID: 31097627 PMCID: PMC6886437 DOI: 10.3324/haematol.2018.205930] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 05/09/2019] [Indexed: 01/04/2023] Open
Abstract
Olaptesed pegol (NOX-A12) is a pegylated structured L-oligoribonucleotide that binds and neutralizes CXCL12, a chemokine tightly regulating the life cycle of chronic lymphocytic leukemia cells. The resulting inhibition of CXCR4 and CXCR7 signaling reduces the protective activity of the bone marrow and lymph node microenvironment. CXCL12 inhibition mobilizes chronic lymphocytic leukemia cells into the circulation and prevents their homing into the protective niches. In this phase I/II study, 28 patients with relapsed/refractory chronic lymphocytic leukemia were treated with olaptesed pegol in combination with bendamustine and rituximab. Combination treatment was preceded by single escalating pilot doses of olaptesed pegol in the first ten patients for evaluation of safety and pharmacokinetics. Peak concentrations and systemic exposure of olaptesed pegol were dose-linear; plasma elimination was monophasic with a 53.2 h half-life. A rapid increase in circulating chronic lymphocytic leukemia cells was observed already 1 h after administration of olaptesed pegol and lasted for at least 72 h. Single-agent treatment was well tolerated and no dose-limiting toxicity was observed. The combination regimen yielded an overall response rate of 86%, with 11% of patients achieving a complete response and 75% a partial response. Notably, all ten high-risk patients, including four with a 17p deletion, responded to treatment. The median progression-free survival was 15.4 (95% confidence interval: 12.2, 26.2) months while the median overall survival was not reached with >80% of patients alive after a median follow-up of 28 months. Olaptesed pegol was well tolerated and did not result in additional toxicity when combined with bendamustine and rituximab (ClinicalTrials.gov identifier: NCT01486797). Further clinical development of this novel CXCL12 inhibitor is thus warranted.
Collapse
Affiliation(s)
- Michael Steurer
- Division of Hematology and Oncology, Innsbruck Medical University, Innsbruck, Austria
| | - Marco Montillo
- Department of Hematology, Niguarda Cancer Center, Niguarda Hospital, Milan, Italy
| | - Lydia Scarfò
- Università Vita-Salute San Raffaele and IRCCS Istituto Scientifico San Raffaele, Milan, Italy
| | - Francesca R Mauro
- Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | | | - Sophie Wildner
- Division of Hematology and Oncology, Innsbruck Medical University, Innsbruck, Austria
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padua, Padua, Italy
| | - Ann Janssens
- Department of Hematology, Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - Sonja Burgstaller
- Department of Internal Medicine IV, Wels-Grieskirchen Hospital, Wels, Austria
| | | | - Thomas Dümmler
- NOXXON Pharma, Berlin, Germany.,current affiliation: Mologen AG, Berlin, Germany and
| | | | - Matthias Baumann
- NOXXON Pharma, Berlin, Germany.,current affiliation: Mologen AG, Berlin, Germany and
| | | | | | - Paolo Ghia
- Università Vita-Salute San Raffaele and IRCCS Istituto Scientifico San Raffaele, Milan, Italy
| | - Robin Foà
- Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | | | - Marco Gobbi
- Haematology Clinic, Department of Internal Medicine, University of Genoa, and Ospedale Policlinico S. Martino, Clinica Ematologica, Genoa, Italy
| |
Collapse
|
18
|
Shachar I, Barak A, Lewinsky H, Sever L, Radomir L. SLAMF receptors on normal and malignant B cells. Clin Immunol 2018; 204:23-30. [PMID: 30448442 DOI: 10.1016/j.clim.2018.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
Abstract
The Signaling Lymphocyte Activation Molecule family (SLAMF) is a collection of nine surface receptors expressed mainly on hematopoietic cells, and was found to modulate the behavior of immune cells. SLAMF receptors are expressed on B cells in health and disease. Each SLAM receptor has a unique differential expression pattern during the development and activation of B cells. Furthermore, recent findings have revealed a principal role for this family of receptors in B cell malignancies, emphasizing their importance in the control of malignant cell survival, cell to cell communication within the tumor microenvironment, retention in the supporting niches and regulation of T cell anti-tumor response. This review summarizes the latest studies regarding SLAMF expression and behavior in B cells and in B cell pathologies, and discusses the therapeutic potential of these receptors.
Collapse
Affiliation(s)
- Idit Shachar
- Department of Immunology, Weizmann Institute of Science, Israel.
| | - Avital Barak
- Department of Immunology, Weizmann Institute of Science, Israel
| | - Hadas Lewinsky
- Department of Immunology, Weizmann Institute of Science, Israel
| | - Lital Sever
- Department of Immunology, Weizmann Institute of Science, Israel
| | - Lihi Radomir
- Department of Immunology, Weizmann Institute of Science, Israel
| |
Collapse
|
19
|
Gordiienko I, Shlapatska L, Kovalevska L, Sidorenko SP. SLAMF1/CD150 in hematologic malignancies: Silent marker or active player? Clin Immunol 2018; 204:14-22. [PMID: 30616923 DOI: 10.1016/j.clim.2018.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
SLAMF1/CD150 receptor is a founder of signaling lymphocyte activation molecule (SLAM) family of cell-surface receptors. It is widely expressed on cells within hematopoietic system. In hematologic malignancies CD150 cell surface expression is restricted to cutaneous T-cell lymphomas, few types of B-cell non-Hodgkin's lymphoma, near half of cases of chronic lymphocytic leukemia, Hodgkin's lymphoma, and multiple myeloma. Differential expression among various types of hematological malignancies allows considering CD150 as diagnostical and potential prognostic marker. Moreover, CD150 may be a target for antibody-based or measles virus oncolytic therapy. Due to CD150 signaling properties it is involved in regulation of malignant cell fate decision and tumor microenvironment in Hodgkin's lymphoma and chronic lymphocytic leukemia. This review summarizes evidence for the important role of CD150 in pathogenesis of hematologic malignancies.
Collapse
Affiliation(s)
- Inna Gordiienko
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine.
| | - Larysa Shlapatska
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Larysa Kovalevska
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Svetlana P Sidorenko
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
20
|
Peled A, Klein S, Beider K, Burger JA, Abraham M. Role of CXCL12 and CXCR4 in the pathogenesis of hematological malignancies. Cytokine 2018; 109:11-16. [PMID: 29903571 DOI: 10.1016/j.cyto.2018.02.020] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/18/2018] [Accepted: 02/20/2018] [Indexed: 12/25/2022]
Abstract
The chemokine receptor CXCR4 and its ligand stromal cell-derived factor-1 (SDF-1/CXCL12) are important players in the cross-talk among lymphoma, myeloma and leukemia cells and their microenvironments. In hematological malignancies and solid tumors, the overexpression of CXCR4 on the cell surface has been shown to be responsible for disease progression, increasing tumor cell survival and chemoresistance and metastasis to organs with high CXCL12 levels (e.g., lymph nodes and bone marrow (BM)). Furthermore, the overexpression of CXCR4 has been found to have prognostic significance for disease progression in many type of tumors including lymphoma, leukemia, glioma, and prostate, breast, colorectal, renal, and hepatocellular carcinomas. In leukemia, CXCR4 expression granted leukemic blasts a higher capacity to seed into BM niches, thereby protecting leukemic cells from chemotherapy-induced apoptosis, and was correlated with shorter disease-free survival. In contrast, neutralizing the interaction of CXCL12/CXCR4 with a variety of antagonists induced apoptosis and differentiation and increased the chemosensitivity of lymphoma, myeloma, and leukemia cells. The role of CXCL12 and CXCR4 in the pathogenesis of hematological malignancies and the clinical therapeutic potential of CXCR4 antagonists in these diseases is discussed.
Collapse
MESH Headings
- Apoptosis/immunology
- Cell Survival/physiology
- Chemokine CXCL12/metabolism
- Disease Progression
- Hematologic Neoplasms/drug therapy
- Hematologic Neoplasms/pathology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Multiple Myeloma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Prognosis
- Receptors, CXCR4/metabolism
- Tumor Microenvironment/physiology
Collapse
Affiliation(s)
- Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, P.O.B 12000, Jerusalem 91120, Israel.
| | - Shiri Klein
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, P.O.B 12000, Jerusalem 91120, Israel
| | - Katia Beider
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Israel
| | - Jan A Burger
- Department of Leukemia, The University of Texas Houston, TX, USA
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, P.O.B 12000, Jerusalem 91120, Israel
| |
Collapse
|
21
|
Insel PA, Sriram K, Wiley SZ, Wilderman A, Katakia T, McCann T, Yokouchi H, Zhang L, Corriden R, Liu D, Feigin ME, French RP, Lowy AM, Murray F. GPCRomics: GPCR Expression in Cancer Cells and Tumors Identifies New, Potential Biomarkers and Therapeutic Targets. Front Pharmacol 2018; 9:431. [PMID: 29872392 PMCID: PMC5972277 DOI: 10.3389/fphar.2018.00431] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/12/2018] [Indexed: 12/16/2022] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of targets for approved drugs, are rarely targeted for cancer treatment, except for certain endocrine and hormone-responsive tumors. Limited knowledge regarding GPCR expression in cancer cells likely has contributed to this lack of use of GPCR-targeted drugs as cancer therapeutics. We thus undertook GPCRomic studies to define the expression of endoGPCRs (which respond to endogenous molecules such as hormones, neurotransmitters and metabolites) in multiple types of cancer cells. Using TaqMan qPCR arrays to quantify the mRNA expression of ∼340 such GPCRs, we found that human chronic lymphocytic leukemia (CLL) cells/stromal cells associated with CLL, breast cancer cell lines, colon cancer cell lines, pancreatic ductal adenocarcinoma (PDAC) cells, cancer associated fibroblasts (CAFs), and PDAC tumors express 50 to >100 GPCRs, including many orphan GPCRs (which lack known physiologic agonists). Limited prior data exist regarding the expression or function of most of the highly expressed GPCRs in these cancer cells and tumors. Independent results from public cancer gene expression databases confirm the expression of such GPCRs. We propose that highly expressed GPCRs in cancer cells (for example, GPRC5A in PDAC and colon cancer cells and GPR68 in PDAC CAFs) may contribute to the malignant phenotype, serve as biomarkers and/or may be novel therapeutic targets for the treatment of cancer.
Collapse
Affiliation(s)
- Paul A. Insel
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Shu Z. Wiley
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Andrea Wilderman
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Trishna Katakia
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Thalia McCann
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Hiroshi Yokouchi
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Lingzhi Zhang
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Ross Corriden
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Dongling Liu
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Michael E. Feigin
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Randall P. French
- Department of Surgery, University of California, San Diego, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Andrew M. Lowy
- Department of Surgery, University of California, San Diego, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Fiona Murray
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
22
|
A retinoic acid-dependent stroma-leukemia crosstalk promotes chronic lymphocytic leukemia progression. Nat Commun 2018; 9:1787. [PMID: 29725010 PMCID: PMC5934403 DOI: 10.1038/s41467-018-04150-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/14/2018] [Indexed: 12/19/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), the non-hematopoietic stromal microenvironment plays a critical role in promoting tumor cell recruitment, activation, survival, and expansion. However, the nature of the stromal cells and molecular pathways involved remain largely unknown. Here, we demonstrate that leukemic B lymphocytes induce the activation of retinoid acid synthesis and signaling in the microenvironment. Inhibition of RA-signaling in stromal cells causes deregulation of genes associated with adhesion, tissue organization and chemokine secretion including the B-cell chemokine CXCL13. Notably, reducing retinoic acid precursors from the diet or inhibiting RA-signaling through retinoid-antagonist therapy prolong survival by preventing dissemination of leukemia cells into lymphoid tissues. Furthermore, mouse and human leukemia cells could be distinguished from normal B-cells by their increased expression of Rarγ2 and RXRα, respectively. These findings establish a role for retinoids in murine CLL pathogenesis, and provide new therapeutic strategies to target the microenvironment and to control disease progression. The stromal microenvironment plays a key role in the expansion of chronic lymphocytic leukemia. Here, the authors use the Eµ-TCL1 mouse model to show that leukemic B-cells induce the activation of retinoic acid synthesis in stromal cells of the lymphoid microenvironment, and that impacting on retinoic acid signalling via diet or chemical inhibition prolonged survival by preventing leukemia dissemination and accumulation in lymphoid tissues.
Collapse
|
23
|
Kokhaei P, Hojjat-Farsangi M, Mozaffari F, Moshfegh A, Pak F, Rashidy-Pour A, Palma M, Hansson L, Österborg A, Mellstedt H. Autologous T cells expressing the oncogenic transcription factor KLF6-SV1 prevent apoptosis of chronic lymphocytic leukemia cells. PLoS One 2018; 13:e0192839. [PMID: 29432497 PMCID: PMC5809069 DOI: 10.1371/journal.pone.0192839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Crosstalk between leukemic cells and the tumor microenvironment is of importance in chronic lymphocytic leukemia (CLL). T cells seem to sustain the survival of CLL cells by various mechanisms. The Krüppel-like family of transcription factors (KLFs) are identified as regulators of proliferation and cell death. In the present study, we analyzed the expression of the wild type (WT) gene KLF6 and the oncogenic splice variant 1 (KLF6-SV1) at the mRNA level in subsets of T cells from CLL patients (n = 29), multiple myeloma patients (n = 6) and normal donors (n = 10). RNA Silencing was used for wtKLF6 and KLF6-SV1. Tumor cell apoptosis was measured. A significant overexpression of wtKLF6 and KLF6-SV1 in T cells of CLL patients compared to normal donors and myeloma patients was noted (p<0.002). Western blot showed that both wtKLF6 and KLF6-SV1 were expressed in purified T cells from CLL patients. KLF6-SV1 siRNA transfection induced a significant down-regulation of KLF6-SV1 in CLL T cells, which lost the capability to sustain the growth of leukemic cells. However, no such a significant effect was seen after wtKLF6 transfection of the autologous T cells. The results suggest that KLF6-SV1 may play a role in the regulation of survival CLL cells.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Apoptosis/genetics
- Female
- Gene Expression
- Humans
- Kruppel-Like Factor 6/antagonists & inhibitors
- Kruppel-Like Factor 6/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Multiple Myeloma/genetics
- Multiple Myeloma/pathology
- Oncogenes
- Protein Isoforms/antagonists & inhibitors
- Protein Isoforms/genetics
- RNA Interference
- RNA, Messenger/genetics
- RNA, Neoplasm/genetics
- RNA, Small Interfering/genetics
- T-Lymphocytes/metabolism
- Transfection
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Parviz Kokhaei
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohammad Hojjat-Farsangi
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
- The Persian Gulf Marine Biotechnology Medicine Research Center and Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Fariba Mozaffari
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Ali Moshfegh
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Fatemeh Pak
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Physiology Research Center and Department of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Marzia Palma
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Lotta Hansson
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anders Österborg
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Håkan Mellstedt
- Immune and Gene Therapy Laboratory, Cancer Centre Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
24
|
Byford ET, Carr M, Ladikou E, Ahearne MJ, Wagner SD. Circulating Tfh1 (cTfh1) cell numbers and PD1 expression are elevated in low-grade B-cell non-Hodgkin's lymphoma and cTfh gene expression is perturbed in marginal zone lymphoma. PLoS One 2018; 13:e0190468. [PMID: 29293620 PMCID: PMC5749831 DOI: 10.1371/journal.pone.0190468] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/17/2017] [Indexed: 02/07/2023] Open
Abstract
CD4+ T-cell subsets are found in the tumour microenvironment (TME) of low-grade B-cell non-Hodgkin’s lymphomas such as marginal zone lymphoma (MZL) or follicular lymphoma (FL). Both numbers and architecture of activating follicular helper T-cells (Tfh) and suppressive Treg in the TME of FL are associated with clinical outcomes. There has been almost no previous work on CD4+ T-cells in MZL. It is now recognised that circulating CD4+CXCR5+ T-cells are the memory compartment of Tfh cells. We determined differences in number of circulating Tfh (cTfh) cells and cTfh subsets between normal subjects and patients with FL or MZL. Lymphoma patients showed increased numbers of cTfh1 and reduced cTfh17 cells due to decreased expression of the subset-defining marker CCR6 in patients. PD1, a surface marker associated with Tfh cells, showed increased expression on cTfh subsets in patients. Focusing on MZL we determined expression of 96 T-cell associated genes by microfluidic qRT-PCR. Analysis of differentially expressed genes showed significant differences between normal subjects and patients both for bulk cTfh (CCL4) and the cTfh1 subset (JAK3). While our findings require confirmation in larger studies we suggest that analysis of number and gene expression of circulating T-cells might be a source of clinically useful information as is the case for T-cells within lymphoma lymph nodes.
Collapse
Affiliation(s)
- Elliot T. Byford
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematology Research Institute, University of Leicester, Leicester, United Kingdom
| | - Matthew Carr
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematology Research Institute, University of Leicester, Leicester, United Kingdom
| | - Eleni Ladikou
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematology Research Institute, University of Leicester, Leicester, United Kingdom
| | - Matthew J. Ahearne
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematology Research Institute, University of Leicester, Leicester, United Kingdom
| | - Simon D. Wagner
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematology Research Institute, University of Leicester, Leicester, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Crompot E, Van Damme M, Pieters K, Vermeersch M, Perez-Morga D, Mineur P, Maerevoet M, Meuleman N, Bron D, Lagneaux L, Stamatopoulos B. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica 2017; 102:1594-1604. [PMID: 28596280 PMCID: PMC5685228 DOI: 10.3324/haematol.2016.163337] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 06/05/2017] [Indexed: 12/13/2022] Open
Abstract
Interactions between chronic lymphocytic leukemia (CLL) B cells and the bone marrow (BM) microenvironment play a major function in the physiopathology of CLL. Extracellular vesicles (EVs), which are composed of exosomes and microparticles, play an important role in cell communication. However, little is known about their role in CLL / microenvironment interactions. In the present study, EVs purified by ultracentrifugation from BM mesenchymal stromal cell (BM-MSC) cultures were added to CLL B cells. After their integration into CLL B cells, we observed a decrease of leukemic cell spontaneous apoptosis and an increase in their chemoresistance to several drugs, including fludarabine, ibrutinib, idelalisib and venetoclax after 24 hours. Spontaneous (P=0.0078) and stromal cell-derived factor 1α -induced migration capacities of CLL B cells were also enhanced (P=0.0020). A microarray study highlighted 805 differentially expressed genes between leukemic cells cultured with or without EVs. Of these, genes involved in the B-cell receptor pathway such as CCL3/4, EGR1/2/3, and MYC were increased. Interestingly, this signature presents important overlaps with other microenvironment stimuli such as B-cell receptor stimulation, CLL/nurse-like cells co-culture or those provided by a lymph node microenvironment. Finally, we showed that EVs from MSCs of leukemic patients also rescue leukemic cells from spontaneous or drug-induced apoptosis. However, they induce a higher migration and also a stronger gene modification compared to EVs of healthy MSCs. In conclusion, we show that EVs play a crucial role in CLL B cells/BM microenvironment communication.
Collapse
Affiliation(s)
- Emerence Crompot
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Michael Van Damme
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Karlien Pieters
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Perez-Morga
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Philippe Mineur
- Department of Hemato-Oncology, Grand Hôpital de Charleroi, Gilly, Belgium
| | - Marie Maerevoet
- Hematology Department, Jules Bordet Institute, Brussels, Belgium
| | | | - Dominique Bron
- Hematology Department, Jules Bordet Institute, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy, Université Libre de Bruxelles (ULB), Jules Bordet Institute, Belgium
| |
Collapse
|
26
|
Kashyap MK, Amaya-Chanaga CI, Kumar D, Simmons B, Huser N, Gu Y, Hallin M, Lindquist K, Yafawi R, Choi MY, Amine AA, Rassenti LZ, Zhang C, Liu SH, Smeal T, Fantin VR, Kipps TJ, Pernasetti F, Castro JE. Targeting the CXCR4 pathway using a novel anti-CXCR4 IgG1 antibody (PF-06747143) in chronic lymphocytic leukemia. J Hematol Oncol 2017. [PMID: 28526063 DOI: 10.1186/s13045-017-0435-x,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The CXCR4-CXCL12 axis plays an important role in the chronic lymphocytic leukemia (CLL)-microenvironment interaction. Overexpression of CXCR4 has been reported in different hematological malignancies including CLL. Binding of the pro-survival chemokine CXCL12 with its cognate receptor CXCR4 induces cell migration. CXCL12/CXCR4 signaling axis promotes cell survival and proliferation and may contribute to the tropism of leukemia cells towards lymphoid tissues and bone marrow. Therefore, we hypothesized that targeting CXCR4 with an IgG1 antibody, PF-06747143, may constitute an effective therapeutic approach for CLL. METHODS Patient-derived primary CLL-B cells were assessed for cytotoxicity in an in vitro model of CLL microenvironment. PF-06747143 was analyzed for cell death induction and for its potential to interfere with the chemokine CXCL12-induced mechanisms, including migration and F-actin polymerization. PF-06747143 in vivo efficacy was determined in a CLL murine xenograft tumor model. RESULTS PF-06747143, a novel-humanized IgG1 CXCR4 antagonist antibody, induced cell death of patient-derived primary CLL-B cells, in presence or absence of stromal cells. Moreover, cell death induction by the antibody was independent of CLL high-risk prognostic markers. The cell death mechanism was dependent on CXCR4 expression, required antibody bivalency, involved reactive oxygen species production, and did not require caspase activation, all characteristics reminiscent of programmed cell death (PCD). PF-06747143 also induced potent B-CLL cytotoxicity via Fc-driven antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity activity (CDC). PF-06747143 had significant combinatorial effect with standard of care (SOC) agents in B-CLL treatment, including rituximab, fludarabine (F-ara-A), ibrutinib, and bendamustine. In a CLL xenograft model, PF-06747143 decreased tumor burden and improved survival as a monotherapy, and in combination with bendamustine. CONCLUSIONS We show evidence that PF-06747143 has biological activity in CLL primary cells, supporting a rationale for evaluation of PF-06747143 for the treatment of CLL patients.
Collapse
Affiliation(s)
- Manoj K Kashyap
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Carlos I Amaya-Chanaga
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Deepak Kumar
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Brett Simmons
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Nanni Huser
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Yin Gu
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Max Hallin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.,Present Address: Mirati Therapeutics, San Diego, CA, USA
| | - Kevin Lindquist
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Rolla Yafawi
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, CA, USA
| | - Michael Y Choi
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ale-Ali Amine
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Laura Z Rassenti
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cathy Zhang
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Shu-Hui Liu
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Tod Smeal
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.,Present Address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Valeria R Fantin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.,Present Address: ORIC Pharmaceuticals, South San Francisco, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Flavia Pernasetti
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.
| | - Januario E Castro
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA. .,CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
27
|
Kashyap MK, Amaya-Chanaga CI, Kumar D, Simmons B, Huser N, Gu Y, Hallin M, Lindquist K, Yafawi R, Choi MY, Amine AA, Rassenti LZ, Zhang C, Liu SH, Smeal T, Fantin VR, Kipps TJ, Pernasetti F, Castro JE. Targeting the CXCR4 pathway using a novel anti-CXCR4 IgG1 antibody (PF-06747143) in chronic lymphocytic leukemia. J Hematol Oncol 2017; 10:112. [PMID: 28526063 PMCID: PMC5438492 DOI: 10.1186/s13045-017-0435-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The CXCR4-CXCL12 axis plays an important role in the chronic lymphocytic leukemia (CLL)-microenvironment interaction. Overexpression of CXCR4 has been reported in different hematological malignancies including CLL. Binding of the pro-survival chemokine CXCL12 with its cognate receptor CXCR4 induces cell migration. CXCL12/CXCR4 signaling axis promotes cell survival and proliferation and may contribute to the tropism of leukemia cells towards lymphoid tissues and bone marrow. Therefore, we hypothesized that targeting CXCR4 with an IgG1 antibody, PF-06747143, may constitute an effective therapeutic approach for CLL. METHODS Patient-derived primary CLL-B cells were assessed for cytotoxicity in an in vitro model of CLL microenvironment. PF-06747143 was analyzed for cell death induction and for its potential to interfere with the chemokine CXCL12-induced mechanisms, including migration and F-actin polymerization. PF-06747143 in vivo efficacy was determined in a CLL murine xenograft tumor model. RESULTS PF-06747143, a novel-humanized IgG1 CXCR4 antagonist antibody, induced cell death of patient-derived primary CLL-B cells, in presence or absence of stromal cells. Moreover, cell death induction by the antibody was independent of CLL high-risk prognostic markers. The cell death mechanism was dependent on CXCR4 expression, required antibody bivalency, involved reactive oxygen species production, and did not require caspase activation, all characteristics reminiscent of programmed cell death (PCD). PF-06747143 also induced potent B-CLL cytotoxicity via Fc-driven antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity activity (CDC). PF-06747143 had significant combinatorial effect with standard of care (SOC) agents in B-CLL treatment, including rituximab, fludarabine (F-ara-A), ibrutinib, and bendamustine. In a CLL xenograft model, PF-06747143 decreased tumor burden and improved survival as a monotherapy, and in combination with bendamustine. CONCLUSIONS We show evidence that PF-06747143 has biological activity in CLL primary cells, supporting a rationale for evaluation of PF-06747143 for the treatment of CLL patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- CHO Cells
- Cell Death/drug effects
- Cricetulus
- Female
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice, Inbred BALB C
- Mice, SCID
- Reactive Oxygen Species/immunology
- Receptors, CXCR4/analysis
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/immunology
- Signal Transduction/drug effects
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Manoj K Kashyap
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Carlos I Amaya-Chanaga
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Deepak Kumar
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Brett Simmons
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Nanni Huser
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Yin Gu
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Max Hallin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
- Present Address: Mirati Therapeutics, San Diego, CA, USA
| | - Kevin Lindquist
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Rolla Yafawi
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, CA, USA
| | - Michael Y Choi
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ale-Ali Amine
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
| | - Laura Z Rassenti
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cathy Zhang
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
| | - Shu-Hui Liu
- Oncology Research & Development-Rinat Biotechnology Unit, Pfizer Worldwide Research & Development, South San Francisco, CA, USA
| | - Tod Smeal
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
- Present Address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Valeria R Fantin
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA
- Present Address: ORIC Pharmaceuticals, South San Francisco, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Flavia Pernasetti
- Oncology Research & Development, Pfizer Worldwide Research & Development, 10646 Science Center Drive, San Diego, CA, 92121, USA.
| | - Januario E Castro
- Moores Cancer Center, University of California San Diego, 3855 Health Science Drive, La Jolla, CA, 92093-0820, USA.
- CLL Research Consortium, and Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Gusella M, Bolzonella C, Paolini R, Rodella E, Bertolaso L, Scipioni C, Bellini S, Cuneo A, Pasini F, Ramazzina E. Plasma matrix metalloprotease 9 correlates with blood lymphocytosis, leukemic cell invasiveness, and prognosis in B-cell chronic lymphocytic leukemia. Tumour Biol 2017; 39:1010428317694325. [DOI: 10.1177/1010428317694325] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The complex biology underlying chronic lymphocytic leukemia cell migration and tissue invasiveness is not yet completely understood and might provide novel predictive markers and therapeutic targets. A total of 36 patients out of treatment from at least 3 months were enrolled and followed up for a median period of 44.2 months (range: 4.4–99.2). Matrix metalloprotease 9 and tissue inhibitor of metalloproteases 1 plasma levels and production/release from lymphoid cells were measured by zymography and enzyme-linked immunosorbent assay (ELISA) analysis. Malignant and normal lymphocyte mobility and matrix-degradation capability were studied using a Boyden chamber system, with and without autologous plasma. Free matrix metalloprotease 9 plasma levels were related with blood lymphocytosis, especially in more advanced stages (p = 0.003), and higher concentrations were associated with an increased disease progression risk (hazard ratio = 9.0, 95% confidence interval = 1.5–13.8). Leukemic cells expressed and secreted very little matrix metalloprotease 9. On the contrary, normal lymphocytes derived from the same leukemic patients showed matrix metalloprotease 9 intracellular levels that were lower in subjects with higher blood lymphocytosis (p = 0.024) and more advanced stages (p = 0.03); the released quantities were inversely associated with matrix metalloprotease 9 plasma concentrations (p = 0.035). Leukemic cells had a reduced spontaneous mobility and matrix-degradation capability that were stimulated by autologous plasma (p = 0.001) and normal lymphocytes (p = 0.005), respectively. Matrix metalloprotease 9 affected cell invasiveness depending on concentration and disease stage. In conclusion, chronic lymphocytic leukemia cells have a reduced mobility, matrix-degradation capability, and matrix metalloprotease 9 production compared to their own autologous normal lymphocytes. They are exposed to matrix metalloprotease 9 of prevalently systemic origin whose higher levels are associated with both leukemic and normal lymphocyte accumulation in the peripheral blood and have a negative prognostic value.
Collapse
Affiliation(s)
- Milena Gusella
- Department of Oncology, Azienda ULSS 18 Rovigo, Rovigo, Italy
| | | | | | | | - Laura Bertolaso
- Department of Oncology, Azienda ULSS 18 Rovigo, Rovigo, Italy
| | - Cinzia Scipioni
- Department of Transfusion Medicine, Azienda ULSS 18 Rovigo, Rovigo, Italy
| | - Silvia Bellini
- Department of Transfusion Medicine, Azienda ULSS 18 Rovigo, Rovigo, Italy
| | - Antonio Cuneo
- Department of Medical Sciences, Section of Hematology, University of Ferrara, Ferrara, Italy
| | - Felice Pasini
- Department of Oncology, Azienda ULSS 18 Rovigo, Rovigo, Italy
| | | |
Collapse
|
29
|
Bone marrow stroma-induced resistance of chronic lymphocytic leukemia cells to arsenic trioxide involves Mcl-1 upregulation and is overcome by inhibiting the PI3Kδ or PKCβ signaling pathways. Oncotarget 2016; 6:44832-48. [PMID: 26540567 PMCID: PMC4792595 DOI: 10.18632/oncotarget.6265] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/22/2015] [Indexed: 11/25/2022] Open
Abstract
CLL remains an incurable disease in spite of the many new compounds being studied. Arsenic trioxide (ATO) induces apoptosis in all CLL cell types and could constitute an efficient therapy. To further explore this, we have studied the influence of stromal cells, key components of the CLL microenvironment, on the response of CLL cells to ATO. Bone marrow stromal cells induced CLL cell resistance to 2 μM ATO and led to activation of Lyn, ERK, PI3K and PKC, as well as NF-κB and STAT3. Mcl-1, Bcl-xL, and Bfl-1 were also upregulated after the co-culture. Inhibition experiments indicated that PI3K and PKC were involved in the resistance to ATO induced by stroma. Moreover, idelalisib and sotrastaurin, specific inhibitors for PI3Kδ and PKCβ, respectively, inhibited Akt phosphorylation, NF-κB/STAT3 activation and Mcl-1 upregulation, and rendered cells sensitive to ATO. Mcl-1 was central to the mechanism of resistance to ATO, since: 1) Mcl-1 levels correlated with the CLL cell response to ATO, and 2) blocking Mcl-1 expression or function with specific siRNAs or inhibitors overcame the protecting effect of stroma. We have therefore identified the mechanism involved in the CLL cell resistance to ATO induced by bone marrow stroma and show that idelalisib or sotrastaurin block this mechanism and restore sensibility to ATO. Combination of ATO with these inhibitors may thus constitute an efficient treatment for CLL.
Collapse
|
30
|
Rombout A, Verhasselt B, Philippé J. Lipoprotein lipase in chronic lymphocytic leukemia: function and prognostic implications. Eur J Haematol 2016; 97:409-415. [PMID: 27504855 DOI: 10.1111/ejh.12789] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2016] [Indexed: 12/17/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a clinically heterogeneous disease characterized by the accumulation of a clonal population of B cells in peripheral blood, bone marrow, and lymphoid organs. More than 10 years ago, lipoprotein lipase (LPL) mRNA was identified as being strongly expressed in patients experiencing a more aggressive phenotype, while CLL patients with an indolent disease course lack expression of this marker. Since then, several reports confirmed the capability of LPL to predict CLL disease evolution at the moment of diagnosis. In contrast, data on the functional implications of LPL in CLL are scarce. LPL exerts a central role in overall lipid metabolism and transport, but plays additional, non-catalytic roles as well. Which of those is more important in the pathogenesis of CLL remains largely unclear. Here, we review the current knowledge on the prognostic and biological relevance of LPL in CLL.
Collapse
Affiliation(s)
- Ans Rombout
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Bruno Verhasselt
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Jan Philippé
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent University, Ghent, Belgium.
| |
Collapse
|
31
|
Cavallini C, Lovato O, Bertolaso A, Zoratti E, Malpeli G, Mimiola E, Tinelli M, Aprili F, Tecchio C, Perbellini O, Scarpa A, Zamò A, Cassatella MA, Pizzolo G, Scupoli MT. Expression and function of the TL1A/DR3 axis in chronic lymphocytic leukemia. Oncotarget 2016; 6:32061-74. [PMID: 26393680 PMCID: PMC4741659 DOI: 10.18632/oncotarget.5201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/04/2015] [Indexed: 01/15/2023] Open
Abstract
TNF-like ligand 1A (TL1A) and its unique receptor death receptor 3 (DR3) acts as broad T-cell costimulator involved in regulatory mechanisms of adaptive immune response under physiological and pathological settings. Moreover, we have recently shown that TL1A negatively regulates B-cell proliferation. Despite increasing interest on the TL1A/DR3-axis functions, very little is known on its expression and role in leukemia. In this study, we investigated the expression and function of TL1A/DR3 axis in chronic lymphocytic leukemia (CLL). DR3 was differentially expressed in activated CLL cells and predominantly detected in patients with early clinical stage disease. Soluble TL1A has been revealed in the sera of CLL patients where higher TL1A levels were associated with early stage disease. T cells, monocytes and leukemic B cells have been identified as major sources of TL1A in CLL. The relevance of these findings has been sustained by functional data showing that exogenous TL1A reduces CLL proliferation induced by stimulation of the B cell receptor. Overall, these data document the expression of the TL1A/DR3 axis in early-stage CLL. They also identify a novel function for TL1A as a negative regulator of leukemic cell proliferation that may influence the CLL physiopathology and clinical outcome at an early-stage disease.
Collapse
Affiliation(s)
- Chiara Cavallini
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy
| | - Ornella Lovato
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy
| | - Anna Bertolaso
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy
| | - Elisa Zoratti
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Giorgio Malpeli
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Elda Mimiola
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Martina Tinelli
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Fiorenza Aprili
- Department of Pathology and Diagnostics, Laboratory of Cytogenetics, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Cristina Tecchio
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Omar Perbellini
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Alberto Zamò
- Department of Pathology and Diagnostics, Section of Pathological Anatomy, University of Verona, Verona, Italy
| | - Marco Antonio Cassatella
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
| | - Giovanni Pizzolo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Maria Teresa Scupoli
- Interdepartmental Laboratory of Medical Research (LURM), University of Verona, Verona, Italy.,Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy.,Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| |
Collapse
|
32
|
Alsagaby SA, Brennan P, Pepper C. Key Molecular Drivers of Chronic Lymphocytic Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 16:593-606. [PMID: 27601002 DOI: 10.1016/j.clml.2016.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/29/2016] [Accepted: 08/02/2016] [Indexed: 01/01/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is an adult neoplastic disease of B cells characterized by variable clinical outcomes. Although some patients have an aggressive form of the disease and often encounter treatment failure and short survival, others have more stable disease with long-term survival and little or no need for theraphy. In the past decade, significant advances have been made in our understanding of the molecular drivers that affect the natural pathology of CLL. The present review describes what is known about these key molecules in the context of their role in tumor pathogenicity, prognosis, and therapy.
Collapse
Affiliation(s)
- Suliman A Alsagaby
- Department of Medical Laboratory, College of Science, Majmaah University, Al-Zuli, Kingdom of Saudi Arabia; Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.
| | - Paul Brennan
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Chris Pepper
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
33
|
CD84 mediates CLL-microenvironment interactions. Oncogene 2016; 36:628-638. [PMID: 27452524 DOI: 10.1038/onc.2016.238] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/04/2016] [Accepted: 06/01/2016] [Indexed: 12/21/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a malignant disease of small mature lymphocytes. Signals from the CLL microenvironment promote progression of the disease and induce drug resistance. This phenomenon is largely dependent on direct contact between the malignant B cells and stromal cells. CD84 belongs to the signaling lymphocyte activation molecule family of immunoreceptors, which self-associates, forming an orthogonal homophilic dimer. We therefore hypothesized that CD84 may bridge between CLL cells and their microenvironment, promoting cell survival. Our in vitro results show that CD84 expressed on CLL cells interact with CD84 expressed on cells in their microenvironment, inducing cell survival in both sides. Blocking CD84 in vitro and in vivo disrupt the interaction of CLL cells with their microenvironment, resulting in induced cell death. Thus, our findings suggest novel therapeutic strategies based on the blockade of this CD84-dependent survival pathway.
Collapse
|
34
|
|
35
|
Butrym A, Gebura K, Iwaszko M, Kuliczkowski K, Bogunia-Kubik K, Mazur G. Dual role of the CXCL12 polymorphism in patients with chronic lymphocytic leukemia. HLA 2016; 87:432-8. [DOI: 10.1111/tan.12810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/12/2016] [Accepted: 04/12/2016] [Indexed: 01/03/2023]
Affiliation(s)
- A. Butrym
- Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
- Department of Physiology; Wroclaw Medical University; Wroclaw Poland
| | - K. Gebura
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy; Polish Academy of Sciences; Wroclaw Poland
| | - M. Iwaszko
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy; Polish Academy of Sciences; Wroclaw Poland
| | - K. Kuliczkowski
- Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - K. Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy; Polish Academy of Sciences; Wroclaw Poland
- Department of Internal, Occupational Diseases, Hypertension and Clinical Oncology; Wroclaw Medical University; Wroclaw Poland
| | - G. Mazur
- Department of Internal, Occupational Diseases, Hypertension and Clinical Oncology; Wroclaw Medical University; Wroclaw Poland
| |
Collapse
|
36
|
Jain N, O'Brien S. Targeted therapies for CLL: Practical issues with the changing treatment paradigm. Blood Rev 2016; 30:233-44. [PMID: 26776345 DOI: 10.1016/j.blre.2015.12.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/02/2015] [Accepted: 12/14/2015] [Indexed: 11/21/2022]
Abstract
Chemoimmunotherapy (CIT) such as FCR (fludarabine, cyclophosphamide, rituximab) has been the standard first-line therapy for younger patients with CLL. In the last few years, several novel targeted therapies have been developed for patients with CLL. These include B-cell receptor (BCR) inhibitors such as Bruton tyrosine kinase (BTK) inhibitors, PI3 kinase inhibitors, and Syk inhibitors, novel anti-CD20 monoclonal antibodies such as ofatumumab and obinutuzumab, and Bcl-2 antagonists such as venetoclax (ABT-199). Strategies targeting the immune system such as lenalidomide, chimeric antigen receptor (CAR) T cells, and more recently, checkpoint inhibitors, are in clinical development. Ibrutinib and idelalisib are already approved for patients with relapsed and refractory CLL. Ibrutinib is also approved for first-line treatment of CLL patients with del(17p). Several ongoing phase III clinical trials with novel therapies will further define the role of targeted agents in CLL.
Collapse
MESH Headings
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Combined Modality Therapy
- Disease Management
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Molecular Targeted Therapy
- Treatment Outcome
Collapse
Affiliation(s)
- Nitin Jain
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Susan O'Brien
- Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA, USA.
| |
Collapse
|
37
|
Li PP, Lu K, Geng LY, Zhou XX, Li XY, Wang X. Bruton's tyrosine kinase inhibitor restrains Wnt signaling in chronic lymphocytic leukemia. Mol Med Rep 2016; 13:4934-8. [PMID: 27082823 DOI: 10.3892/mmr.2016.5111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 03/29/2016] [Indexed: 11/06/2022] Open
Abstract
The B-cell receptor (BCR) signaling pathway serves an important role in the pathogenesis of chronic lymphocytic leukemia (CLL), and has been identified as a novel and effective therapeutic target of CLL, with particular focus its kinase factor, BTK. Previous studies have focused on combining the BTK inhibitor with additional chemotherapeutic agents to improve the prognosis of patients with CLL. Further investigation into the mechanism of the BTK inhibitor would promote an understanding of the pathogenesis of CLL. The current study investigated the association between ibrutinib and the Wnt signaling pathway, additionally focussing upon one of its regulators, metadherin (MTDH), which has been identified to be overexpressed in CLL and is considered a promoter of the Wnt pathway. The experiments in the current study were performed in the MEC-1 CLL cell line. Results indicated that MTDH, β-catenin and lymphoid-enhancing factor-1 were inhibited subsequent to ibrutinib treatment. The results indicate that in CLL, ibrutinib is likely to possess an inhibitory role in Wnt signaling.
Collapse
Affiliation(s)
- Pei-Pei Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Ling-Yun Geng
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiang-Xiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin-Yu Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
38
|
Herman SEM, Wiestner A. Preclinical modeling of novel therapeutics in chronic lymphocytic leukemia: the tools of the trade. Semin Oncol 2016; 43:222-32. [PMID: 27040700 DOI: 10.1053/j.seminoncol.2016.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the last decade our understanding of chronic lymphocytic leukemia (CLL) biology and pathogenesis has increased substantially. These insights have led to the development of several new agents with novel mechanisms of action prompting a change in therapeutic approaches from chemotherapy-based treatments to targeted therapies. Multiple preclinical models for drug development in CLL are available; however, with the advent of these targeted agents, it is becoming clear that not all models and surrogate readouts of efficacy are appropriate for all drugs. In this review we discuss in vitro and in vivo preclinical models, with a particular focus on the benefits and possible pitfalls of different model systems in the evaluation of novel therapeutics for the treatment of CLL.
Collapse
Affiliation(s)
- Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
39
|
Kashyap MK, Kumar D, Jones H, Amaya-Chanaga CI, Choi MY, Melo-Cardenas J, Ale-Ali A, Kuhne MR, Sabbatini P, Cohen LJ, Shelat SG, Rassenti LZ, Kipps TJ, Cardarelli PM, Castro JE. Ulocuplumab (BMS-936564 / MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget 2016; 7:2809-2822. [PMID: 26646452 PMCID: PMC4823073 DOI: 10.18632/oncotarget.6465] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
The CXCR4 receptor (Chemokine C-X-C motif receptor 4) is highly expressed in different hematological malignancies including chronic lymphocytic leukemia (CLL). The CXCR4 ligand (CXCL12) stimulates CXCR4 promoting cell survival and proliferation, and may contribute to the tropism of leukemia cells towards lymphoid tissues. Therefore, strategies targeting CXCR4 may constitute an effective therapeutic approach for CLL. To address that question, we studied the effect of Ulocuplumab (BMS-936564), a fully human IgG4 anti-CXCR4 antibody, using a stroma--CLL cells co-culture model. We found that Ulocuplumab (BMS-936564) inhibited CXCL12 mediated CXCR4 activation-migration of CLL cells at nanomolar concentrations. This effect was comparable to AMD3100 (Plerixafor--Mozobil), a small molecule CXCR4 inhibitor. However, Ulocuplumab (BMS-936564) but not AMD3100 induced apoptosis in CLL at nanomolar concentrations in the presence or absence of stromal cell support. This pro-apoptotic effect was independent of CLL high-risk prognostic markers, was associated with production of reactive oxygen species and did not require caspase activation. Overall, these findings are evidence that Ulocuplumab (BMS-936564) has biological activity in CLL, highlight the relevance of the CXCR4-CXCL12 pathway as a therapeutic target in CLL, and provide biological rationale for ongoing clinical trials in CLL and other hematological malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michelle R. Kuhne
- Bristol-Myers Squibb, Department of Cell Biology and Physiology, Redwood City, CA, USA
| | - Peter Sabbatini
- Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Lewis J. Cohen
- Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Suresh G. Shelat
- Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | | | - Thomas J. Kipps
- UCSD-Moores Cancer Center, La Jolla, CA, USA
- CLL Research Consortium, La Jolla, CA, USA
| | - Pina M. Cardarelli
- Bristol-Myers Squibb, Department of Cell Biology and Physiology, Redwood City, CA, USA
| | - Januario E. Castro
- UCSD-Moores Cancer Center, La Jolla, CA, USA
- CLL Research Consortium, La Jolla, CA, USA
| |
Collapse
|
40
|
Vitale C, Falchi L, Ten Hacken E, Gao H, Shaim H, Van Roosbroeck K, Calin G, O'Brien S, Faderl S, Wang X, Wierda WG, Rezvani K, Reuben JM, Burger JA, Keating MJ, Ferrajoli A. Ofatumumab and Lenalidomide for Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia: Correlation between Responses and Immune Characteristics. Clin Cancer Res 2016; 22:2359-67. [PMID: 26733610 DOI: 10.1158/1078-0432.ccr-15-2476] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/12/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE We evaluated efficacy and tolerability of the combination of ofatumumab and lenalidomide in patients with relapsed/refractory chronic lymphocytic leukemia (CLL), and explored whether immune system characteristics could influence the response to treatment. EXPERIMENTAL DESIGN Thirty-four patients were enrolled in this phase II study. Ofatumumab was administered at a dose of 300 mg on day 1, 1,000 mg on days 8, 15, and 22 during course 1, 1,000 mg on day 1 during courses 3-6, and once every other course during courses 7-24 (28-day courses). Oral lenalidomide (10 mg daily) was started on day 9 and continued for as long as a clinical benefit was observed. RESULTS The overall response rate was 71%. Eight patients (24%) achieved a complete remission (CR) or CR with incomplete recovery of blood counts, including 9% with minimal residual disease-negative CR. The median progression-free survival was 16 months, and the estimated 5-year survival was 53%. The most common treatment-related toxicity was neutropenia (grade >2 in 18% of the 574 patient courses). The most frequent infectious complications were pneumonia and neutropenic fever (24% and 9% of patients, respectively). We observed that patients who achieved a CR had at baseline higher numbers and a better preserved function of T cells and natural killer cells compared with non-responders. CONCLUSIONS The combination of ofatumumab and lenalidomide is a well-tolerated regimen that induces durable responses in the majority of patients with relapsed/refractory CLL. Our correlative data suggest a role of competent immune system in supporting the efficacy of this treatment. Clin Cancer Res; 22(10); 2359-67. ©2016 AACR.
Collapse
Affiliation(s)
- Candida Vitale
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lorenzo Falchi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elisa Ten Hacken
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Gao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hila Shaim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katrien Van Roosbroeck
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan Faderl
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jan A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
41
|
Li H, Wu X, Hou S, Malek M, Kielkowska A, Noh E, Makondo KJ, Du Q, Wilkins JA, Johnston JB, Gibson SB, Lin F, Marshall AJ. Phosphatidylinositol-3,4-Bisphosphate and Its Binding Protein Lamellipodin Regulate Chemotaxis of Malignant B Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2015; 196:586-95. [DOI: 10.4049/jimmunol.1500630] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 11/16/2015] [Indexed: 01/14/2023]
|
42
|
Martínez-Moreno M, Leiva M, Aguilera-Montilla N, Sevilla-Movilla S, Isern de Val S, Arellano-Sánchez N, Gutiérrez NC, Maldonado R, Martínez-López J, Buño I, García-Marco JA, Sánchez-Mateos P, Hidalgo A, García-Pardo A, Teixidó J. In vivo adhesion of malignant B cells to bone marrow microvasculature is regulated by α4β1 cytoplasmic-binding proteins. Leukemia 2015; 30:861-72. [PMID: 26658839 DOI: 10.1038/leu.2015.332] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/23/2015] [Accepted: 11/24/2015] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) and chronic lymphocytic leukemia (CLL) cells must attach to the bone marrow (BM) microvasculature before lodging in the BM microenvironment. Using intravital microscopy (IVM) of the BM calvariae we demonstrate that the α4β1 integrin is required for MM and CLL cell firm arrest onto the BM microvasculature, while endothelial P-selectin and E-selectin mediate cell rolling. Talin, kindlin-3 and ICAP-1 are β1-integrin-binding partners that regulate β1-mediated cell adhesion. We show that talin and kindlin-3 cooperatively stimulate high affinity and strength of α4β1-dependent MM and CLL cell attachment, whereas ICAP-1 negatively regulates this adhesion. A functional connection between talin/kindlin-3 and Rac1 was found to be required for MM cell attachment mediated by α4β1. Importantly, IVM analyses with talin- and kindlin-3-silenced MM cells indicate that these proteins are needed for cell arrest on the BM microvasculature. Instead, MM cell arrest is repressed by ICAP-1. Moreover, MM cells silenced for talin and kindlin-3, and cultured on α4β1 ligands showed higher susceptibility to bortezomib-mediated cell apoptosis. Our results highlight the requirement of α4β1 and selectins for the in vivo attachment of MM and CLL cells to the BM microvasculature, and indicate that talin, kindlin-3 and ICAP-1 differentially control physiological adhesion by regulating α4β1 activity.
Collapse
Affiliation(s)
- M Martínez-Moreno
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - M Leiva
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - N Aguilera-Montilla
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - S Sevilla-Movilla
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - S Isern de Val
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - N Arellano-Sánchez
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - N C Gutiérrez
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain
| | - R Maldonado
- Section of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - J Martínez-López
- Section of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - I Buño
- Section of Hematology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - J A García-Marco
- Hematology Unit, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - P Sánchez-Mateos
- Section of Immuno-Oncology, Hospital General Universitario Gregorio Marañón, Complutense University School of Medicine, Madrid, Spain
| | - A Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - A García-Pardo
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - J Teixidó
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
43
|
Niemann CU, Herman SEM, Maric I, Gomez-Rodriguez J, Biancotto A, Chang BY, Martyr S, Stetler-Stevenson M, Yuan CM, Calvo KR, Braylan RC, Valdez J, Lee YS, Wong DH, Jones J, Sun C, Marti GE, Farooqui MZH, Wiestner A. Disruption of in vivo Chronic Lymphocytic Leukemia Tumor-Microenvironment Interactions by Ibrutinib--Findings from an Investigator-Initiated Phase II Study. Clin Cancer Res 2015; 22:1572-82. [PMID: 26660519 DOI: 10.1158/1078-0432.ccr-15-1965] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental interactions for proliferation and survival that are at least partially mediated through B-cell receptor (BCR) signaling. Ibrutinib, a Bruton tyrosine kinase inhibitor, disrupts BCR signaling and leads to the egress of tumor cells from the microenvironment. Although the on-target effects on CLL cells are well defined, the impact on the microenvironment is less well studied. We therefore sought to characterize the in vivo effects of ibrutinib on the tumor microenvironment. EXPERIMENTAL DESIGN Patients received single-agent ibrutinib on an investigator-initiated phase II trial. Serial blood and tissue samples were collected pretreatment and during treatment. Changes in cytokine levels, cellular subsets, and microenvironmental interactions were assessed. RESULTS Serum levels of key chemokines and inflammatory cytokines decreased significantly in patients on ibrutinib. Furthermore, ibrutinib treatment decreased circulating tumor cells and overall T-cell numbers. Most notably, a reduced frequency of the Th17 subset of CD4(+)T cells was observed concurrent with reduced expression of activation markers and PD-1 on T cells. Consistent with direct inhibition of T cells, ibrutinib inhibited Th17 differentiation of murine CD4(+)T cells in vitro Finally, in the bone marrow microenvironment, we found that ibrutinib disaggregated the interactions of macrophages and CLL cells, inhibited secretion of CXCL13, and decreased the chemoattraction of CLL cells. CONCLUSIONS In conjunction with inhibition of BCR signaling, these changes in the tumor microenvironment likely contribute to the antitumor activity of ibrutinib and may impact the efficacy of immunotherapeutic strategies in patients with CLL. See related commentary by Bachireddy and Wu, p. 1547.
Collapse
Affiliation(s)
- Carsten U Niemann
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland. Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Irina Maric
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, Maryland
| | | | - Angelique Biancotto
- Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, Maryland
| | | | - Sabrina Martyr
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | | | - Constance M Yuan
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Katherine R Calvo
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, Maryland
| | - Raul C Braylan
- Department of Laboratory Medicine, Clinical Research Center, NIH, Bethesda, Maryland
| | - Janet Valdez
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Yuh Shan Lee
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Deanna H Wong
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Jade Jones
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland. Medical Research Scholars Program, NIH, Bethesda, Maryland
| | - Clare Sun
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Gerald E Marti
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Mohammed Z H Farooqui
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
44
|
Sutton LA, Rosenquist R. The complex interplay between cell-intrinsic and cell-extrinsic factors driving the evolution of chronic lymphocytic leukemia. Semin Cancer Biol 2015; 34:22-35. [DOI: 10.1016/j.semcancer.2015.04.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 01/08/2023]
|
45
|
Lyn sustains oncogenic signaling in chronic lymphocytic leukemia by strengthening SET-mediated inhibition of PP2A. Blood 2015; 125:3747-55. [DOI: 10.1182/blood-2014-12-619155] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/21/2015] [Indexed: 01/12/2023] Open
Abstract
Key Points
Cytosolic HSP90-bound Lyn mediates resistance to apoptosis by strengthening PP2A/SET interaction in CLL cells. FTY720-analogues antagonizing the PP2A/SET interaction and Lyn inhibitors may provide a therapeutic approach of CLL.
Collapse
|
46
|
Initial treatment of CLL: integrating biology and functional status. Blood 2015; 126:463-70. [PMID: 26065656 DOI: 10.1182/blood-2015-04-585067] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/23/2015] [Indexed: 11/20/2022] Open
Abstract
A better understanding of the biology of chronic lymphocytic leukemia (CLL) has led to significant advances in therapeutic strategies for patients with CLL. Chemoimmunotherapy (CIT) has been the standard first-line therapy for CLL. Age and comorbidities can help decide which patients may benefit from a CIT approach. FCR (fludarabine, cyclophosphamide, and rituximab) is the current standard treatment option for younger patients with CLL. For older patients and for patients with renal dysfunction, bendamustine and rituximab may be a better option. For older patients with comorbidities who may not be able to tolerate intensive CIT, the combination treatment of chlorambucil and obinutuzumab or ofatumumab is an option. For patients with del(17p), ibrutinib is the treatment of choice. Several ongoing phase 3 clinical trials with novel therapies will further refine the frontline therapy of CLL.
Collapse
|
47
|
Wei M, Wang X, Song Z, Jiao M, Ding J, Meng LH, Zhang A. Targeting PI3Kδ: Emerging Therapy for Chronic Lymphocytic Leukemia and Beyond. Med Res Rev 2015; 35:720-52. [DOI: 10.1002/med.21341] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Manman Wei
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| | - Xiang Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| | - Zilan Song
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| | - Mingkun Jiao
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| | - Jian Ding
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| | - Ling-Hua Meng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM); Chinese Academy of Sciences; Shanghai 201203 China
| |
Collapse
|
48
|
Lu K, Fang XS, Feng LL, Jiang YJ, Zhou XX, Liu X, Li PP, Chen N, Ding M, Wang N, Zhang J, Wang X. The STAT3 inhibitor WP1066 reverses the resistance of chronic lymphocytic leukemia cells to histone deacetylase inhibitors induced by interleukin-6. Cancer Lett 2015; 359:250-8. [PMID: 25636517 DOI: 10.1016/j.canlet.2015.01.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/14/2015] [Accepted: 01/17/2015] [Indexed: 10/24/2022]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine produced by a variety of cell types, including fibroblasts, endothelial cells, lymphocytes, and bone marrow stromal cells (BMSCs). Levels of IL-6 are increased in serum of CLL patients and correlated with adverse clinical features and short survival. In our study, we observed that IL-6 induced the resistance of CLL cells to pan-histone deacetylase (HDAC) inhibitors vorinostat (SAHA) and panobinostat (LBH589). Furthermore, low concentrations of SAHA and LBH589 enhanced the activation of the signal transducer and activator of transcription 3 (STAT3) signaling pathway induced by IL-6 in CLL cells. All of these effects were blocked by the STAT3-selective inhibitor, WP1066. Meanwhile, WP1066 decreased the expressions of Mcl-1 and Bcl-xL protein induced by IL-6 with or without low concentrations of HDAC inhibitors. Co-culture of CLL cells with BMSCs could also facilitate the activation of STAT3 and protected CLL cells from apoptosis when treated with HDAC inhibitors, and this cytoprotection was reversed by WP1066. The present study indicated that IL-6 or co-culture with BMSCs prevented HDAC inhibitor-induced apoptosis of CLL cells. This prevention was mediated by activation of the STAT3 signaling pathway. Moreover, WP1066 reversed the resistance of CLL cells to SAHA and LBH589 induced by either IL-6 or co-culture with BMSCs. Our findings suggest that targeting the STAT3 pathway may be a novel way to improve the efficacy of the HDAC inhibitor in CLL patients by overcoming antiapoptotic signaling of the microenvironment.
Collapse
Affiliation(s)
- Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xiao-sheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Li-li Feng
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Yu-jie Jiang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xiang-xiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xin Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Pei-pei Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Na Chen
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Mei Ding
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Na Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Jie Zhang
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Diagnostics, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|