1
|
Ferrer G, Palacios F, Chiu PY, Wong K, Bueno-Costa A, Barrientos JC, Kolitz JE, Allen SL, Rai KR, Chen SS, Sherry B, Chiorazzi N. CLL crosstalk with naïve T cells enhances the differentiation of IL-22-producing T cells and CLL -cell survival. Leukemia 2025; 39:499-502. [PMID: 39578533 PMCID: PMC11794130 DOI: 10.1038/s41375-024-02463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/29/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024]
Affiliation(s)
- Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
- Cancer Epigenetics, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain.
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Madrid, Spain.
| | - Florencia Palacios
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Chronic Lymphocytic Leukemia laboratory Research, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Pui Yan Chiu
- Center for Immunology & Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Kelly Wong
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Alberto Bueno-Costa
- Cancer Epigenetics, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Madrid, Spain
| | - Jacqueline C Barrientos
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Jonathan E Kolitz
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Steven L Allen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Kanti R Rai
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Barbara Sherry
- Center for Immunology & Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| |
Collapse
|
2
|
Aroldi A, Mauri M, Ramazzotti D, Villa M, Malighetti F, Crippa V, Cocito F, Borella C, Bossi E, Steidl C, Scollo C, Voena C, Chiarle R, Mologni L, Piazza R, Gambacorti‐Passerini C. Effects of blocking CD24 and CD47 'don't eat me' signals in combination with rituximab in mantle-cell lymphoma and chronic lymphocytic leukaemia. J Cell Mol Med 2023; 27:3053-3064. [PMID: 37654003 PMCID: PMC10568669 DOI: 10.1111/jcmm.17868] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 09/02/2023] Open
Abstract
Mantle-cell lymphoma (MCL) is a B-cell non-Hodgkin Lymphoma (NHL) with a poor prognosis, at high risk of relapse after conventional treatment. MCL-associated tumour microenvironment (TME) is characterized by M2-like tumour-associated macrophages (TAMs), able to interact with cancer cells, providing tumour survival and resistance to immuno-chemotherapy. Likewise, monocyte-derived nurse-like cells (NLCs) present M2-like profile and provide proliferation signals to chronic lymphocytic leukaemia (CLL), a B-cell malignancy sharing with MCL some biological and phenotypic features. Antibodies against TAMs targeted CD47, a 'don't eat me' signal (DEMs) able to quench phagocytosis by TAMs within TME, with clinical effectiveness when combined with Rituximab in pretreated NHL. Recently, CD24 was found as valid DEMs in solid cancer. Since CD24 is expressed during B-cell differentiation, we investigated and identified consistent CD24 in MCL, CLL and primary human samples. Phagocytosis increased when M2-like macrophages were co-cultured with cancer cells, particularly in the case of paired DEMs blockade (i.e. anti-CD24 + anti-CD47) combined with Rituximab. Similarly, unstimulated CLL patients-derived NLCs provided increased phagocytosis when DEMs blockade occurred. Since high levels of CD24 were associated with worse survival in both MCL and CLL, anti-CD24-induced phagocytosis could be considered for future clinical use, particularly in association with other agents such as Rituximab.
Collapse
Affiliation(s)
- Andrea Aroldi
- Hematology DivisionSan Gerardo HospitalMonzaItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Mario Mauri
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Daniele Ramazzotti
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Matteo Villa
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | | | - Valentina Crippa
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | | | | | - Elisa Bossi
- Hematology DivisionSan Gerardo HospitalMonzaItaly
| | - Carolina Steidl
- Lymphoma Unit, Department of Onco‐HematologyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Chiara Scollo
- Transfusion Medicine UnitSan Gerardo HospitalMonzaItaly
| | - Claudia Voena
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
- Department of PathologyBoston Children's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Division of HematopathologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
| | - Luca Mologni
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Rocco Piazza
- Hematology DivisionSan Gerardo HospitalMonzaItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Carlo Gambacorti‐Passerini
- Hematology DivisionSan Gerardo HospitalMonzaItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| |
Collapse
|
3
|
Integrin Signaling Shaping BTK-Inhibitor Resistance. Cells 2022; 11:cells11142235. [PMID: 35883678 PMCID: PMC9322986 DOI: 10.3390/cells11142235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Integrins are adhesion molecules that function as anchors in retaining tumor cells in supportive tissues and facilitating metastasis. Beta1 integrins are known to contribute to cell adhesion-mediated drug resistance in cancer. Very late antigen-4 (VLA-4), a CD49d/CD29 heterodimer, is a beta1 integrin implicated in therapy resistance in both solid tumors and haematological malignancies such as chronic lymphocytic leukemia (CLL). A complex inside-out signaling mechanism activates VLA-4, which might include several therapeutic targets for CLL. Treatment regimens for this disease have recently shifted towards novel agents targeting BCR signaling. Bruton’s tyrosine kinase (BTK) is a component of B cell receptor signaling and BTK inhibitors such as ibrutinib are highly successful; however, their limitations include indefinite drug administration, the development of therapy resistance, and toxicities. VLA-4 might be activated independently of BTK, resulting in an ongoing interaction of CD49d-expressing leukemic cells with their surrounding tissue, which may reduce the success of therapy with BTK inhibitors and increases the need for alternative therapies. In this context, we discuss the inside-out signaling cascade culminating in VLA-4 activation, consider the advantages and disadvantages of BTK inhibitors in CLL and elucidate the mechanisms behind cell adhesion-mediated drug resistance.
Collapse
|
4
|
Jung B, Ferrer G, Chiu PY, Aslam R, Ng A, Palacios F, Wysota M, Cardillo M, Kolitz JE, Allen SL, Barrientos JC, Rai KR, Chiorazzi N, Sherry B. Activated CLL cells regulate IL17F producing Th17 cells in miR155 dependent and outcome specific manners. JCI Insight 2022; 7:158243. [PMID: 35511436 DOI: 10.1172/jci.insight.158243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) results from expansion of a CD5+ B-cell clone that requires interactions with other cell types, including T cells. Moreover, CLL patients have elevated circulating IL17A+ and IL17F+ CD4+ T cells (Th17s), with higher IL17A+Th17s correlating with better outcomes. We report that CLL Th17s express more miR155, a Th17 differentiation regulator, than control Th17s, despite naïve CD4+ T cell (TN) basal miR155 levels being similar in both. We also found that CLL cells directly regulate miR155 levels in TN, thereby affecting Th17 differentiation by documenting that: co-culturing TN with resting (Brest) or activated (Bact) CLL cells alters the magnitude and direction of T-cell miR155 levels; CLL Bact promote IL17A+ and IL17F+ T cell generation by a miR155-dependent mechanism, confirmed by miR155 inhibition; co-cultures of TN with CLL Bact lead to a linear correlation between the degree and direction of T-cell miR155 expression changes and IL17F production, but not IL17A; Bact-mediated changes in TN miR155 expression correlate with outcome, irrespective of IGHV mutation status, a strong prognostic indicator. Together, the results identify a previously unrecognized CLL Bact-dependent mechanism, upregulation of TN miR155 expression and subsequent enhancement of IL17F+ Th17 generation, that favors better clinical courses.
Collapse
Affiliation(s)
- Byeongho Jung
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Gerardo Ferrer
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Pui Yan Chiu
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Insitute for Medical Research, Manhasset, United States of America
| | - Rukhsana Aslam
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Anita Ng
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Florencia Palacios
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Michael Wysota
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Martina Cardillo
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Jonathan E Kolitz
- Department of Medicine, Northwell Health, New Hyde Park, United States of America
| | - Steven L Allen
- Department of Medicine, Northwell Health, New Hyde Park, United States of America
| | | | - Kanti R Rai
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Barbara Sherry
- Center for Immunology & Inflammation, Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Manhasset, United States of America
| |
Collapse
|
5
|
Palacios F, Yan XJ, Ferrer G, Chen SS, Vergani S, Yang X, Gardner J, Barrientos JC, Rock P, Burack R, Kolitz JE, Allen SL, Kharas MG, Abdel-Wahab O, Rai KR, Chiorazzi N. Musashi 2 influences chronic lymphocytic leukemia cell survival and growth making it a potential therapeutic target. Leukemia 2021; 35:1037-1052. [PMID: 33504942 PMCID: PMC8024198 DOI: 10.1038/s41375-020-01115-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/04/2020] [Accepted: 12/14/2020] [Indexed: 01/30/2023]
Abstract
Progression of chronic lymphocytic leukemia (CLL) results from the expansion of a small fraction of proliferating leukemic B cells. When comparing the global gene expression of recently divided CLL cells with that of previously divided cells, we found higher levels of genes involved in regulating gene expression. One of these was the oncogene Musashi 2 (MSI2), an RNA-binding protein that induces or represses translation. While there is an established role for MSI2 in normal and malignant stem cells, much less is known about its expression and role in CLL. Here we report for the first time ex vivo and in vitro experiments that MSI2 protein levels are higher in dividing and recently divided leukemic cells and that downregulating MSI2 expression or blocking its function eliminates primary human and murine CLL and mature myeloid cells. Notably, mature T cells and hematopoietic stem and progenitor cells are not affected. We also confirm that higher MSI2 levels correlate with poor outcome markers, shorter time-to-first-treatment, and overall survival. Thus, our data highlight an important role for MSI2 in CLL-cell survival and proliferation and associate MSI2 with poor prognosis in CLL patients. Collectively, these findings pinpoint MSI2 as a potentially valuable therapeutic target in CLL.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents
- Apoptosis/drug effects
- Biomarkers, Tumor
- Caspase 3/metabolism
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Survival/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Disease Models, Animal
- Gene Expression
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Gene Knockdown Techniques
- Humans
- Immunophenotyping
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Molecular Targeted Therapy
- Prognosis
- RNA, Small Interfering
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Florencia Palacios
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xiao-Jie Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Stefano Vergani
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xuejing Yang
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Gardner
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaqueline C Barrientos
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Philip Rock
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Richard Burack
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Jonathan E Kolitz
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Steven L Allen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Michael G Kharas
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kanti R Rai
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Northwell Health, Manhasset and New Hyde Park, New York, NY, USA.
| |
Collapse
|
6
|
Ibrutinib and venetoclax target distinct subpopulations of CLL cells: implication for residual disease eradication. Blood Cancer J 2021; 11:39. [PMID: 33602908 PMCID: PMC7893066 DOI: 10.1038/s41408-021-00429-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/21/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Ibrutinib inhibits Bruton tyrosine kinase while venetoclax is a specific inhibitor of the anti-apoptotic protein BCL2. Both drugs are highly effective as monotherapy against chronic lymphocytic leukemia (CLL), and clinical trials using the combination therapy have produced remarkable results in terms of rate of complete remission and frequency of undetectable minimal residual disease. However, the laboratory rationale behind the success of the drug combination is still lacking. A better understanding of how these two drugs synergize would eventually help develop other rational combination strategies. Using an ex vivo model that promotes CLL proliferation, we show that modeled ibrutinib proliferative responses, but not viability responses, correlate well with patients’ actual clinical responses. Importantly, we demonstrate for the first time that ibrutinib and venetoclax act on distinct CLL subpopulations that have different proliferative capacities. While the dividing subpopulation of CLL responds to ibrutinib, the resting subpopulation preferentially responds to venetoclax. The combination of these targeted therapies effectively reduced both the resting and dividing subpopulations in most cases. Our laboratory findings help explain several clinical observations and contribute to the understanding of tumor dynamics. Additionally, our proliferation model may be used to identify novel drug combinations with the potential of eradicating residual disease.
Collapse
|
7
|
MNDA controls the expression of MCL-1 and BCL-2 in chronic lymphocytic leukemia cells. Exp Hematol 2020; 88:68-82.e5. [PMID: 32682001 DOI: 10.1016/j.exphem.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023]
Abstract
The myeloid nuclear differentiation antigen (MNDA) is a stress-induced protein that promotes degradation of the anti-apoptotic factor MCL-1 and apoptosis in myeloid cells. MNDA is also expressed in normal lymphoid cells and in B-cell clones isolated from individuals with chronic lymphocytic leukemia (CLL), a disease characterized by abnormal apoptosis control. We found that MNDA expression levels inversely correlate with the amount of the anti-apoptotic proteins MCL-1 and BCL-2 in human CLL samples. We report that in response to chemotherapeutic agents that induce genotoxic stress, MNDA exits its typical nucleolar localization and accumulates in the nucleoplasm of CLL and lymphoid cells. Then, MNDA binds chromatin at Mcl1 and Bcl2 genes and affects the transcriptional competence of RNA polymerase II. Our data also reveal that MNDA specifically associates with Mcl1 and Bcl2 (pre-) mRNAs and favors their rapid turnover as a prompt response to genotoxic stress. We propose that this rapid dynamic tuning of RNA levels, which leads to the destabilization of Mcl1 and Bcl2 transcripts, represents a post-transcriptional mechanism of apoptosis control in CLL cells. These results provide an explanation of previous clinical data and corroborate the finding that higher MNDA expression levels in CLL are associated with a better clinical course.
Collapse
|
8
|
VLA-4 Expression and Activation in B Cell Malignancies: Functional and Clinical Aspects. Int J Mol Sci 2020; 21:ijms21062206. [PMID: 32210016 PMCID: PMC7139737 DOI: 10.3390/ijms21062206] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Lineage commitment and differentiation of hematopoietic cells takes place in well-defined microenvironmental surroundings. Communication with other cell types is a vital prerequisite for the normal functions of the immune system, while disturbances in this communication support the development and progression of neoplastic disease. Integrins such as the integrin very late antigen-4 (VLA-4; CD49d/CD29) control the localization of healthy as well as malignant B cells within the tissue, and thus determine the patterns of organ infiltration. Malignant B cells retain some key characteristics of their normal counterparts, with B cell receptor (BCR) signaling and integrin-mediated adhesion being essential mediators of tumor cell homing, survival and proliferation. It is thus not surprising that targeting the BCR pathway using small molecule inhibitors has proved highly effective in the treatment of B cell malignancies. Attenuation of BCR-dependent lymphoma–microenvironment interactions was, in this regard, described as a main mechanism critically contributing to the efficacy of these agents. Here, we review the contribution of VLA-4 to normal B cell differentiation on the one hand, and to the pathophysiology of B cell malignancies on the other hand. We describe its impact as a prognostic marker, its interplay with BCR signaling and its predictive role for novel BCR-targeting therapies, in chronic lymphocytic leukemia and beyond.
Collapse
|
9
|
Rosati E, Baldoni S, De Falco F, Del Papa B, Dorillo E, Rompietti C, Albi E, Falzetti F, Di Ianni M, Sportoletti P. NOTCH1 Aberrations in Chronic Lymphocytic Leukemia. Front Oncol 2018; 8:229. [PMID: 29998084 PMCID: PMC6030253 DOI: 10.3389/fonc.2018.00229] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/05/2018] [Indexed: 01/13/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable B-cell neoplasm characterized by highly variable clinical outcomes. In recent years, genomic and molecular studies revealed a remarkable heterogeneity in CLL, which mirrored the clinical diversity of this disease. These studies profoundly enhanced our understanding of leukemia cell biology and led to the identification of new biomarkers with potential prognostic and therapeutic significance. Accumulating evidence indicates a key role of deregulated NOTCH1 signaling and NOTCH1 mutations in CLL. This review highlights recent discoveries that improve our understanding of the pathophysiological NOTCH1 signaling in CLL and the clinical impact of NOTCH1 mutations in retrospective and prospective trials. In addition, we discuss the rationale for a therapeutic strategy aiming at inhibiting NOTCH1 signaling in CLL, along with an overview on the currently available NOTCH1-directed approaches.
Collapse
Affiliation(s)
- Emanuela Rosati
- Department of Experimental Medicine, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| | - Stefano Baldoni
- Department of Life, Hematology Section, Health and Environmental Sciences, University of L'Aquila, Perugia, Italy
| | - Filomena De Falco
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| | - Beatrice Del Papa
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| | - Erica Dorillo
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| | - Chiara Rompietti
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| | - Elisa Albi
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| | - Franca Falzetti
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Aging Sciences, University of Chieti Pescara, Chieti, Italy.,Department of Hematology, Transfusion Medicine and Biotechnologies, Ospedale Civile, Pescara, Italy
| | - Paolo Sportoletti
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
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
|
11
|
Ganghammer S, Hutterer E, Hinterseer E, Brachtl G, Asslaber D, Krenn PW, Girbl T, Berghammer P, Geisberger R, Egle A, Zucchetto A, Kruschinski A, Gattei V, Chigaev A, Greil R, Hartmann TN. CXCL12-induced VLA-4 activation is impaired in trisomy 12 chronic lymphocytic leukemia cells: a role for CCL21. Oncotarget 2016; 6:12048-60. [PMID: 25895128 PMCID: PMC4494922 DOI: 10.18632/oncotarget.3660] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/27/2015] [Indexed: 12/18/2022] Open
Abstract
Homing to distinct lymphoid organs enables chronic lymphocytic leukemia (CLL) cells to receive pro-survival and proliferative signals. Cytogenetic aberrations can significantly affect CLL cell compartmentalization. Trisomy 12 (tri12) defines a CLL subgroup with specific clinical features and increased levels of the negative prognostic marker CD49d, the α4-subunit of the integrin VLA-4, which is a key regulator of CLL cell homing to bone marrow (BM). Chemokine-induced inside-out VLA-4 activation, particularly via the CXCL12-CXCR4 axis, increases the arrest of various cell types on VCAM-1 presenting endothelium. Here, we demonstrate that high CD49d expression in tri12 CLL is accompanied by decreased CXCR4 expression. Dissecting functional consequences of these alterations, we observed that tri12 CLL cell homing to murine BM is not affected by CXCR4-CXCL12 blockage using AMD3100 or olaptesed pegol/NOX-A12. In line, CCL21-CCR7 rather than CXCL12-CXCR4 interactions triggered VLA-4-mediated arrests of tri12 CLL cells to VCAM-1 under blood flow conditions. Concordantly, in real-time kinetic analyses we found CCL21 but not CXCL12 being capable to induce inside-out VLA-4 conformational changes in this CLL subgroup. Our results provide novel insights into the peculiar clinico-biological behaviour of tri12 CLL and emphasize its specific chemokine and integrin utilization during pathophysiologically and therapeutically relevant interactions with the microenvironment.
Collapse
Affiliation(s)
- Sylvia Ganghammer
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Evelyn Hutterer
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Elisabeth Hinterseer
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Gabriele Brachtl
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Daniela Asslaber
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Peter William Krenn
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Tamara Girbl
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Petra Berghammer
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Roland Geisberger
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Alexander Egle
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico, Aviano, Italy
| | | | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico, Aviano, Italy
| | - Alexandre Chigaev
- Department of Pathology and Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Richard Greil
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| | - Tanja Nicole Hartmann
- Laboratory for Immunological and Molecular Cancer Research, 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Austria.,Salzburg Cancer Research Institute, Salzburg, Austria
| |
Collapse
|
12
|
Poret N, Fu Q, Guihard S, Cheok M, Miller K, Zeng G, Quesnel B, Troussard X, Galiègue-Zouitina S, Shelley CS. CD38 in Hairy Cell Leukemia Is a Marker of Poor Prognosis and a New Target for Therapy. Cancer Res 2016; 75:3902-11. [PMID: 26170397 DOI: 10.1158/0008-5472.can-15-0893] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hairy cell leukemia (HCL) is characterized by underexpression of the intracellular signaling molecule RhoH. Reconstitution of RhoH expression limits HCL pathogenesis in a mouse model, indicating this could represent a new therapeutic strategy. However, while RhoH reconstitution is theoretically possible as a therapy, it is technically immensely challenging as an appropriately functional RhoH protein needs to be specifically targeted. Because of this problem, we sought to identify druggable proteins on the HCL surface that were dependent upon RhoH underexpression. One such protein was identified as CD38. Analysis of 51 HCL patients demonstrated that 18 were CD38-positive. Interrogation of the clinical record of 23 relapsed HCL patients demonstrated those that were CD38-positive had a mean time to salvage therapy 71 months shorter than patients who were CD38-negative. Knockout of the CD38 gene in HCL cells increased apoptosis, inhibited adherence to endothelial monolayers, and compromised ability to produce tumors in vivo. Furthermore, an anti-CD38 antibody proved effective against pre-existing HCL tumors. Taken together, our data indicate that CD38 expression in HCL drives poor prognosis by promoting survival and heterotypic adhesion. Our data also indicate that CD38-positive HCL patients might benefit from treatments based on CD38 targeting.
Collapse
Affiliation(s)
- Nicolas Poret
- Institut National de la Santé et de la Recherche Medicale UMR-S1172, Centre Jean-Pierre Aubert, Institut pour la Recherche sur le Cancer de Lille and Université de Lille, Lille, France
| | - Qiangwei Fu
- Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse, Wisconsin
| | - Soizic Guihard
- Institut National de la Santé et de la Recherche Medicale UMR-S1172, Centre Jean-Pierre Aubert, Institut pour la Recherche sur le Cancer de Lille and Université de Lille, Lille, France
| | - Meyling Cheok
- Institut National de la Santé et de la Recherche Medicale UMR-S1172, Centre Jean-Pierre Aubert, Institut pour la Recherche sur le Cancer de Lille and Université de Lille, Lille, France
| | - Katie Miller
- Department of Biology, Saint Mary's University of Minnesota, Winona, Minnesota
| | - Gordon Zeng
- Department of Pathology, Gundersen Health System, La Crosse, Wisconsin
| | - Bruno Quesnel
- Institut National de la Santé et de la Recherche Medicale UMR-S1172, Centre Jean-Pierre Aubert, Institut pour la Recherche sur le Cancer de Lille and Université de Lille, Lille, France. Service des Maladies du Sang, Hôpital Huriez, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Xavier Troussard
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Sylvie Galiègue-Zouitina
- Institut National de la Santé et de la Recherche Medicale UMR-S1172, Centre Jean-Pierre Aubert, Institut pour la Recherche sur le Cancer de Lille and Université de Lille, Lille, France.
| | - Carl Simon Shelley
- Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse, Wisconsin.
| |
Collapse
|
13
|
Kellner J, Wierda W, Shpall E, Keating M, McNiece I. Isolation of a novel chronic lymphocytic leukemic (CLL) cell line and development of an in vivo mouse model of CLL. Leuk Res 2016; 40:54-9. [PMID: 26601610 PMCID: PMC11770973 DOI: 10.1016/j.leukres.2015.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/28/2015] [Accepted: 10/14/2015] [Indexed: 11/29/2022]
Abstract
Leukemic cell lines have become important tools for studies of disease providing a monoclonal cell population that can be extensively expanded in vitro while preserving leukemic cellular characteristics. However, studies of chronic lymphocytic leukemia (CLL) have been impeded in part by the lack of continuous human cell lines. CLL cells have a high spontaneous apoptosis rate in vitro and exhibit minimal proliferation in xenograft models. Therefore, there is a need for development of primary CLL cell lines and we describe the isolation of such a line from the bone marrow of a CLL patient (17p deletion and TP53 mutation) which has been in long term culture for more than 12 months with continuous proliferation. The CLL cell line (termed MDA-BM5) which was generated in vitro with continuous co-culture on autologous stromal cells is CD19+CD5+ and shows an identical pattern of somatic hypermutation as determined in the patient's bone marrow (BM), confirming the origin of the cells from the original CLL clone. MDA-BM5 cells were readily transplantable in NOD/SCID gamma null mice (NSG) with disease developing in the BM, liver and spleen. BM cells from quaternary serial transplantation in NSG mice demonstrated the presence of CD19+CD5+ cells with Ig restricted to lambda which is consistent with the original patient cells. These studies describe a new CLL cell line from a patient with del(17p) that provides a unique model for in vitro and in vivo studies.
Collapse
Affiliation(s)
- Joshua Kellner
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, MD Anderson Cancer Centre, Houston, TX, United States
| | - William Wierda
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, MD Anderson Cancer Centre, Houston, TX, United States
| | - Michael Keating
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Ian McNiece
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, MD Anderson Cancer Centre, Houston, TX, United States.
| |
Collapse
|
14
|
Lamanna N. New oral small molecules in the treatment of chronic lymphocytic leukemia. Cancer 2015; 121:1917-26. [PMID: 25690403 DOI: 10.1002/cncr.29130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 09/15/2014] [Accepted: 10/07/2014] [Indexed: 12/19/2022]
Abstract
There has been a dramatic change in therapy for chronic lymphocytic leukemia (CLL) over the last 20 years. In 1990, available therapy produced complete responses in <5% of treated patients. This is in marked contrast to modern regimens, which are reported to reliably produce complete responses in approximately 40% to 50% of patients. This remarkable improvement has been attributable to combination chemoimmunotherapy agents that have contributed to the backbone of therapy for patients with CLL. However, the disease is still incurable and these modern treatment regimens have been somewhat limited to the treatment of younger, physically "fit" patients with CLL due to their increased toxicity, including enhanced myelosuppression and immunosuppression. In addition, because patients receive multiple therapies during the course of their lifetime, the mounting toxicities as well as decreased efficacy often limit the repeated use of these more aggressive combination therapies. Fortunately, over the past 5 years, there has been an explosion of new active agents that have demonstrated remarkable activity in patients with recurrent/refractory disease as well as those who harbor poor cytogenetic abnormalities. The current review focuses on some of the novel small molecules that have either been approved or are at the forefront of clinical development in the treatment of patients with CLL.
Collapse
Affiliation(s)
- Nicole Lamanna
- Leukemia Service, Department of Medicine, Columbia University Medical Center, New York, New York
| |
Collapse
|
15
|
Messmer D, Lorrain K, Stebbins K, Bravo Y, Stock N, Cabrera G, Correa L, Chen A, Jacintho J, Chiorazzi N, Yan XJ, Spaner D, Prasit P, Lorrain D. A Selective Novel Peroxisome Proliferator-Activated Receptor (PPAR)-α Antagonist Induces Apoptosis and Inhibits Proliferation of CLL Cells In Vitro and In Vivo. Mol Med 2015; 21:410-9. [PMID: 26070013 DOI: 10.2119/molmed.2015.00139] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/04/2015] [Indexed: 01/10/2023] Open
Abstract
Tumor-specific metabolic changes can reveal new therapeutic targets. Our findings implicate a supporting role for fatty acid metabolism in chronic lymphocytic leukemia (CLL) cell survival. Peroxisome proliferator-activated receptor (PPAR)-α, a major transcriptional regulator of fatty acid oxidation, was recently shown to be upregulated in CLL. To evaluate PPARα as a potential therapeutic target, we developed a highly selective, potent small molecule antagonist of PPARα, NXT629. NXT629 inhibited agonist-induced transcription of PPARα-regulated genes, demonstrating target engagement in CLL cells. Furthermore, NXT629 induced apoptosis of CLL cells even in the presence of a protective microenvironment. To mimic the proliferative lymphoid compartment of CLL, we examined the activity of NXT629 on CLL cells that were stimulated to proliferate in vitro. NXT629 reduced the number of leukemia cells undergoing cell division. In addition, in two xenograft mouse models of CLL (one a model for nondividing and one for dividing CLL), NXT629 reduced the number of viable CLL cells in vivo. Overall, these results suggest that fatty acid metabolism promotes survival and proliferation of primary CLL cells and that inhibiting PPARα gene regulation could be a new therapeutic approach to treating CLL.
Collapse
Affiliation(s)
- Davorka Messmer
- Inception Sciences, San Diego, California, United States of America
| | - Kymmy Lorrain
- Inception Sciences, San Diego, California, United States of America
| | - Karin Stebbins
- Inception Sciences, San Diego, California, United States of America
| | - Yalda Bravo
- Inception Sciences, San Diego, California, United States of America
| | - Nicholas Stock
- Inception Sciences, San Diego, California, United States of America
| | | | - Lucia Correa
- Inception Sciences, San Diego, California, United States of America
| | - Austin Chen
- Inception Sciences, San Diego, California, United States of America
| | - Jason Jacintho
- Inception Sciences, San Diego, California, United States of America
| | - Nicholas Chiorazzi
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York, United States of America
| | - Xiao Jie Yan
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York, United States of America
| | - David Spaner
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Peppi Prasit
- Inception Sciences, San Diego, California, United States of America
| | - Daniel Lorrain
- Inception Sciences, San Diego, California, United States of America
| |
Collapse
|
16
|
Filip AA, Ciseł B, Wąsik-Szczepanek E. Guilty bystanders: nurse-like cells as a model of microenvironmental support for leukemic lymphocytes. Clin Exp Med 2015; 15:73-83. [PMID: 24337970 PMCID: PMC4308641 DOI: 10.1007/s10238-013-0268-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 11/29/2013] [Indexed: 12/03/2022]
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is one of the most common leukemias among the elderly and, despite many efforts, still stays incurable. Recent studies point to the microenvironment as the critical factor providing leukemic lymphocytes with pro-survival signals. Thus, the neighboring cells appear to be a perfect target for antileukemic therapy. Nurse-like cells (NLCs) largely contribute to CLL microenvironmental support. We developed the CLL lymphocyte/NLC co-culture model for the investigation of microenvironmental interactions. Viability and apoptosis were investigated in CLL lymphocytes treated with dexamethasone (DEX) and chlorambucil (CLB), with and without NLCs' support. For the first time, the capacity of DEX and CLB to affect NLCs viability was also evaluated. Apoptosis-associated gene expression profiles of leukemic lymphocytes ex vivo and cultured with NLCs were assessed by expression arrays. CLL lymphocytes escaped spontaneous apoptosis for several months when cultured with NLCs. The presence of NLCs significantly reduced apoptosis induced with DEX and CLB (p < 0.001; p = 0.012, respectively), and their protective effect was more evident than the effect of recombinant SDF1. Both DEX and CLB also decreased NLCs viability, but to a lesser extent (mean viability in DEX-treated cultures was 37.79% in NLCs compared to 29.24% in lymphocytes). NLCs induced the expression of important anti-apoptotic genes in cultured CLL lymphocytes; median expression of BCL2, SURVIVIN, BCL2A1, and XIAP was significantly higher as compared to ex vivo status. The CLL lymphocyte/NLC co-culture makes up the convenient and close to the natural-state model for studying the relationship between leukemic cells and the microenvironment. Direct cell-to-cell contact with NLCs increases the expression of anti-apoptotic genes in CLL lymphocytes, thus protecting them against induced apoptosis. As the effect of antileukemic drugs is not so apparent in NLCs, the combined therapy targeted at both lymphocytes and the microenvironment should be considered for CLL patients. Simultaneous aiming at the disruption of several different signaling pathways and/or anti-apoptotic proteins may further improve treatment efficiency.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Apoptosis/drug effects
- B-Lymphocytes/drug effects
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Bystander Effect/genetics
- Cell Survival/drug effects
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Chlorambucil/pharmacology
- Coculture Techniques
- Dexamethasone/pharmacology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Minor Histocompatibility Antigens
- Monocytes/drug effects
- Monocytes/metabolism
- Monocytes/pathology
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- X-Linked Inhibitor of Apoptosis Protein/genetics
- X-Linked Inhibitor of Apoptosis Protein/metabolism
Collapse
Affiliation(s)
- Agata A Filip
- Department of Cancer Genetics, Medical University of Lublin, Radziwillowska 11, 20-080, Lublin, Poland,
| | | | | |
Collapse
|
17
|
Wiestner A. BCR pathway inhibition as therapy for chronic lymphocytic leukemia and lymphoplasmacytic lymphoma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:125-134. [PMID: 25696845 DOI: 10.1182/asheducation-2014.1.125] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Chronic lymphocytic leukemia (CLL) and lymphoplasmacytic lymphoma (LPL) are malignancies of mature B cells. In LPL, mutations of the adaptor protein MYD88 (L265P) in the Toll-like receptor pathway have been recognized recently as being a hallmark of the disease and indicate a dependence of the tumor on this pathway. In CLL, functional studies have implicated BCR activation in the tissue microenvironment as a pivotal pathway in the pathogenesis. Bruton's tyrosine kinase (BTK) and the PI3Kδ isoform are essential for BCR signaling and also seem to be required for signal transduction in LPL cells, even if the role of BCR signaling in this disease remains less well defined. Ibrutinib, a covalent inhibitor of BTK approved by the Food and Drug Administration as a second-line treatment for CLL, and idelalisib, a selective inhibitor of PI3Kδ, achieve excellent clinical responses in both diseases irrespective of classic markers indicating high-risk disease. Several additional inhibitors targeting BTK and PI3Kδ, as well as the spleen tyrosine kinase, have entered clinical trials. This review discusses the biologic basis for kinase inhibitors as targeted therapy for CLL and LPL and summarizes the clinical experience with these agents.
Collapse
Affiliation(s)
- Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
18
|
Development and characterization of a physiologically relevant model of lymphocyte migration in chronic lymphocytic leukemia. Blood 2014; 123:3607-17. [PMID: 24637360 DOI: 10.1182/blood-2013-12-544569] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
There is growing evidence that lymphocyte trafficking contributes to the clinical course of chronic lymphocytic leukemia (CLL), but to date, only static in vitro cultures have been used to study these phenomena. To address this lack of data, we have developed a dynamic in vitro model in which CLL cells experience shear forces equivalent to those in capillary beds and are made to flow through capillary-like hollow fibers lined with endothelial cells. CLL cells treated in this way increased their expression of CD62L and CXCR4 (both P < .0001) and of CD49d and CD5 (both P = .003) directly as a result of the shear force. Furthermore, CLL cells migrated through the endothelium into the "extravascular" space (mean migration, 1.37% ± 2.14%; n = 21). Migrated CLL cells had significantly higher expression of CD49d (P = .02), matrix metallopeptidase-9 (P = .004), CD38 (P = .009), CD80 (P = .04), and CD69 (P = .04) compared with CLL cells that remained in the circulation. The degree of migration observed strongly correlated with CD49d expression (r(2), 0.47; P = .01), and treatment with the CD49d-blocking antibody natalizumab resulted in significantly decreased migration (P = .01). Taken together, our data provide evidence for a novel, dynamic, and tractable in vitro model of lymphocyte migration and confirm that CD49d is a critical regulator of this process in CLL.
Collapse
|
19
|
Hill RJ, Lou Y, Tan SL. B-cell antigen receptor signaling in chronic lymphocytic leukemia: therapeutic targets and translational opportunities. Int Rev Immunol 2014; 32:377-96. [PMID: 23886341 DOI: 10.3109/08830185.2013.818141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
B-cell chronic lymphocytic leukemia (CLL) is characterized by clonally expanded and molecularly heterogeneous populations of B lymphocytes with impaired apoptotic mechanisms. This occurs as a result of multiple genetic and epigenetic abnormalities, including chromosomal aberrations and enhancer region hypomethylation, often impinging on intracellular signaling pathways that are essential to normal B-cell activation, proliferation, and survival. The B-cell antigen receptor (BCR) signaling is one such pathway usurped by malignant B cells, as exemplified by the early phase clinical success achieved by small-molecule agents targeting key players involved in the pathway. Such new targeted agents, including those that inhibit the function of Spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), phosphatidylinositol 3-kinases (PI3K), and B-cell lymphoma 2 (BCL-2), along with the current standard therapy comprising chemo-immunotherapies with or without B-cell depleting biologic agent rituximab (anti-CD20 monoclonal antibody), should expand the armamentarium for CLL therapy. We review the therapeutic agents currently in clinical development which target different effectors of the malignant BCR signaling, and discuss their overlapping and discriminating translational opportunities in the context of CLL treatment.
Collapse
Affiliation(s)
- Ronald J Hill
- Principia Biopharma, South San Francisco, CA 94080, USA.
| | | | | |
Collapse
|
20
|
|
21
|
Chavez JC, Sahakian E, Pinilla-Ibarz J. Ibrutinib: an evidence-based review of its potential in the treatment of advanced chronic lymphocytic leukemia. CORE EVIDENCE 2013; 8:37-45. [PMID: 23717217 PMCID: PMC3662532 DOI: 10.2147/ce.s34068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Indexed: 12/28/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a heterogeneous disease with a variable course, and remains an incurable disease. Frequent relapses and eventual resistance to fludarabine characterize symptomatic CLL and portends a dismal prognosis for patients. Growing evidence has shown that signaling pathways such as the B cell receptor and NFkB are implicated in the survival and proliferation of the CLL cells which are ultimately associated with persistence of the disease. The Bruton's tyrosine kinase pathway regulates downstream activation of the B cell receptor and has emerged as an attractive target. Ibrutinib inhibits the Bruton's tyrosine kinase pathway, and consequently induces apoptosis of B cells. Phase I and II studies have shown impressive response rates with an excellent safety profile in patients with refractory/relapsed CLL and elderly treatment-naïve CLL patients. This paper reviews the preclinical and clinical data for ibrutinib when used in the treatment of CLL. Recent studies showing the benefit of combination therapy using ibrutinib, monoclonal antibodies, and chemoimmunotherapy are also discussed.
Collapse
Affiliation(s)
- Julio C Chavez
- H Lee Moffitt Cancer and Research Institute, Division of Malignant Hematology, and University of South Florida, Tampa, FL, USA
| | | | | |
Collapse
|
22
|
Modeling tumor-host interactions of chronic lymphocytic leukemia in xenografted mice to study tumor biology and evaluate targeted therapy. Leukemia 2013; 27:2311-21. [PMID: 23619564 PMCID: PMC4126654 DOI: 10.1038/leu.2013.131] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 01/06/2023]
Abstract
Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental factors for proliferation and survival. In particular, the B-cell receptor (BCR) and NF-κB pathways are activated in the lymph node microenvironment. Thus, model systems mimicking tumor-host interactions are important tools to study CLL biology and pathogenesis. We investigated whether the recently established NOD/scid/γcnull (NSG) mouse xenograft model can recapitulate the effects of the human microenvironment. We assessed, therefore, tumor characteristics previously defined in lymph node-resident CLL cells, including proliferation, and activation of the BCR and NF-κB pathways. We found that the murine spleen microenvironment supported CLL cell proliferation and activation to a similar degree than the human lymph node, including induction of BCR and NF-κB signaling in the xenografted cells. Next, we used this model to study ibrutinib, a Bruton's tyrosine kinase inhibitor in clinical development. Ibrutinib inhibited BCR and NF-κB signaling induced by the microenvironment, decreased proliferation, induced apoptosis, and reduced the tumor burden in vivo. Thus, our data demonstrate that the spleen of xenografted NSG mice can, in part, recapitulate the role of the human lymph node for CLL cells. In addition, we show that ibrutinib effectively disrupts tumor-host interactions essential for CLL cell proliferation and survival in vivo.
Collapse
|
23
|
Filip AA, Ciseł B, Koczkodaj D, Wąsik-Szczepanek E, Piersiak T, Dmoszyńska A. Circulating microenvironment of CLL: are nurse-like cells related to tumor-associated macrophages? Blood Cells Mol Dis 2013; 50:263-70. [PMID: 23313631 DOI: 10.1016/j.bcmd.2012.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/12/2012] [Indexed: 11/28/2022]
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is one of the most common hematologic malignancies in Western countries. Accumulation of leukemic lymphocytes in peripheral blood, bone marrow and secondary lymphatic organs of CLL patients is due to decreased apoptosis rather than to increased proliferation. The former is driven by signals from a specific microenvironment, created by stromal cells of mesenchymal origin, follicular dendritic cells, T lymphocytes and others. Nurse-like cells (NLCs) were first described to differentiate from peripheral blood mononuclear cells of CLL patients in vitro, then they have been also found in proliferation centers of their lymphatic tissues. Like tumor-associated macrophages (TAMs) in solid tumors, nurse-like cells promote survival of CLL lymphocytes. NLC gene expression patterns suggest their similarity to TAMs and differ between patients depending on ZAP70 protein expression status. NLC number in vitro corresponds with CD14 expressing cell count and beta-2-microglobulin serum level, and positively correlates with leukemic lymphocyte viability. As NLCs strongly express genes for adhesion molecules and secrete chemokines of antiapoptotic activity, they should be considered as a target for anti-microenvironment therapy of this incurable disease.
Collapse
Affiliation(s)
- Agata A Filip
- Department of Cancer Genetics, Medical University of Lublin, Radziwillowska 11, 20-080 Lublin, Poland.
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
AbstractChronic lymphocytic leukemia (CLL) is a malignancy of mature B cells that depend on host factors in the tissue microenvironment for survival and proliferation. In vitro, CLL cells rapidly undergo apoptosis unless microenvironmental factors are provided that support their survival. Signaling pathways activated in the microenvironment in vivo include the B-cell receptor (BCR) and NF-κB pathways. Thus, CLL is a disease “addicted to the host” and is dependent on pathways that promote normal B-cell development, expansion, and survival; this is particularly true in the case of the BCR signaling cascade. Small-molecule inhibitors of kinases that are essential for BCR signal transduction abrogate the stimulating effects of the microenvironment on CLL cells. The orally administered tyrosine kinase inhibitors fostamatinib and ibrutinib and the phosphatidylinositol 3-kinase inhibitor GS-1101 have induced impressive responses in relapsed and refractory CLL patients, mostly with moderate side effects. Reductions in lymphadenopathy and splenomegaly are seen within weeks and are frequently accompanied by a transient rise in absolute lymphocyte count that is asymptomatic and probably the result of changes in CLL cell trafficking. This review discusses the biologic basis for kinase inhibitors as targeted therapy of CLL and summarizes the exciting early clinical experience with these agents.
Collapse
|
25
|
Girbl T, Hinterseer E, Grössinger EM, Asslaber D, Oberascher K, Weiss L, Hauser-Kronberger C, Neureiter D, Kerschbaum H, Naor D, Alon R, Greil R, Hartmann TN. CD40-Mediated Activation of Chronic Lymphocytic Leukemia Cells Promotes Their CD44-Dependent Adhesion to Hyaluronan and Restricts CCL21-Induced Motility. Cancer Res 2012; 73:561-70. [DOI: 10.1158/0008-5472.can-12-2749] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Audrito V, Vaisitti T, Serra S, Bologna C, Brusa D, Malavasi F, Deaglio S. Targeting the microenvironment in chronic lymphocytic leukemia offers novel therapeutic options. Cancer Lett 2012; 328:27-35. [PMID: 22910767 DOI: 10.1016/j.canlet.2012.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/10/2012] [Accepted: 08/13/2012] [Indexed: 01/13/2023]
Abstract
Chronic lymphocytic leukemia (CLL) cells display features consistent with a defect in apoptosis and exhibit prolonged survival in vivo. Survival of these malignant cells is influenced by interactions with non-leukemic cells located in permissive niches in lymphoid organs. Leukemic cells subvert the normal architecture of the lymphoid organs, recruiting stromal cells, dendritic cells and T lymphocytes, all reported as playing active roles in the survival and proliferation of CLL. The same survival-promoting environment also rescues/protects leukemic cells from cytotoxic therapies, giving way to disease relapse. This review summarizes and discusses current knowledge about the intricate network of soluble and cell-bound signals regulating the life and death of CLL cells in different districts. At the same time, it seeks to hone in on which discrete molecular elements are best suited as targets for treating this still incurable disease.
Collapse
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, School of Medicine, Turin, Italy; Human Genetics Foundation (HuGeF), Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy of mature B cells that depend on host factors in the tissue microenvironment for survival and proliferation. In vitro, CLL cells rapidly undergo apoptosis unless microenvironmental factors are provided that support their survival. Signaling pathways activated in the microenvironment in vivo include the B-cell receptor (BCR) and NF-κB pathways. Thus, CLL is a disease "addicted to the host" and is dependent on pathways that promote normal B-cell development, expansion, and survival; this is particularly true in the case of the BCR signaling cascade. Small-molecule inhibitors of kinases that are essential for BCR signal transduction abrogate the stimulating effects of the microenvironment on CLL cells. The orally administered tyrosine kinase inhibitors fostamatinib and ibrutinib and the phosphatidylinositol 3-kinase inhibitor GS-1101 have induced impressive responses in relapsed and refractory CLL patients, mostly with moderate side effects. Reductions in lymphadenopathy and splenomegaly are seen within weeks and are frequently accompanied by a transient rise in absolute lymphocyte count that is asymptomatic and probably the result of changes in CLL cell trafficking. This review discusses the biologic basis for kinase inhibitors as targeted therapy of CLL and summarizes the exciting early clinical experience with these agents.
Collapse
|
28
|
Gilling CE, Mittal AK, Chaturvedi NK, Iqbal J, Aoun P, Bierman PJ, Bociek RG, Weisenburger DD, Joshi SS. Lymph node-induced immune tolerance in chronic lymphocytic leukaemia: a role for caveolin-1. Br J Haematol 2012; 158:216-231. [PMID: 22571278 DOI: 10.1111/j.1365-2141.2012.09148.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 03/23/2012] [Indexed: 11/28/2022]
Abstract
Emerging evidence indicates that the tumour microenvironment (TME) regulates the behaviour of chronic lymphocytic leukaemia (CLL). However, the precise mechanism and molecules involved in this process remain unknown. Gene expression profiles of CLL cells from lymph node (LN), bone marrow (BM) and peripheral blood (PB) indicate overexpression of a tolerogenic signature in CLL cells in lymph nodes (LN-CLL). Based on their role in B cell biology, the progression of CLL, or immune regulation, a few genes of this 83-gene signature were selected for further analyses. We observed a significant correlation between the clinical outcomes and the expression of CAV1 (P = 0·041), FGFR1 isoform 8 (P = 0·032), PTPN6 (P = 0·031) and ZWINT (P < 0·001). CAV1, a molecule involved in the regulation of tumour progression in other cancers, was seven-fold higher in LN-CLL cells compared to BM- and PB-CLL cells. Knockdown of CAV1 expression in CLL cells resulted in significantly decreased migration (P = 0·016) and proliferation (P = 0·04). When CAV1 was knocked down in B and T cell lines, we observed an inability to form immune synapses. Furthermore, CAV1 knockdown in CLL cells impaired their ability to form immune synapses with autologous T lymphocytes and allogeneic, healthy T cells. Subsequent analyses of microarray data showed differential expression of cytoskeletal genes, specifically those involved in actin polymerization. Therefore, we report a novel role for CAV1 in tumour-induced immunosuppression during the progression of CLL.
Collapse
Affiliation(s)
- Christine E Gilling
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amit K Mittal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K Chaturvedi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Patricia Aoun
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Philip J Bierman
- Internal Medicine-Oncology/Hematology Section, University of Nebraska Medical Center, Omaha, NE, USA
| | - Robert G Bociek
- Internal Medicine-Oncology/Hematology Section, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dennis D Weisenburger
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shantaram S Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
29
|
Chapman CM, Sun X, Roschewski M, Aue G, Farooqui M, Stennett L, Gibellini F, Arthur D, Pérez-Galán P, Wiestner A. ON 01910.Na is selectively cytotoxic for chronic lymphocytic leukemia cells through a dual mechanism of action involving PI3K/AKT inhibition and induction of oxidative stress. Clin Cancer Res 2012; 18:1979-91. [PMID: 22351695 DOI: 10.1158/1078-0432.ccr-11-2113] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE Chronic lymphocytic leukemia (CLL), a malignancy of mature B cells, is incurable with chemotherapy. Signals from the microenvironment support leukemic cell survival and proliferation and may confer chemotherapy resistance. ON 01910.Na (Rigosertib), a multikinase phosphoinositide 3-kinase (PI3K) inhibitor, is entering phase III trials for myelodysplastic syndrome. Our aim was to analyze the efficacy of ON 01910.Na against CLL cells in vitro and investigate the molecular effects of this drug on tumor biology. EXPERIMENTAL DESIGN Cytotoxicity of ON 01910.Na against CLL cells from 34 patients was determined in vitro with flow cytometry of cells stained with Annexin V and CD19. Global gene expression profiling on Affymetrix microarrays, flow cytometry, Western blotting, and cocultures with stroma cells were used to delineate ON 01910.Na mechanism of action. RESULTS ON 01910.Na induced apoptosis in CLL B cells without significant toxicity against T cells or normal B cells. ON 01910.Na was equally active against leukemic cells associated with a more aggressive disease course [immunoglobulin heavy-chain variable region unmutated, adverse cytogenetics] than against cells without these features. Gene expression profiling revealed two main mechanisms of action: PI3K/AKT inhibition and induction of ROS that resulted in an oxidative stress response through activating protein 1 (AP-1), c-jun-NH(2)-terminal kinase, and ATF3 culminating in the upregulation of NOXA. ROS scavengers and shRNA mediated knockdown of ATF3- and NOXA-protected cells from drug-induced apoptosis. ON 01910.Na also abrogated the prosurvival effect of follicular dendritic cells on CLL cells and reduced SDF-1-induced migration of leukemic cells. CONCLUSIONS These data support the clinical development of ON 01910.Na in CLL.
Collapse
Affiliation(s)
- Colby M Chapman
- Hematology Branch, NHLBI, NIH, Bld 10, CRC 3-5140, 10 Center Drive, 20892-1202 Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Malavasi F, Deaglio S, Damle R, Cutrona G, Ferrarini M, Chiorazzi N. CD38 and chronic lymphocytic leukemia: a decade later. Blood 2011; 118:3470-8. [PMID: 21765022 PMCID: PMC3574275 DOI: 10.1182/blood-2011-06-275610] [Citation(s) in RCA: 157] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/28/2011] [Indexed: 11/20/2022] Open
Abstract
This review highlights a decade of investigations into the role of CD38 in CLL. CD38 is accepted as a dependable marker of unfavorable prognosis and as an indicator of activation and proliferation of cells when tested. Leukemic clones with higher numbers of CD38(+) cells are more responsive to BCR signaling and are characterized by enhanced migration. In vitro activation through CD38 drives CLL proliferation and chemotaxis via a signaling pathway that includes ZAP-70 and ERK1/2. Finally, CD38 is under a polymorphic transcriptional control after external signals. Consequently, CD38 appears to be a global molecular bridge to the environment, promoting survival/proliferation over apoptosis. Together, this evidence contributes to the current view of CLL as a chronic disease in which the host's microenvironment promotes leukemic cell growth and also controls the sequential acquisition and accumulation of genetic alterations. This view relies on the existence of a set of surface molecules, including CD38, which support proliferation and survival of B cells on their way to and after neoplastic transformation. The second decade of studies on CD38 in CLL will tell if the molecule is an effective target for antibody-mediated therapy in this currently incurable leukemia.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/genetics
- ADP-ribosyl Cyclase 1/metabolism
- ADP-ribosyl Cyclase 1/physiology
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Models, Biological
- Molecular Targeted Therapy/methods
- Molecular Targeted Therapy/trends
- Research/trends
- Time Factors
- Tumor Microenvironment/physiology
Collapse
Affiliation(s)
- Fabio Malavasi
- Department of Genetics, Biology and Biochemistry, University of Torino School of Medicine, Torino, Italy.
| | | | | | | | | | | |
Collapse
|
31
|
Ponzoni M, Doglioni C, Caligaris-Cappio F. Chronic lymphocytic leukemia: the pathologist's view of lymph node microenvironment. Semin Diagn Pathol 2011; 28:161-6. [PMID: 21842701 DOI: 10.1053/j.semdp.2011.02.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chronic lymphocytic leukemia (CLL), an indolent B-cell malignancy frequently diagnosed in the elderly, is characterized by the relentless accumulation of CD5+ monoclonal B cells that proliferate in the appropriate tissue microenvironments. Despite many advances achieved by molecular and functional studies, our knowledge of the reciprocal relationship between the CLL cell and its microenvironment at the tissue level is still largely incomplete. In this review we present the relevant current information on the tissue microenvironmental features of CLL, focusing on the events that appear to occur in the lymph node. Special attention is devoted to analyzing the properties of both neoplastic and nonneoplastic bystander cells within proliferation centers, the mysterious structures that likely represent the actual proliferative compartment.
Collapse
Affiliation(s)
- Maurilio Ponzoni
- Pathology Unit, Department of Oncology, University Scientific Institute San Raffaele, Milan, Italy.
| | | | | |
Collapse
|
32
|
Vaisitti T, Audrito V, Serra S, Bologna C, Brusa D, Malavasi F, Deaglio S. NAD+-metabolizing ecto-enzymes shape tumor-host interactions: the chronic lymphocytic leukemia model. FEBS Lett 2011; 585:1514-20. [PMID: 21514298 DOI: 10.1016/j.febslet.2011.04.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/13/2011] [Accepted: 04/14/2011] [Indexed: 11/28/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is an essential co-enzyme that can be released in the extracellular milieu. Here, it may elicit signals through binding purinergic receptors. Alternatively, NAD(+) may be dismantled to adenosine, up-taken by cells and transformed to reconstitute the intracellular nucleotide pool. An articulated ecto-enzyme network is responsible for the nucleotide-nucleoside conversion. CD38 is the main mammalian enzyme that hydrolyzes NAD(+), generating Ca(2+)-active metabolites. Evidence suggests that this extracellular network may be altered or used by tumor cells to (i) nestle in protected areas, and (ii) evade the immune response. We have exploited chronic lymphocytic leukemia as a model to test the role of the ecto-enzyme network, starting by analyzing the individual elements that make up the whole picture.
Collapse
Affiliation(s)
- Tiziana Vaisitti
- Department of Genetics, Biology and Biochemistry, University of Turin, Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
33
|
Balogh Z, Reiniger L, Rajnai H, Csomor J, Szepesi Á, Balogh A, Deák L, Gagyi É, Bödör C, Matolcsy A. High rate of neoplastic cells with genetic abnormalities in proliferation centers of chronic lymphocytic leukemia. Leuk Lymphoma 2011; 52:1080-4. [DOI: 10.3109/10428194.2011.555889] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood 2010; 117:563-74. [PMID: 20940416 DOI: 10.1182/blood-2010-05-284984] [Citation(s) in RCA: 685] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL), an incurable malignancy of mature B lymphocytes, involves blood, bone marrow, and secondary lymphoid organs such as the lymph nodes (LN). A role of the tissue microenvironment in the pathogenesis of CLL is hypothesized based on in vitro observations, but its contribution in vivo remains ill-defined. To elucidate the effects of tumor-host interactions in vivo, we purified tumor cells from 24 treatment-naive patients. Samples were obtained concurrently from blood, bone marrow, and/or LN and analyzed by gene expression profiling. We identified the LN as a key site in CLL pathogenesis. CLL cells in the LN showed up-regulation of gene signatures, indicating B-cell receptor (BCR) and nuclear factor-κB activation. Consistent with antigen-dependent BCR signaling and canonical nuclear factor-κB activation, we detected phosphorylation of SYK and IκBα, respectively. Expression of BCR target genes was stronger in clinically more aggressive CLL, indicating more effective BCR signaling in this subtype in vivo. Tumor proliferation, quantified by the expression of the E2F and c-MYC target genes and verified with Ki67 staining by flow cytometry, was highest in the LN and was correlated with clinical disease progression. These data identify the disruption of tumor microenvironment interactions and the inhibition of BCR signaling as promising therapeutic strategies in CLL. This study is registered at http://clinicaltrials.gov as NCT00019370.
Collapse
|
35
|
D'Arena G, Laurenti L, Minervini MM, Deaglio S, Bonello L, De Martino L, De Padua L, Savino L, Tarnani M, De Feo V, Cascavilla N. Regulatory T-cell number is increased in chronic lymphocytic leukemia patients and correlates with progressive disease. Leuk Res 2010; 35:363-8. [PMID: 20880586 DOI: 10.1016/j.leukres.2010.08.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 08/17/2010] [Accepted: 08/19/2010] [Indexed: 12/15/2022]
Abstract
Regulatory T-cells (Treg) actively maintain immunological self-tolerance and play a significant role in the progression of cancer. Treg cell numbers have been evaluated in 80 patients with previously untreated chronic lymphocytic leukemia (CLL) and in 40 normal healthy volunteers. Treg cells are higher in CLL patients than in controls and correlate with disease status (more advanced clinical stage, peripheral blood B-cell lymphocytosis, absolute CD38+ B-cell number, and more elevated LDH levels). No correlation was found with ZAP-70 expression, IgVH mutational status and cytogenetic abnormalities. This data shows that Treg cell number is abnormal in CLL patients.
Collapse
Affiliation(s)
- Giovanni D'Arena
- IRCCS Casa Sollievo della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Antiapoptotic effect of interleukin-2 (IL-2) in B-CLL cells with low and high affinity IL-2 receptors. Ann Hematol 2010; 89:1125-32. [PMID: 20544350 DOI: 10.1007/s00277-010-0994-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 05/11/2010] [Indexed: 10/19/2022]
Abstract
Although B chronic lymphocytic leukemia (B-CLL) cells express the alpha chain of the interleukin-2 (IL-2) receptor CD25, little is known about the effect of IL-2 on apoptosis in B-CLL cells. We have shown previously that stimulation of B-CLL cells with a CpG-oligonucleotide induces IL-2 high affinity receptors. In our current work, we analyzed the effect of IL-2 on apoptosis in resting B-CLL cells and in our model of activated B-CLL cells (CD25 high cells). IL-2 had modest antiapoptotic activity in resting B-CLL cells. In contrast, IL-2 was much more potent to prevent apoptosis in activated cells. Prevention of cell death was also associated with the maintenance of the mitochondrial membrane potential. While only limited regulation of apoptosis controlling proteins was observed in resting B-CLL cells, IL-2 had strong effects on MCL-1, Bcl-xl, and survivin expression and inhibited Bax cleavage in CD25 high cells. Interestingly, expression of Bcl-2 was reduced. Addition of IL-2 to activated B-CLL cells caused rapid phosphorylation of Akt, while IL-2 failed to significantly phosphorylate Akt in resting B-CLL cells. Pharmacological inhibition of Akt by LY294002 restored sensitivity of activated B-CLL cells to fludarabine. IL-2 might be an important survival factor in activated B-CLL cells and might contribute to disease progression by upregulation of several critical antiapoptotic proteins.
Collapse
|
37
|
|