1
|
Biersack B, Höpfner M. Emerging role of MYB transcription factors in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:15. [PMID: 38835346 PMCID: PMC11149108 DOI: 10.20517/cdr.2023.158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/19/2024] [Accepted: 04/04/2024] [Indexed: 06/06/2024]
Abstract
Decades ago, the viral myeloblastosis oncogene v-myb was identified as a gene responsible for the development of avian leukemia. However, the relevance of MYB proteins for human cancer diseases, in particular for solid tumors, remained basically unrecognized for a very long time. The human family of MYB transcription factors comprises MYB (c-MYB), MYBL2 (b-MYB), and MYBL1 (a-MYB), which are overexpressed in several cancers and are associated with cancer progression and resistance to anticancer drugs. In addition to overexpression, the presence of activated MYB-fusion proteins as tumor drivers was described in certain cancers. The identification of anticancer drug resistance mediated by MYB proteins and their underlying mechanisms are of great importance in understanding failures of current therapies and establishing new and more efficient therapy regimens. In addition, new drug candidates targeting MYB transcription factor activity and signaling have emerged as a promising class of potential anticancer therapeutics that could tackle MYB-dependent drug-resistant cancers in a more selective way. This review describes the correlation of MYB transcription factors with the formation and persistence of cancer resistance to various approved and investigational anticancer drugs.
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Bayreuth 95440, Germany
| | - Michael Höpfner
- Institute for Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin 10117, Germany
| |
Collapse
|
2
|
Tejera Nevado P, Tešan Tomić T, Atefyekta A, Fehr A, Stenman G, Andersson MK. Synthetic oleanane triterpenoids suppress MYB oncogene activity and sensitize T-cell acute lymphoblastic leukemia cells to chemotherapy. Front Oncol 2023; 13:1126354. [PMID: 37077825 PMCID: PMC10106619 DOI: 10.3389/fonc.2023.1126354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy with poor prognosis. The MYB oncogene encodes a master transcription factor that is activated in the majority of human T-ALLs. In the present study, we have performed a large-scale screening with small-molecule drugs to find clinically useful inhibitors of MYB gene expression in T-ALL. We identified several pharmacological agents that potentially could be used to treat MYB-driven malignancies. In particular, treatment with the synthetic oleanane triterpenoids (OTs) bardoxolone methyl and omaveloxolone decreased MYB gene activity and expression of MYB downstream target genes in T-ALL cells with constitutive MYB gene activation. Notably, treatment with bardoxolone methyl and omaveloxolone led to a dose-dependent reduction in cell viability and induction of apoptosis at low nanomolar concentrations. In contrast, normal bone marrow-derived cells were unaffected at these concentrations. Bardoxolone methyl and omaveloxolone treatment downregulated the expression of DNA repair genes and sensitized T-ALL cells to doxorubicin, a drug that is part of the standard therapy of T-ALL. OT treatment may thus potentiate DNA-damaging chemotherapy through attenuation of DNA repair. Taken together, our results indicate that synthetic OTs may be useful in the treatment of T-ALL and potentially also in other MYB-driven malignancies.
Collapse
|
3
|
Zhao A, Zhou H, Yang J, Li M, Niu T. Epigenetic regulation in hematopoiesis and its implications in the targeted therapy of hematologic malignancies. Signal Transduct Target Ther 2023; 8:71. [PMID: 36797244 PMCID: PMC9935927 DOI: 10.1038/s41392-023-01342-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/03/2023] [Accepted: 01/19/2023] [Indexed: 02/18/2023] Open
Abstract
Hematologic malignancies are one of the most common cancers, and the incidence has been rising in recent decades. The clinical and molecular features of hematologic malignancies are highly heterogenous, and some hematologic malignancies are incurable, challenging the treatment, and prognosis of the patients. However, hematopoiesis and oncogenesis of hematologic malignancies are profoundly affected by epigenetic regulation. Studies have found that methylation-related mutations, abnormal methylation profiles of DNA, and abnormal histone deacetylase expression are recurrent in leukemia and lymphoma. Furthermore, the hypomethylating agents and histone deacetylase inhibitors are effective to treat acute myeloid leukemia and T-cell lymphomas, indicating that epigenetic regulation is indispensable to hematologic oncogenesis. Epigenetic regulation mainly includes DNA modifications, histone modifications, and noncoding RNA-mediated targeting, and regulates various DNA-based processes. This review presents the role of writers, readers, and erasers of DNA methylation and histone methylation, and acetylation in hematologic malignancies. In addition, this review provides the influence of microRNAs and long noncoding RNAs on hematologic malignancies. Furthermore, the implication of epigenetic regulation in targeted treatment is discussed. This review comprehensively presents the change and function of each epigenetic regulator in normal and oncogenic hematopoiesis and provides innovative epigenetic-targeted treatment in clinical practice.
Collapse
Affiliation(s)
- Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Hui Zhou
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Jinrong Yang
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Meng Li
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Kumar A, Emdad L, Fisher PB, Das SK. Targeting epigenetic regulation for cancer therapy using small molecule inhibitors. Adv Cancer Res 2023; 158:73-161. [PMID: 36990539 DOI: 10.1016/bs.acr.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Cancer cells display pervasive changes in DNA methylation, disrupted patterns of histone posttranslational modification, chromatin composition or organization and regulatory element activities that alter normal programs of gene expression. It is becoming increasingly clear that disturbances in the epigenome are hallmarks of cancer, which are targetable and represent attractive starting points for drug creation. Remarkable progress has been made in the past decades in discovering and developing epigenetic-based small molecule inhibitors. Recently, epigenetic-targeted agents in hematologic malignancies and solid tumors have been identified and these agents are either in current clinical trials or approved for treatment. However, epigenetic drug applications face many challenges, including low selectivity, poor bioavailability, instability and acquired drug resistance. New multidisciplinary approaches are being designed to overcome these limitations, e.g., applications of machine learning, drug repurposing, high throughput virtual screening technologies, to identify selective compounds with improved stability and better bioavailability. We provide an overview of the key proteins that mediate epigenetic regulation that encompass histone and DNA modifications and discuss effector proteins that affect the organization of chromatin structure and function as well as presently available inhibitors as potential drugs. Current anticancer small-molecule inhibitors targeting epigenetic modified enzymes that have been approved by therapeutic regulatory authorities across the world are highlighted. Many of these are in different stages of clinical evaluation. We also assess emerging strategies for combinatorial approaches of epigenetic drugs with immunotherapy, standard chemotherapy or other classes of agents and advances in the design of novel epigenetic therapies.
Collapse
|
5
|
Bochicchio MT, Di Battista V, Poggio P, Carrà G, Morotti A, Brancaccio M, Lucchesi A. Understanding Aberrant Signaling to Elude Therapy Escape Mechanisms in Myeloproliferative Neoplasms. Cancers (Basel) 2022; 14:cancers14040972. [PMID: 35205715 PMCID: PMC8870427 DOI: 10.3390/cancers14040972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/01/2023] Open
Abstract
Aberrant signaling in myeloproliferative neoplasms may arise from alterations in genes coding for signal transduction proteins or epigenetic regulators. Both mutated and normal cells cooperate, altering fragile balances in bone marrow niches and fueling persistent inflammation through paracrine or systemic signals. Despite the hopes placed in targeted therapies, myeloid proliferative neoplasms remain incurable diseases in patients not eligible for stem cell transplantation. Due to the emergence of drug resistance, patient management is often very difficult in the long term. Unexpected connections among signal transduction pathways highlighted in neoplastic cells suggest new strategies to overcome neoplastic cell adaptation.
Collapse
Affiliation(s)
- Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Valeria Di Battista
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| |
Collapse
|
6
|
Bartalucci N, Galluzzi L. Philadelphia-negative myeloproliferative neoplasms: From origins to new perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 366:ix-xx. [PMID: 35153008 DOI: 10.1016/s1937-6448(22)00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; DENOThe Excellence Center, University of Florence, Florence, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| |
Collapse
|
7
|
The JAK2 inhibitor TG101209 exhibits anti-tumor and chemotherapeutic sensitizing effects on Burkitt lymphoma cells by inhibiting the JAK2/STAT3/c-MYB signaling axis. Cell Death Discov 2021; 7:268. [PMID: 34588425 PMCID: PMC8481535 DOI: 10.1038/s41420-021-00655-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/18/2021] [Accepted: 09/09/2021] [Indexed: 11/30/2022] Open
Abstract
Constitutive activation of JAK2/STAT3 is a major oncogenic signaling event involved in the development of Burkitt lymphoma (BL). In the present study, we investigated the antilymphoma activity of TG101209, a specific JAK2 inhibitor, on EBV-positive and EBV-negative Burkitt lymphoma cell lines and primary BL cells. The results showed that TG101209 had a significant antilymphoma effect by inhibiting BL cell growth and inducing apoptosis along with cell differentiation toward mature B cells in vitro. We also found that TG101209 displayed significant synergistic action and a sensitizing effect on the anti-Burkitt lymphoma activity of doxorubicin. In vivo experiments indicated that TG101209 could suppress tumor growth and prolong the overall survival of BL cell-bearing mice. The mechanistic study indicated that TG101209, by suppressing the JAK2/STAT3/c-MYB signaling axis and crosstalk between the downstream signaling pathways, plays an antilymphoma role. These data suggested that TG101209 may be a promising agent or alternative choice for the treatment of BL.
Collapse
|
8
|
Impact of NFE2 mutations on AML transformation and overall survival in patients with myeloproliferative neoplasms (MPN). Blood 2021; 138:2142-2148. [PMID: 33945619 DOI: 10.1182/blood.2020010402] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/23/2021] [Indexed: 11/20/2022] Open
|
9
|
Reeves BN, Beckman JD. Novel Pathophysiological Mechanisms of Thrombosis in Myeloproliferative Neoplasms. Curr Hematol Malig Rep 2021; 16:304-313. [PMID: 33876389 DOI: 10.1007/s11899-021-00630-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Thrombosis remains a leading cause of morbidity and mortality in BCR/ABL negative myeloproliferative neoplasms (MPN). Circulating blood cells are both increased in quantity and qualitatively abnormal in MPN, resulting in an increased thrombotic risk. Herein, we review recently elucidated mechanisms of MPN thrombosis and discuss implications of drugs currently under investigation for MPN. RECENT FINDINGS Recent studies highlight that in JAK2V617F granulocytes and platelets, thrombo-inflammatory genes are upregulated. Furthermore, in JAK2V617F granulocytes, protein expression of integrin CD11b, tissue factor, and leukocyte alkaline phosphatase are all increased. Overall, myeloid cells, namely neutrophils, may contribute in several ways, such as through increased adhesion via β1 integrin binding to VCAM1, increased infiltration, and enhanced inducibility to extrude neutrophil extracellular traps. Non-myeloid inflammatory cells may also contribute via secretion of cytokines. With regard to red blood cells, number, rigidity, adhesion, and generation of microvesicles may lead to increased vascular resistance as well as increased cell-cell interactions that promote rolling and adhesion. Platelets may also contribute in a similar fashion. Lastly, the vasculature is also increasingly appreciated, as several studies have demonstrated increased endothelial expression of pro-coagulant and pro-adhesive proteins, such as von Willebrand factor or P-selectin in JAK2V617F endothelial cells. With the advent of molecular diagnostics, MPN therapeutics are advancing beyond cytoreduction. Our increased understanding of pro-inflammatory and thrombotic pathophysiology in MPN provides a rational basis for evaluation of in-development MPN therapeutics to reduce thrombosis.
Collapse
Affiliation(s)
- Brandi N Reeves
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
- Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan D Beckman
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, 420 Delaware St. SE, MMC 480, Minneapolis, MN, 55455, USA.
| |
Collapse
|
10
|
Rambaldi A, Iurlo A, Vannucchi AM, Martino B, Guarini A, Ruggeri M, von Bubnoff N, De Muro M, McMullin MF, Luciani S, Martinelli V, Nogai A, Rosti V, Ricco A, Bettica P, Manzoni S, Di Tollo S. Long-term safety and efficacy of givinostat in polycythemia vera: 4-year mean follow up of three phase 1/2 studies and a compassionate use program. Blood Cancer J 2021; 11:53. [PMID: 33677466 PMCID: PMC7936975 DOI: 10.1038/s41408-021-00445-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
Polycythemia vera (PV) is a BCR-ABL1-negative myeloproliferative neoplasm (MPN) characterized by excessive proliferation of erythroid, myeloid, and megakaryocytic components in the bone marrow, mainly due to a Janus kinase 2 gene mutation (JAK2V617F). Givinostat, a histone-deacetylase inhibitor that selectively targets JAK2V617F cell growth, has demonstrated good efficacy and safety in three phase 1/2 studies in patients with PV. This manuscript focuses on the 4-year mean (2.8 year median) follow-up of an open-label, long-term study that enrolled 51 patients with PV (out of a total of 54 with MPN) who received clinical benefit from givinostat in these previous studies or on compassionate use, and who continued to receive givinostat at the last effective and tolerated dose. The primary objectives are to determine givinostat's long-term safety and tolerability, and efficacy evaluated by the investigators according to internationally recognized response criteria. During follow-up, only 10% of PV patients reported Grade 3 treatment-related adverse events (AEs), while none had Grade 4 or 5 treatment-related AEs. The overall response rate for the duration of follow-up was always greater than 80% in patients with PV. In conclusion, givinostat demonstrated a good safety and efficacy profile in patients with PV, data supporting long-term use in this population.
Collapse
Affiliation(s)
- Alessandro Rambaldi
- Department of Oncology and Hematology University of Milan, and Azienda SocioSanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.
| | - Alessandra Iurlo
- Hematology Division, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandro M Vannucchi
- Center Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, University of Florence, Florence, Italy
| | - Bruno Martino
- Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Haematology Unit, Reggio Calabria, Italy
| | - Attilio Guarini
- Hematology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Marco Ruggeri
- U.O. Haematology, San Bortolo Hospital, Vicenza, Italy
| | - Nikolas von Bubnoff
- Department of Haematology, Oncology and Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Haematology and Oncology, Medical Center, University of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Marianna De Muro
- Hematology and Stem Cells Transplantation Unit, Campus Bio-Medico, University Hospital, Rome, Italy
| | | | - Stefania Luciani
- Dipartimento Oncologia-Ematologia, U.O. Complessa Ematologia Clinica, Presidio Ospedaliero "Spirito Santo"- A.S.L. Azienda Sanitaria Locale, Pescara, Italy
| | - Vincenzo Martinelli
- Dipartimento di Medicina Clinica e Chirurgia, Ematologia, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Axel Nogai
- Division of Hematology and Oncology at Campus Benjamin Franklin (CBF), Charité, Berlin, Germany
| | - Vittorio Rosti
- Fondazione I.R.C.C.S. Policlinico San Matteo di Pavia, Centro per lo Studio e la Cura della Mielofibrosi, Laboratorio Biochimica, Biotecnologie e Diagnostica Avanzata, Pavia, Italy
| | - Alessandra Ricco
- Azienda Ospedaliero-Universitaria Policlinico Consorziale di Bari, U. O. Ematologia con Trapianto - Ambulatorio, Bari, Italy
| | - Paolo Bettica
- Clinical R&D Department, Italfarmaco S.p.A, Cinisello Balsamo, Italy
| | - Sara Manzoni
- Clinical R&D Department, Italfarmaco S.p.A, Cinisello Balsamo, Italy
| | - Silvia Di Tollo
- Clinical R&D Department, Italfarmaco S.p.A, Cinisello Balsamo, Italy
| |
Collapse
|
11
|
Yung Y, Lee E, Chu HT, Yip PK, Gill H. Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22020659. [PMID: 33440869 PMCID: PMC7827471 DOI: 10.3390/ijms22020659] [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: 12/09/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 02/02/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in haematopoiesis. They are characterized by stem cell-derived clonal myeloproliferation. The key MPNs comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). CML is defined by the presence of the Philadelphia (Ph) chromosome and BCR-ABL1 fusion gene. Despite effective cytoreductive agents and targeted therapy, complete CML/MPN stem cell eradication is rarely achieved. In this review article, we discuss the novel agents and combination therapy that can potentially abnormal hematopoietic stem cells in CML and MPNs and the CML/MPN stem cell-sustaining bone marrow microenvironment.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Autophagy
- Biomarkers, Tumor
- Cell Survival/drug effects
- Cell Transformation, Neoplastic/genetics
- Combined Modality Therapy
- Disease Susceptibility
- Genetic Predisposition to Disease
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Molecular Targeted Therapy
- Myeloproliferative Disorders/etiology
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/therapy
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Philadelphia Chromosome
- Signal Transduction/drug effects
- Stem Cell Niche
- Tumor Microenvironment
Collapse
Affiliation(s)
| | | | | | | | - Harinder Gill
- Correspondence: ; Tel.: +852-2255-4542; Fax: +852-2816-2863
| |
Collapse
|
12
|
Smalley JP, Cowley SM, Hodgkinson JT. Bifunctional HDAC Therapeutics: One Drug to Rule Them All? Molecules 2020; 25:E4394. [PMID: 32987782 PMCID: PMC7583022 DOI: 10.3390/molecules25194394] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Histone deacetylase (HDAC) enzymes play crucial roles in epigenetic gene expression and are an attractive therapeutic target. Five HDAC inhibitors have been approved for cancer treatment to date, however, clinical applications have been limited due to poor single-agent drug efficacy and side effects associated with a lack of HDAC isoform or complex selectivity. An emerging strategy aiming to address these limitations is the development of bifunctional HDAC therapeutics-single molecules comprising a HDAC inhibitor conjugated to another specificity targeting moiety. This review summarises the recent advancements in novel types of dual-targeting HDAC modulators, including proteolysis-targeting chimeras (PROTACs), with a focus on HDAC isoform and complex selectivity, and the future potential of such bifunctional molecules in achieving enhanced drug efficacy and therapeutic benefits in treating disease.
Collapse
Affiliation(s)
- Joshua P. Smalley
- Leicester Institute of Structural and Chemical Biology, School of Chemistry, University of Leicester, George Porter Building, University Road, Leicester LE1 7RH, UK;
| | - Shaun M. Cowley
- Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK;
| | - James T. Hodgkinson
- Leicester Institute of Structural and Chemical Biology, School of Chemistry, University of Leicester, George Porter Building, University Road, Leicester LE1 7RH, UK;
| |
Collapse
|
13
|
Bartalucci N, Guglielmelli P, Vannucchi AM. Polycythemia vera: the current status of preclinical models and therapeutic targets. Expert Opin Ther Targets 2020; 24:615-628. [PMID: 32366208 DOI: 10.1080/14728222.2020.1762176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Polycythemia vera (PV) is the most common myeloproliferative neoplasm (MPN). PV is characterized by erythrocytosis, leukocytosis, thrombocytosis, increased hematocrit, and hemoglobin in the peripheral blood. Splenomegaly and myelofibrosis often occur in PV patients. Almost all PV patients harbor a mutation in the JAK2 gene, mainly represented by the JAK2V617F point mutation. AREAS COVERED This article examines the recent in vitro and in vivo available models of PV and moreover, it offers insights on emerging biomarkers and therapeutic targets. The evidence from mouse models, resembling a PV-like phenotype generated by different technical approaches, is discussed. The authors searched PubMed, books, and clinicaltrials.gov for original and review articles and drugs development status including the terms Myeloproliferative Neoplasms, Polycythemia Vera, erythrocytosis, hematocrit, splenomegaly, bone marrow fibrosis, JAK2V617F, Hematopoietic Stem Cells, MPN cytoreductive therapy, JAK2 inhibitor, histone deacetylase inhibitor, PV-like phenotype, JAK2V617F BMT, transgenic JAK2V617F mouse, JAK2 physiologic promoter. EXPERT OPINION Preclinical models of PV are valuable tools for enabling an understanding of the pathophysiology and the molecular mechanisms of the disease. These models provide new biological insights on the contribution of concomitant mutations and the efficacy of novel drugs in a 'more faithful' setting. This may facilitate an enhanced understanding of pathogenetic mechanisms and targeted therapy.
Collapse
Affiliation(s)
- Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, Center Research and Innovation of Myeloproliferative Neoplasms - CRIMM, Azienda Ospedaliera Universitaria Careggi, University of Florence , Florence, Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, Center Research and Innovation of Myeloproliferative Neoplasms - CRIMM, Azienda Ospedaliera Universitaria Careggi, University of Florence , Florence, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, Center Research and Innovation of Myeloproliferative Neoplasms - CRIMM, Azienda Ospedaliera Universitaria Careggi, University of Florence , Florence, Italy
| |
Collapse
|
14
|
Chifotides HT, Bose P, Verstovsek S. Givinostat: an emerging treatment for polycythemia vera. Expert Opin Investig Drugs 2020; 29:525-536. [PMID: 32693648 PMCID: PMC7534842 DOI: 10.1080/13543784.2020.1761323] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Polycythemia vera (PV), a Philadelphia chromosome-negative myeloproliferative neoplasm, is characterized by panmyelosis, pancytosis, and a JAK2 mutation. Patients are at increased risk of thrombohemorrhagic events, and progression to myelofibrosis or acute leukemia. Current treatments include aspirin, phlebotomy, and cytoreductive drugs (most commonly hydroxyurea). Givinostat is a potent, class I/II histone deacetylase (HDAC) inhibitor that is in phase I/II clinical trials in PV. Givinostat was well tolerated and yielded promising clinico-hematological responses. A phase III study of givinostat versus hydroxyurea in high-risk PV patients is planned. AREAS COVERED We present an overview of PV, current treatment guidelines, and the putative mechanism(s) of action of givinostat. We discuss the preclinical and clinical studies of givinostat in PV and briefly review approved and investigational competitor compounds. EXPERT OPINION HDAC inhibitors have long been known to be active in PV, but chronic toxicities can be challenging. Givinostat, however, is active and well tolerated, and is entering a pivotal Phase III randomized trial. Givinostat offers the possibility of replacing hydroxyurea as the standard first-line cytoreductive choice for PV patients. This would completely change the current therapeutic paradigm and guidelines for PV management. Although surrogate clinical study endpoints may suffice for regulatory purposes, thrombosis reduction and prevention of disease progression remain most important to patients and clinicians.
Collapse
Affiliation(s)
- Helen T. Chifotides
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
15
|
Shariati L, Rohani F, Heidari Hafshejani N, Kouhpayeh S, Boshtam M, Mirian M, Rahimmanesh I, Hejazi Z, Modarres M, Pieper IL, Khanahmad H. Disruption of
SOX6
gene using CRISPR/Cas9 technology for gamma‐globin reactivation: An approach towards gene therapy of β‐thalassemia. J Cell Biochem 2018; 119:9357-9363. [DOI: 10.1002/jcb.27253] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/20/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Laleh Shariati
- Applied Physiology Research Center Cardiovascular Research Institute, Isfahan University of Medical Sciences Isfahan Iran
- Isfahan Cardiovascular Research Center Cardiovascular Research Institute, Isfahan University of Medical Sciences Isfahan Iran
| | - Fattah Rohani
- Department of Clinical Sciences Faculty of Veterinary Medicine, University of Shahrekord Shahrekord Iran
| | - Nahid Heidari Hafshejani
- Department of Genetics and Molecular Biology School of Medicine, Isfahan University of Medical Sciences Isfahan Iran
| | - Shirin Kouhpayeh
- Isfahan Neurosciences Research Center, Alzahra Research Institute Isfahan University of Medical Sciences Isfahan Iran
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center Cardiovascular Research Institute, Isfahan University of Medical Sciences Isfahan Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Science Research Center, School of Pharmacy and Pharmaceutical Science Isfahan University of Medical Sciences Isfahan Iran
| | - Ilnaz Rahimmanesh
- Department of Genetics and Molecular Biology School of Medicine, Isfahan University of Medical Sciences Isfahan Iran
| | - Zahra Hejazi
- Department of Genetics and Molecular Biology School of Medicine, Isfahan University of Medical Sciences Isfahan Iran
| | - Mehran Modarres
- Department of Genetics and Molecular Biology School of Medicine, Isfahan University of Medical Sciences Isfahan Iran
| | - Ina Laura Pieper
- Institute of Life Science, College of Medicine Swansea University Medical School Swansea United Kingdom
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology School of Medicine, Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
16
|
Savino AM, Sarno J, Trentin L, Vieri M, Fazio G, Bardini M, Bugarin C, Fossati G, Davis KL, Gaipa G, Izraeli S, Meyer LH, Nolan GP, Biondi A, Te Kronnie G, Palmi C, Cazzaniga G. The histone deacetylase inhibitor givinostat (ITF2357) exhibits potent anti-tumor activity against CRLF2-rearranged BCP-ALL. Leukemia 2017; 31:2365-2375. [PMID: 28331226 DOI: 10.1038/leu.2017.93] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 01/05/2017] [Accepted: 02/06/2017] [Indexed: 12/18/2022]
Abstract
Leukemias bearing CRLF2 and JAK2 gene alterations are characterized by aberrant JAK/STAT signaling and poor prognosis. The HDAC inhibitor givinostat/ITF2357 has been shown to exert anti-neoplastic activity against both systemic juvenile idiopathic arthritis and myeloproliferative neoplasms through inhibition of the JAK/STAT pathway. These findings led us to hypothesize that givinostat might also act against CRLF2-rearranged BCP-ALL, which lack effective therapies. Here, we found that givinostat inhibited proliferation and induced apoptosis of BCP-ALL CRLF2-rearranged cell lines, positive for exon 16 JAK2 mutations. Likewise, givinostat killed primary cells, but not their normal hematopoietic counterparts, from patients carrying CRLF2 rearrangements. At low doses, givinostat downregulated the expression of genes belonging to the JAK/STAT pathway and inhibited STAT5 phosphorylation. In vivo, givinostat significantly reduced engraftment of human blasts in patient-derived xenograft models of CRLF2-positive BCP-ALL. Importantly, givinostat killed ruxolitinib-resistant cells and potentiated the effect of current chemotherapy. Thus, givinostat in combination with conventional chemotherapy may represent an effective therapeutic option for these difficult-to-treat subsets of ALL. Lastly, the selective killing of cancer cells by givinostat may allow the design of reduced intensity regimens in CRLF2-rearranged Down syndrome-associated BCP-ALL patients with an overall benefit in terms of both toxicity and related complications.
Collapse
Affiliation(s)
- A M Savino
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy.,Department of Pediatric Hematology and Oncology, Leukemia Research Section, Edmond and Lily Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J Sarno
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - L Trentin
- Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - M Vieri
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - G Fazio
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - M Bardini
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - C Bugarin
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - G Fossati
- Preclinical R&D Department, Italfarmaco S.p.A., Cinisello Balsamo, Milan, Italy
| | - K L Davis
- Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.,Hematology and Oncology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - G Gaipa
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - S Izraeli
- Department of Pediatric Hematology and Oncology, Leukemia Research Section, Edmond and Lily Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - L H Meyer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - G P Nolan
- Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.,Hematology and Oncology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - A Biondi
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - G Te Kronnie
- Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - C Palmi
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - G Cazzaniga
- Tettamanti Research Center, Department of Pediatrics, University of Milano Bicocca, Fondazione MBBM, Monza, Italy
| |
Collapse
|
17
|
Bose P, Verstovsek S. Investigational histone deacetylase inhibitors (HDACi) in myeloproliferative neoplasms. Expert Opin Investig Drugs 2016; 25:1393-1403. [PMID: 27756180 DOI: 10.1080/13543784.2016.1250882] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The Philadelphia chromosome negative myeloproliferative neoplasms (MPN) mainly comprise polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis (MF, primary or post-PV/ET). Therapy in PV and ET focuses on minimizing thrombosis and bleeding risk, while in MF, prolongation of survival is an important goal. Different cytoreductive agents are employed in high risk PV and ET, while the JAK inhibtior ruxolitinib is the cornerstone of therapy in MF. Histone deacetylase inhibitors (HDACi) are pleiotropic agents with diverse epigenetic and non-epigenetic actions, selectively in transformed cells. A number of HDACi have been or are being investigated in MPN. Areas covered: The mechanisms of action of HDACI in neoplastic cells are summarized, and the preclinical rationale and data supporting their development in MPN specifically examined, particularly their synergism with JAK inhibitors. Major findings of clinical trials of HDACi, both alone and in combination with ruxolitinib, in MPN are then discussed, with particular attention to their toxicities and disease-modifying effects. Expert opinion: HDACi are clearly active in MPN, and there is good preclinical rationale for this. Their combination with ruxolitinib in MF is promising, but the long-term tolerability of these agents is an important concern. Further development in PV or ET appears unlikely.
Collapse
Affiliation(s)
- Prithviraj Bose
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Srdan Verstovsek
- a Department of Leukemia , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
18
|
Shariati L, Khanahmad H, Salehi M, Hejazi Z, Rahimmanesh I, Tabatabaiefar MA, Modarressi MH. Genetic disruption of the KLF1 gene to overexpress the γ-globin gene using the CRISPR/Cas9 system. J Gene Med 2016; 18:294-301. [PMID: 27668420 DOI: 10.1002/jgm.2928] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/03/2016] [Accepted: 09/22/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND β-thalassemia comprises a major group of human genetic disorders involving a decrease in or an end to the normal synthesis of the β-globin chains of hemoglobin. KLF1 is a key regulatory molecule involved in the γ- to β-globin gene switching process directly inducing the expression of the β-globin gene and indirectly repressing γ-globin. The present study aimed to investigate the ability of an engineered CRISPR/Cas9 system with respect to disrupting the KLF1 gene to inhibit the γ- to β-hemoglobin switching process in K562 cells. METHODS We targeted three sites on the KLF1 gene, two of which are upstream of codon 288 in exon 2 and the other site being in exon 3. RESULTS The average indel percentage in the cells transfected with CRISPR a, b and c was approximately 24%. Relative quantification was performed for the assessment of γ-globin expression. The levels of γ-globin mRNA on day 5 of differentiation were 8.1-, 7.7- and 1.8-fold in the cells treated with CRISPR/Cas9 a, b and c, respectively,compared to untreated cells. The measurement of HbF expression levels confirmed the same results. CONCLUSIONS The findings obtained in the present study support the induction of an indel mutation in the KLF1 gene leading to a null allele. As a result, the effect of KLF1 on the expression of BCL11A is decreased and its inhibitory effect on γ-globin gene expression is removed. Application of CRISPR technology to induce an indel in the KLF1 gene in adult erythroid progenitors may provide a method for activating fetal hemoglobin expression in individuals with β-thalassemia or sickle cell disease.
Collapse
Affiliation(s)
- Laleh Shariati
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansoor Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Hejazi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
19
|
Yang EG, Mustafa N, Tan EC, Poulsen A, Ramanujulu PM, Chng WJ, Yen JJY, Dymock BW. Design and Synthesis of Janus Kinase 2 (JAK2) and Histone Deacetlyase (HDAC) Bispecific Inhibitors Based on Pacritinib and Evidence of Dual Pathway Inhibition in Hematological Cell Lines. J Med Chem 2016; 59:8233-62. [DOI: 10.1021/acs.jmedchem.6b00157] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Eugene Guorong Yang
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | - Nurulhuda Mustafa
- Department
of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228
| | - Eng Chong Tan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Anders Poulsen
- Experimental Therapeutics Centre, 31 Biopolis Way, 03-01 Nanos, Singapore 138669
- Department
of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543
| | - Pondy Murugappan Ramanujulu
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
- Life Sciences
Institute, National University of Singapore, Centre for Life Sciences, Level
5, 28 Medical Drive, Singapore 117456
| | - Wee Joo Chng
- Department
of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228
- Cancer
Science Institute, Singapore, National University of Singapore, Singapore 117599
- National
University Cancer Institute of Singapore, National University Health SystemSingapore 119074
| | - Jeffrey J. Y. Yen
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Brian W. Dymock
- Department
of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543
| |
Collapse
|
20
|
Pierini T, Di Giacomo D, Pierini V, Gorello P, Barba G, Lema Fernandez AG, Pellanera F, Iannotti T, Falzetti F, La Starza R, Mecucci C. MYB deregulation from a EWSR1-MYB fusion at leukemic evolution of a JAK2 (V617F) positive primary myelofibrosis. Mol Cytogenet 2016; 9:68. [PMID: 27594918 PMCID: PMC5009546 DOI: 10.1186/s13039-016-0277-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/22/2016] [Indexed: 02/08/2023] Open
Abstract
Background Although Philadelphia-negative myeloproliferative neoplasms (Ph-MPN) are usually not aggressive, the type and the number of molecular lesions impact greatly on leukemic transformation. Indeed, the molecular background underlying progression is still largely unexplored even though ASXL1, IDH1/2, SRSF2, and TP53 mutations, together with adverse karyotypic changes, place the patient at high risk of leukemic transformation. Case presentation Our patient, a 64-year old man with a diagnosis of JAK2V617F primary myelofibrosis (PMF) had an unusually rapid leukemic transformation. Genomic profiling showed that TET2 and SRSF2 mutations were also present. At leukemic transformation, the patient developed a complex chromosome rearrangement producing a EWSR1-MYB fusion. Remarkably, the expression of MYB and of its target BCL2 was, respectively, ≥4.7 and ≥2.8 fold higher at leukemic transformation than after chemotherapy, when the patient obtained the hematological remission. At this time point, the EWSR1-MYB fusion disappeared while JAK2V617F, TET2, and SRSF2 mutations, as well as PMF morphological features persisted. Conclusions Rapid leukemic transformation of JAK2V617F PMF was closely linked to a previously undescribed putative EWSR1-MYB transcription factor which was detected only at disease evolution. We hypothesize that the EWSR1-MYB contributed to leukemia transformation through at least two mechanisms: 1) it sustained MYB expression, and consequently deregulated its target BCL2, a putative onco-suppressor gene; and 2) ectopic EWSR1-MYB expression probably fulfilled its own oncogenic potential as demonstrated for other MYB-fusions. As our study confirmed that MYB is recurrently involved in chronic as well as leukemic transformation of PMF, it appears to be a valid molecular marker for tailored treatments. Electronic supplementary material The online version of this article (doi:10.1186/s13039-016-0277-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tiziana Pierini
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Danika Di Giacomo
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Valentina Pierini
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Paolo Gorello
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Gianluca Barba
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Anair Graciela Lema Fernandez
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Fabrizia Pellanera
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Tamara Iannotti
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Franca Falzetti
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Roberta La Starza
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Cristina Mecucci
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| |
Collapse
|
21
|
Whetton AD, Azmi NC, Pearson S, Jaworska E, Zhang L, Blance R, Kendall AC, Nicolaou A, Taylor S, Williamson AJ, Pierce A. MPL W515L expression induces TGFβ secretion and leads to an increase in chemokinesis via phosphorylation of THOC5. Oncotarget 2016; 7:10739-55. [PMID: 26919114 PMCID: PMC4905435 DOI: 10.18632/oncotarget.7639] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/15/2016] [Indexed: 02/03/2023] Open
Abstract
The thrombopoietin receptor (MPL) has been shown to be mutated (MPL W515L) in myelofibrosis and thrombocytosis yet new approaches to treat this disorder are still required. We have previously shown that transcriptome and proteomic effects do not correlate well in oncogene-mediated leukemogenesis. We therefore investigated the effects of MPL W515L using proteomics. The consequences of MPL W515L expression on over 3300 nuclear and 3500 cytoplasmic proteins were assessed using relative quantification mass spectrometry. We demonstrate that MPL W515L expression markedly modulates the CXCL12/CXCR4/CD45 pathway associated with stem and progenitor cell chemotactic movement. We also demonstrated that MPL W515L expressing cells displayed increased chemokinesis which required the MPL W515L-mediated dysregulation of MYC expression via phosphorylation of the RNA transport protein THOC5 on tyrosine 225. In addition MPL W515L expression induced TGFβ secretion which is linked to sphingosine 1-phosphate production and the increased chemokinesis. These studies identify several pathways which offer potential targets for therapeutic intervention in the treatment of MPL W515L-driven malignancy. We validate our approach by showing that CD34+ cells from MPL W515L positive patients display increased chemokinesis and that treatment with a combination of MYC and sphingosine kinase inhibitors leads to the preferential killing of MPL W515L expressing cells.
Collapse
Affiliation(s)
- Anthony D. Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Norhaida Che Azmi
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Stella Pearson
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Ewa Jaworska
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Liqun Zhang
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Rognvald Blance
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Alexandra C. Kendall
- Manchester Pharmacy School, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Anna Nicolaou
- Manchester Pharmacy School, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Samuel Taylor
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Andrew J.K. Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| | - Andrew Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, The University of Manchester, Manchester, UK
| |
Collapse
|
22
|
Abstract
Cystic fibrosis (CF) lung disease is characterized by persistent and unresolved inflammation, with elevated proinflammatory and decreased anti-inflammatory cytokines, and greater numbers of immune cells. Hyperinflammation is recognized as a leading cause of lung tissue destruction in CF. Hyper-inflammation is not solely observed in the lungs of CF patients, since it may contribute to destruction of exocrine pancreas and, likely, to defects in gastrointestinal tract tissue integrity. Paradoxically, despite the robust inflammatory response, and elevated number of immune cells (such as neutrophils and macrophages), CF lungs fail to clear bacteria and are more susceptible to infections. Here, we have summarized the current understanding of immune dysregulation in CF, which may drive hyperinflammation and impaired host defense.
Collapse
Affiliation(s)
- Emanuela M Bruscia
- Section of Respiratory Medicine, Department of Pediatrics, Yale University School of Medicine, 330 Cedar Street, FMP, Room#524, New Haven, CT 06520, USA.
| | - Tracey L Bonfield
- Division of Pulmonology, Allergy and Immunology, Department of Pediatrics, Case Western Reserve University School of Medicine, 0900 Euclid Avenue, Cleveland, OH 44106-4948, USA.
| |
Collapse
|
23
|
Tedjaseputra A, Galli S, Ibrahim M, Harrison CN, McLornan DP. Histone deacetylase inhibitors in myeloproliferative neoplasms: current roles and future prospects. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1149467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
24
|
Kaplan JB, Stein BL, McMahon B, Giles FJ, Platanias LC. Evolving Therapeutic Strategies for the Classic Philadelphia-Negative Myeloproliferative Neoplasms. EBioMedicine 2016; 3:17-25. [PMID: 26870834 PMCID: PMC4739416 DOI: 10.1016/j.ebiom.2016.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/03/2016] [Accepted: 01/11/2016] [Indexed: 12/15/2022] Open
Abstract
Despite the emergence of JAK inhibitors, there is a need for disease-modifying treatments for Philadelphia-negative myeloproliferative neoplasms (MPNs). JAK inhibitors ameliorate symptoms and address splenomegaly, but because of the heterogeneous contributors to the disease process, JAK inhibitor monotherapy incompletely addresses the burden of disease. The ever-growing understanding of MPN pathogenesis has provided the rationale for testing novel and targeted therapeutic agents, as monotherapies or in combination, in preclinical and clinical settings. A number of intriguing options have emerged, and it is hoped that further progress will lead to significant changes in the natural history of MPNs.
Collapse
Affiliation(s)
- Jason B Kaplan
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Northwestern University, Chicago, IL, United States; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Northwestern University, Chicago, IL, United States
| | - Brandon McMahon
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Francis J Giles
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Northwestern University, Chicago, IL, United States
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Northwestern University, Chicago, IL, United States; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| |
Collapse
|
25
|
Gasiorek JJ, Blank V. Regulation and function of the NFE2 transcription factor in hematopoietic and non-hematopoietic cells. Cell Mol Life Sci 2015; 72:2323-35. [PMID: 25721735 PMCID: PMC11114048 DOI: 10.1007/s00018-015-1866-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/27/2015] [Accepted: 02/16/2015] [Indexed: 01/01/2023]
Abstract
The NFE2 transcription factor was identified over 25 years ago. The NFE2 protein forms heterodimers with small MAF proteins, and the resulting complex binds to regulatory elements in a large number of target genes. In contrast to other CNC transcription family members including NFE2L1 (NRF1), NFE2L2 (NRF2) and NFE2L3 (NRF3), which are widely expressed, earlier studies had suggested that the major sites of NFE2 expression are hematopoietic cells. Based on cell culture studies it was proposed that this protein acts as a critical regulator of globin gene expression. However, the knockout mouse model displayed only mild erythroid abnormalities, while the major phenotype was a defect in megakaryocyte biogenesis. Indeed, absence of NFE2 led to severely impaired platelet production. A series of recent data, also summarized here, shed new light on the various functional roles of NFE2 and the regulation of its activity. NFE2 is part of a complex regulatory network, including transcription factors such as GATA1 and RUNX1, controlling megakaryocytic and/or erythroid cell function. Surprisingly, it was recently found that NFE2 also has a role in non-hematopoietic tissues, such as the trophoblast, in which it is also expressed, as well as the bone, opening the door to new research areas for this transcription factor. Additional data showed that NFE2 function is controlled by a series of posttranslational modifications. Important strides have been made with respect to the clinical significance of NFE2, linking this transcription factor to hematological disorders such as polycythemias.
Collapse
Affiliation(s)
- Jadwiga J. Gasiorek
- Lady Davis Institute for Medical Research, McGill University, 3755 Chemin de la Côte Sainte-Catherine, Montreal, QC H3T 1E2 Canada
- Department of Medicine, McGill University, Montreal, QC Canada
| | - Volker Blank
- Lady Davis Institute for Medical Research, McGill University, 3755 Chemin de la Côte Sainte-Catherine, Montreal, QC H3T 1E2 Canada
- Department of Medicine, McGill University, Montreal, QC Canada
- Department of Physiology, McGill University, Montreal, QC Canada
| |
Collapse
|
26
|
Mets E, Van der Meulen J, Van Peer G, Boice M, Mestdagh P, Van de Walle I, Lammens T, Goossens S, De Moerloose B, Benoit Y, Van Roy N, Clappier E, Poppe B, Vandesompele J, Wendel HG, Taghon T, Rondou P, Soulier J, Van Vlierberghe P, Speleman F. MicroRNA-193b-3p acts as a tumor suppressor by targeting the MYB oncogene in T-cell acute lymphoblastic leukemia. Leukemia 2015; 29:798-806. [PMID: 25231743 PMCID: PMC4890642 DOI: 10.1038/leu.2014.276] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/07/2014] [Accepted: 08/15/2014] [Indexed: 12/12/2022]
Abstract
The MYB oncogene is a leucine zipper transcription factor essential for normal and malignant hematopoiesis. In T-cell acute lymphoblastic leukemia (T-ALL), elevated MYB levels can arise directly through T-cell receptor-mediated MYB translocations, genomic MYB duplications or enhanced TAL1 complex binding at the MYB locus or indirectly through the TAL1/miR-223/FBXW7 regulatory axis. In this study, we used an unbiased MYB 3'untranslated region-microRNA (miRNA) library screen and identified 33 putative MYB-targeting miRNAs. Subsequently, transcriptome data from two independent T-ALL cohorts and different subsets of normal T-cells were used to select miRNAs with relevance in the context of normal and malignant T-cell transformation. Hereby, miR-193b-3p was identified as a novel bona fide tumor-suppressor miRNA that targets MYB during malignant T-cell transformation thereby offering an entry point for efficient MYB targeting-oriented therapies for human T-ALL.
Collapse
Affiliation(s)
- E Mets
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | | | - G Van Peer
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - M Boice
- Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - P Mestdagh
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - I Van de Walle
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent, Belgium
| | - T Lammens
- Department of Pediatric Hematology—Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - S Goossens
- VIB/Department for Molecular Biomedical Research, Ghent University, Ghent, Belgium
| | - B De Moerloose
- Department of Pediatric Hematology—Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Y Benoit
- Department of Pediatric Hematology—Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - N Van Roy
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - E Clappier
- Genome Rearrangements and Cancer Laboratory, U462 INSERM, Laboratoire Central d'Hématologie and Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - B Poppe
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - J Vandesompele
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - H-G Wendel
- Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - T Taghon
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent, Belgium
| | - P Rondou
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - J Soulier
- Genome Rearrangements and Cancer Laboratory, U462 INSERM, Laboratoire Central d'Hématologie and Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | | | - F Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| |
Collapse
|
27
|
Abstract
Myeloproliferative neoplasms, including polycythemia vera (PV), essential thrombocythemia, and myelofibrosis (MF) (both primary and secondary), are recognized for their burdensome symptom profiles, life-threatening complications, and risk of progression to acute leukemia. Recent advancements in our ability to diagnose and prognosticate these clonal malignancies have paralleled the development of MPN-targeted therapies that have had a significant impact on disease burden and quality of life. Ruxolitinib has shown success in alleviating the symptomatic burden, reducing splenomegaly and improving quality of life in patients with MF. The role and clinical expectations of JAK2 inhibition continues to expand to a variety of investigational arenas. Clinical trials for patients with MF focus on new JAK inhibitors with potentially less myelosuppression( pacritinib) or even activity for anemia (momelotinib). Further efforts focus on combination trials (including a JAK inhibitor base) or targeting new pathways (ie, telomerase). Similarly, therapy for PV continues to evolve with phase 3 trials investigating optimal frontline therapy (hydroxyurea or IFN) and second-line therapy for hydroxyurea-refractory or intolerant PV with JAK inhibitors. In this chapter, we review the evolving data and role of JAK inhibition (alone or in combination) in the management of patients with MPNs.
Collapse
|
28
|
Geyer HL, Mesa RA. Therapy for myeloproliferative neoplasms: when, which agent, and how? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:277-286. [PMID: 25696867 DOI: 10.1182/asheducation-2014.1.277] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Myeloproliferative neoplasms, including polycythemia vera (PV), essential thrombocythemia, and myelofibrosis (MF) (both primary and secondary), are recognized for their burdensome symptom profiles, life-threatening complications, and risk of progression to acute leukemia. Recent advancements in our ability to diagnose and prognosticate these clonal malignancies have paralleled the development of MPN-targeted therapies that have had a significant impact on disease burden and quality of life. Ruxolitinib has shown success in alleviating the symptomatic burden, reducing splenomegaly and improving quality of life in patients with MF. The role and clinical expectations of JAK2 inhibition continues to expand to a variety of investigational arenas. Clinical trials for patients with MF focus on new JAK inhibitors with potentially less myelosuppression (pacritinib) or even activity for anemia (momelotinib). Further efforts focus on combination trials (including a JAK inhibitor base) or targeting new pathways (ie, telomerase). Similarly, therapy for PV continues to evolve with phase 3 trials investigating optimal frontline therapy (hydroxyurea or IFN) and second-line therapy for hydroxyurea-refractory or intolerant PV with JAK inhibitors. In this chapter, we review the evolving data and role of JAK inhibition (alone or in combination) in the management of patients with MPNs.
Collapse
Affiliation(s)
| | - Ruben A Mesa
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
29
|
Brambilla C, Laffaire J, Lantuejoul S, Moro-Sibilot D, Mignotte H, Arbib F, Toffart AC, Petel F, Hainaut P, Rousseaux S, Khochbin S, de Reyniès A, Brambilla E. Lung squamous cell carcinomas with basaloid histology represent a specific molecular entity. Clin Cancer Res 2014; 20:5777-86. [PMID: 25189482 DOI: 10.1158/1078-0432.ccr-14-0459] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The basaloid carcinoma (pure) and the (mixed) basaloid variant of lung squamous cell carcinoma (SCC) have a dismal prognosis but their underlying specific molecular characteristics remain obscure and no therapy has proven to be efficient. EXPERIMENTAL DESIGN To assess their molecular specificity among other lung SCCs we analyzed DNA copy number aberrations and mRNA expression pangenomic profiles of 93 SCCs, including 42 basaloid samples (24 pure, 18 mixed). RESULTS Supervised analyses reveal that pure basaloid tumors display a specific mRNA expression profile, encoding factors controlling the cell cycle, transcription, chromatin, and splicing, with prevalent expression in germline and stem cells, while genes related to squamous differentiation are underexpressed. From this signature, we derived a 2-genes (SOX4, IVL) immunohistochemistry-based predictor that discriminated basaloid tumors (pure and mixed) from non-basaloid tumors with 94% accuracy in an independent series. The pure basaloid tumors are also distinguished through unsupervised analyses. Using a centroid-based predictor, the corresponding molecular subtype was found in 8 independent public datasets (n = 58/533), and was shown to be associated with a very poor survival as compared with other SCCs (adjusted HR = 2.45; P = 0.000001). CONCLUSION This study enlightens the heterogeneity of SCCs that can be subclassified in mRNA expression subtypes. This study demonstrates for the first time that basaloid SCCs constitute a distinct histomolecular entity, which justifies its recognition and distinction from non-basaloid SCCs. In addition, their characteristic molecular profile highlights their intrinsic resistance to cytotoxic chemotherapy and could serve as a guide for targeted therapies.
Collapse
Affiliation(s)
- Christian Brambilla
- Clinique de Pneumologie, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France
| | - Julien Laffaire
- Cartes d'Identité des Tumeurs (CIT) Program, Ligue Nationale Contre le Cancer, Paris, France
| | - Sylvie Lantuejoul
- Département d'Anatomie et Cytologie Pathologique, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France
| | - Denis Moro-Sibilot
- Clinique de Pneumologie, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France
| | - Hélène Mignotte
- Clinique de Pneumologie, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France
| | - François Arbib
- Clinique de Pneumologie, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France
| | - Anne-Claire Toffart
- Clinique de Pneumologie, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France
| | - Fabien Petel
- Cartes d'Identité des Tumeurs (CIT) Program, Ligue Nationale Contre le Cancer, Paris, France
| | - Pierre Hainaut
- International Prevention Research Institute, Lyon, France
| | - Sophie Rousseaux
- INSERM, U823, Université Joseph Fourier, Institut Albert Bonniot, Grenoble, France
| | - Saadi Khochbin
- INSERM, U823, Université Joseph Fourier, Institut Albert Bonniot, Grenoble, France
| | - Aurélien de Reyniès
- Cartes d'Identité des Tumeurs (CIT) Program, Ligue Nationale Contre le Cancer, Paris, France.
| | - Elisabeth Brambilla
- Département d'Anatomie et Cytologie Pathologique, Centre Hospitalier Universitaire de Grenoble, Institut Albert Bonniot, INSERM U823, Université Joseph Fourrier, Grenoble, France.
| |
Collapse
|
30
|
Pieri L, Guglielmelli P, Finazzi G, Vannucchi AM. Givinostat for the treatment of polycythemia vera. Expert Opin Orphan Drugs 2014. [DOI: 10.1517/21678707.2014.934223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
31
|
Ronzoni L, Sonzogni L, Fossati G, Modena D, Trombetta E, Porretti L, Cappellini MD. Modulation of gamma globin genes expression by histone deacetylase inhibitors: an in vitro study. Br J Haematol 2014; 165:714-21. [PMID: 24606390 DOI: 10.1111/bjh.12814] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/05/2014] [Indexed: 02/04/2023]
Abstract
Induction of fetal haemoglobin (HbF) is a promising therapeutic approach for the treatment of β-thalassaemia and sickle cell disease (SCD). Several pharmacological agents, such as hydroxycarbamide (HC) and butyrates, have been shown to induce the γ-globin genes (HBG1, HBG2). However, their therapeutic use is limited due to weak efficacy and an inhibitory effect on erythroid differentiation. Thus, more effective agents are needed. The histone deacetylase (HDAC) inhibitors are potential therapeutic haemoglobin (Hb) inducers able to modulate gene expression through pleiotropic mechanisms. We investigated the effects of a HDAC inhibitor, Givinostat (GVS), on erythropoiesis and haemoglobin synthesis and compared it with sodium butyrate and HC. We used an in vitro erythropoiesis model derived from peripheral CD34⁺ cells of healthy volunteers and SCD donors. GVS effects on erythroid proliferation and differentiation and on Hb synthesis were investigated. We found that GVS at high concentrations delayed erythroid differentiation with no specific effect on HBG1/2 transcription. At a low concentration (1 nmol/l), GVS induced Hb production with no effects on cells proliferation and differentiation. The efficacy of GVS 1 mol/l in Hb induction in vitro was comparable to that of HC and butyrate. Our results support the evaluation of GVS as a new candidate molecule for the treatment of the haemoglobinophathies due to its positive effects on haemoglobin production at low and non-toxic concentrations.
Collapse
Affiliation(s)
- Luisa Ronzoni
- Department of Clinical Sciences and Community, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy; UO Genetica Medica, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Finazzi G, Vannucchi AM, Martinelli V, Ruggeri M, Nobile F, Specchia G, Pogliani EM, Olimpieri OM, Fioritoni G, Musolino C, Cilloni D, Sivera P, Barosi G, Finazzi MC, Tollo SD, Demuth T, Barbui T, Rambaldi A. A phase II study of Givinostat in combination with hydroxycarbamide in patients with polycythaemia vera unresponsive to hydroxycarbamide monotherapy. Br J Haematol 2013; 161:688-694. [PMID: 23573950 DOI: 10.1111/bjh.12332] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 02/26/2013] [Indexed: 12/19/2022]
Abstract
Givinostat, a histone-deacetylase inhibitor (HDACi), inhibits proliferation of cells bearing the JAK2 V617F mutation and has shown significant activity with good tolerability in patients with chronic myeloproliferative neoplasms (MPN). In this multicentre, open-label, phase II study, 44 patients with polycythaemia vera (PV), unresponsive to the maximum tolerated doses (MTD) of hydroxycarbamide (HC), were treated with Givinostat (50 or 100 mg/d) in combination with MTD of HC. The European LeukaemiaNet response criteria were used to assess the primary endpoint after 12 weeks of treatment. Complete or partial response was observed in 55% and 50% of patients receiving 50 or 100 mg of Givinostat, respectively. Control of pruritus was observed in 64% and 67% of patients in the 50 and 100 mg groups, respectively. The combination of Givinostat and HC was well tolerated: eight patients (18%) discontinued, four in each treatment arm; grade 3 adverse events were reported in one patient (4·5%) in each treatment arm. The combined use of Givinostat and HC was safe and clinically effective in HC-unresponsive PV patients.
Collapse
Affiliation(s)
- Guido Finazzi
- Haematology, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Marco Ruggeri
- Department of Cell Therapy and Haematology, San Bortolo Hospital, Vicenza, Italy
| | - Francesco Nobile
- Onco-haematology Department, A.O. "Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | | | - Enrico Maria Pogliani
- Clinical Haematology, New Hospital San.Gerardo, University of Milano Bicocca, Monza, Italy
| | | | | | | | - Daniela Cilloni
- Internal Medicine 2, AOU San Luigi Gonzaga, Orbassano, Italy
| | - Piera Sivera
- SCU Haematology, A.O. "Ordine Mauriziano" Umberto I, Turin, Italy
| | - Giovanni Barosi
- Centre for the Study of Myelofibrosis, IRCCS Policlinico S.Matteo Foundation, Pavia, Italy
| | | | - Silvia Di Tollo
- Clinical R&D Department, Italfarmaco S.p.A., Cinisello Balsamo, Italy
| | - Tim Demuth
- Clinical R&D Department, Italfarmaco S.p.A., Cinisello Balsamo, Italy
| | - Tiziano Barbui
- Haematology, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | | |
Collapse
|
33
|
Amaru Calzada A, Pedrini O, Finazzi G, Leoni F, Mascagni P, Introna M, Rambaldi A, Golay J. Givinostat and hydroxyurea synergize in vitro to induce apoptosis of cells from JAK2V617F myeloproliferative neoplasm patients. Exp Hematol 2013; 41:253-60.e2. [DOI: 10.1016/j.exphem.2012.10.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 10/27/2022]
|
34
|
JAK2 the future: therapeutic strategies for JAK-dependent malignancies. Trends Pharmacol Sci 2012; 33:574-82. [PMID: 22995223 DOI: 10.1016/j.tips.2012.08.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/18/2012] [Accepted: 08/21/2012] [Indexed: 11/23/2022]
Abstract
The Janus kinase (JAK) proteins are a family of intracellular nonreceptor tyrosine kinases involved in cytokine signaling via the JAK-STAT (signal transducers and activators of transcription) pathway. Genetic studies have identified somatic JAK2(V617F) mutations and other mutant alleles that activate JAK-STAT signaling in most patients with myeloproliferative neoplasms (MPNs). As a result, JAK inhibitors have been developed to treat various malignancies and have been shown to be efficacious in both preclinical and clinical settings. However, available ATP-competitive JAK (type I) inhibitors are associated with dose-dependent toxicities, and do not yet reduce disease burden in MPN patients. Recent studies suggest that genetic and epigenetic mechanisms can cause insensitivity to type I JAK inhibitors. Novel therapies include the development of type II JAK inhibitors and the use of alternative strategies to abrogate JAK-STAT signaling, perhaps with histone deacetylase (HDAC) and heat shock protein 90 (HSP90) inhibitors. These innovative therapies may translate to treatment of other diseases that are dependent on JAK signaling, including B-precursor acute lymphoblastic leukemia (B-ALL).
Collapse
|