1
|
Mu H, Zhu X, Jia H, Zhou L, Liu H. Combination Therapies in Chronic Myeloid Leukemia for Potential Treatment-Free Remission: Focus on Leukemia Stem Cells and Immune Modulation. Front Oncol 2021; 11:643382. [PMID: 34055612 PMCID: PMC8155539 DOI: 10.3389/fonc.2021.643382] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Although tyrosine Kinase Inhibitors (TKI) has revolutionized the treatment of chronic myeloid leukemia (CML), patients are not cured with the current therapy modalities. Also, the more recent goal of CML treatment is to induce successful treatment-free remission (TFR) among patients achieving durable deep molecular response (DMR). Together, it is necessary to develop novel, curative treatment strategies. With advancements in understanding the biology of CML, such as dormant Leukemic Stem Cells (LSCs) and impaired immune modulation, a number of agents are now under investigation. This review updates such agents that target LSCs, and together with TKIs, have the potential to eradicate CML. Moreover, we describe the developing immunotherapy for controlling CML.
Collapse
Affiliation(s)
- Hui Mu
- Medical School, Nantong University, Nantong, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Jia
- Medical School, Nantong University, Nantong, China
| | - Lu Zhou
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
2
|
Sasi S, Mohamed M, P C, Yassin MA. Myasthenia Gravis and Myeloproliferative Neoplasms - Mere Association or Paraneoplastic Neurologic Syndrome: A Mini-Review. ACTA BIO-MEDICA : ATENEI PARMENSIS 2021; 92:e2021437. [PMID: 35075066 PMCID: PMC8823564 DOI: 10.23750/abm.v92i6.12180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 08/26/2021] [Indexed: 01/17/2023]
Abstract
Myasthenia Gravis (MG) is a rare neurological condition characterized by muscle weakness that worsens after use. Myeloproliferative Neoplasms (MPNs) are disorders due to stem-cell hyperplasia characterized by an increased peripheral blood cell count, overactive bone marrow, and proliferation of mature hematopoietic cells. MPNs may be Philadelphia (Ph) chromosome-positive or Negative .A systematic review of case reports was conducted by searching PubMed, Scopus, and Google scholar to identify case reports in which there is an association between MG and MPN and know whether MG can be considered a possible neurological paraneoplastic syndrome in patients with MPNs. A total of 13 cases of MPNs associated with MG were identified. The most common type of MPN associated with MG was chronic myeloid leukemia (CML) (10 out of 13 patients). In most of the patients, MG symptoms appeared after a diagnosis of MPN was made. Considering that 10 out of the 13 patients in our cohort had positive auto-antibodies though only 4 of them had thymic hyperplasia, we hypothesize that bone marrow proliferation was responsible for the production of autoantibodies in these patients.As the clonal cell population cannot be eliminated entirely in the bone marrow even after treatment with tyrosine kinase inhibitors (TKI) in Ph +ve MPNs and JAK2 inhibitors in Ph -ve MPNS, MG can occur even in patients who are treated with these agents. A high index of suspicion is needed to diagnose it early, and treatment should be initiated immediately with steroids and anticholinergic agents.
Collapse
Affiliation(s)
- Sreethish Sasi
- Department of Internal Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Mouhand Mohamed
- Department of Internal Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Chitrambika P
- Department of Anaesthesiology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Mohamed A Yassin
- Department of Hematology, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW In this review, we emphasize up-to-date practical cytogenetic and molecular aspects of chronic myeloid leukemia (CML) and summarize current knowledge on tyrosine kinase inhibitor (TKI) resistance and treatment response monitoring of CML. RECENT FINDINGS The introduction of TKIs has changed the natural course of CML and markedly improved patient survival. Over the past decades, many research efforts were devoted to elucidating the leukemogenic mechanisms of BCR-ABL1 and developing novel TKIs. More recent studies have attempted to answer new questions that have emerged in the TKI era, such as the cytogenetic and molecular bases of treatment failure and disease progression, the clinical impact of genetic aberrations in Philadelphia chromosome (Ph)-positive and Ph-negative cells, and the biological significance of Ph secondarily acquired during therapy of other hematological neoplasms. Recent progresses in the understanding of the cytogenetic and molecular mechanisms underlying therapeutic failure and disease progression have improved the risk stratification of CML and will be helpful in the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0072, Houston, TX, 77030, USA
| | - Shimin Hu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0072, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
El Eit R, Itani AR, Nassar F, Rasbieh N, Jabbour M, Santina A, Zaatari G, Mahon FX, Bazarbachi A, Nasr R. Antitumor efficacy of arsenic/interferon in preclinical models of chronic myeloid leukemia resistant to tyrosine kinase inhibitors. Cancer 2019; 125:2818-2828. [PMID: 31034603 DOI: 10.1002/cncr.32130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/12/2019] [Accepted: 03/05/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) are the standard treatment for chronic myeloid leukemia (CML). Despite their clinical success, TKIs are faced with challenges such as treatment resistance, which may be driven by kinase domain mutations, and frequent disease relapse upon the cessation of treatment. The combination of arsenic trioxide (ATO) and interferon-α (IFN) was previously demonstrated to inhibit proliferation and induce apoptosis in CML cell lines, prolong the survival of primary wild-type CML mice, and dramatically decrease the activity of leukemia-initiating cells (LICs). METHODS The ATO/IFN combination was tested in vitro on imatinib (IMN)-resistant K562-R and Ar230-R cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assays were used to evaluate proliferation and apoptosis, respectively. The acridine orange assay was used to assess autophagy, and quantitative reverse transcription-polymerase chain reaction was used to assess the involvement of the hedgehog (Hh) pathway. In vivo, a retroviral transduction/transplantation T315I BCR-ABL CML mouse model was used to assay the effect of the treatment on survival, tumor burden (histopathology and blood counts), and LIC activity (secondary transplantation). RESULTS In vitro, ATO/IFN synergized to inhibit proliferation and induce apoptosis of IMN-resistant cells with variant modes of resistance. Furthermore, the preclinical effects of ATO/IFN were associated with induction of autophagy along with inhibition of the Hh pathway. Most remarkably, ATO/IFN significantly prolonged the survival of primary T315I-CML mice and displayed a dramatic impairment of disease engraftment in secondary mice, which reflected decreased LIC activity. CONCLUSIONS Collectively, the ATO/IFN strategy has been demonstrated to have the potential to lead to durable remissions in TKI-resistant CML preclinical models and to overcome various TKI-specific mechanisms of resistance.
Collapse
Affiliation(s)
- Rabab El Eit
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Abdul Rahman Itani
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Farah Nassar
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Nagham Rasbieh
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Mark Jabbour
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmad Santina
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Ghazi Zaatari
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - François-Xavier Mahon
- French National Institute of Health and Medical Research Unit 876, Laboratory of Hematology and Department of Blood Diseases, University Hospital Center of Bordeaux, Bordeaux Segalen University, Bordeaux, France
| | - Ali Bazarbachi
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon.,Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
5
|
Deregulation of calcium homeostasis in Bcr-Abl-dependent chronic myeloid leukemia. Oncotarget 2018; 9:26309-26327. [PMID: 29899861 PMCID: PMC5995172 DOI: 10.18632/oncotarget.25241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/03/2018] [Indexed: 12/19/2022] Open
Abstract
Background Chronic myeloid leukemia (CML) results from hematopoietic stem cell transformation by the bcr-abl chimeric oncogene, encoding a 210 kDa protein with constitutive tyrosine kinase activity. In spite of the efficiency of tyrosine kinase inhibitors (TKI; Imatinib), other strategies are explored to eliminate CML leukemia stem cells, such as calcium pathways. Results In this work, we showed that Store-Operated Calcium Entry (SOCE) and thrombin induced calcium influx were decreased in Bcr-Abl expressing 32d cells (32d-p210). The 32d-p210 cells showed modified Orai1/STIM1 ratio and reduced TRPC1 expression that could explain SOCE reduction. Decrease in SOCE and thrombin induced calcium entry was associated to reduced Nuclear Factor of Activated T cells (NFAT) nucleus translocation in 32d-p210 cells. We demonstrated that SOCE blockers enhanced cell mobility of 32d-p210 cells and reduced the proliferation rate in both 32d cell lines. TKI treatment slightly reduced the thrombin-induced response, but imatinib restored SOCE to the wild type level. Bcr-Abl is also known to deregulate Protein Kinase C (PKC), which was described to modulate calcium entries. We showed that PKC enhances SOCE and thrombin induced calcium entries in control cells while this effect is lost in Bcr-Abl-expressing cells. Conclusion The tyrosine kinase activity seems to regulate calcium entries probably not directly but through a global cellular reorganization involving a PKC pathway. Altogether, calcium entries are deregulated in Bcr-Abl-expressing cells and could represent an interesting therapeutic target in combination with TKI.
Collapse
|
6
|
Massimino M, Stella S, Tirrò E, Romano C, Pennisi MS, Puma A, Manzella L, Zanghì A, Stagno F, Di Raimondo F, Vigneri P. Non ABL-directed inhibitors as alternative treatment strategies for chronic myeloid leukemia. Mol Cancer 2018; 17:56. [PMID: 29455672 PMCID: PMC5817805 DOI: 10.1186/s12943-018-0805-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/01/2018] [Indexed: 02/07/2023] Open
Abstract
The introduction of ABL Tyrosine Kinase Inhibitors (TKIs) has significantly improved the outcome of Chronic Myeloid Leukemia (CML) patients that, in large part, achieve satisfactory hematological, cytogenetic and molecular remissions. However, approximately 15-20% fail to obtain optimal responses according to the current European Leukemia Network recommendation because of drug intolerance or resistance.Moreover, a plethora of evidence suggests that Leukemic Stem Cells (LSCs) show BCR-ABL1-independent survival. Hence, they are unresponsive to TKIs, leading to disease relapse if pharmacological treatment is discontinued.All together, these biological events generate a subpopulation of CML patients in need of alternative therapeutic strategies to overcome TKI resistance or to eradicate LSCs in order to allow cure of the disease.In this review we update the role of "non ABL-directed inhibitors" targeting signaling pathways downstream of the BCR-ABL1 oncoprotein and describe immunological approaches activating specific T cell responses against CML cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Combined Modality Therapy
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Molecular Targeted Therapy
- Signal Transduction/drug effects
- Treatment Outcome
Collapse
Affiliation(s)
- Michele Massimino
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Stefania Stella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Elena Tirrò
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Chiara Romano
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Maria Stella Pennisi
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Adriana Puma
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy
| | - Antonino Zanghì
- Department of Surgical Medical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Fabio Stagno
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Francesco Di Raimondo
- Division of Hematology and Bone Marrow Transplant, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
- Department of Surgery, Medical and Surgical Specialties, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia, 78, Catania, 95123, Italy.
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, Via Santa Sofia, 78, 95123, Catania, Italy.
| |
Collapse
|
7
|
Atallah E, Ritchie E. Will tyrosine kinase inhibitors be part of the treatment armamentarium for CML in the future? Expert Opin Pharmacother 2018; 19:79-81. [PMID: 29308683 DOI: 10.1080/14656566.2017.1421633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Ehab Atallah
- a Division of Hematology/Oncology , Medical College of Wisconsin and Froedtert Hospital , Milwaukee , WI , USA
| | - Ellen Ritchie
- b Leukemia program , Weill Medical College of Cornell University and The New York Presbyterian Hospital , New York , NY , USA
| |
Collapse
|
8
|
Jain P, Kantarjian HM, Ghorab A, Sasaki K, Jabbour EJ, Nogueras Gonzalez G, Kanagal-Shamanna R, Issa GC, Garcia-Manero G, Kc D, Dellasala S, Pierce S, Konopleva M, Wierda WG, Verstovsek S, Daver NG, Kadia TM, Borthakur G, O'Brien S, Estrov Z, Ravandi F, Cortes JE. Prognostic factors and survival outcomes in patients with chronic myeloid leukemia in blast phase in the tyrosine kinase inhibitor era: Cohort study of 477 patients. Cancer 2017; 123:4391-4402. [PMID: 28743165 DOI: 10.1002/cncr.30864] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/09/2017] [Accepted: 05/31/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Outcomes in patients with chronic myeloid leukemia in blast phase (CML-BP) are historically dismal. Herein, the authors sought to analyze the characteristics, prognostic factors, and survival outcomes in patients with CML-BP in the tyrosine kinase inhibitor (TKI) era. METHODS A total of 477 patients with CML-BP were treated with a TKI at some point during the course of their CML. Cox proportional hazard models identified characteristics that were predictive of survival. Overall survival and failure-free survival were assessed. Optimal cutoff points for specific parameters were identified using classification and regression tree (CART) analysis. RESULTS The median age of the patients was 53 years (range, 16-84 years) and 64% were male. Approximately 80% of patients initially were diagnosed in the chronic phase of CML at a median of 41 months (range, 0.7-298 months) before transformation to CML-BP. De novo CML-BP occurred in 71 patients. Approximately 72% of patients received TKI therapy before CML-BP. The initial therapy for CML-BP included a TKI alone (35%), a TKI with chemotherapy (46%), and non-TKI therapies (19%). The median overall survival was 12 months and the median failure-free survival was 5 months. In multivariate analysis, myeloid immunophenotype, prior TKI, age ≥58 years, lactate dehydrogenase level ≥1227 IU/L, platelet count < 102 K/μL, no history of stem cell transplantation, transition to BP from chronic phase/accelerated phase, and the presence of chromosome 15 aberrations predicted for a significantly increased risk of death. Achievement of major hematologic response and/or complete cytogenetic response to first-line treatment was found to be predictive of better survival. The combination of a TKI with intensive chemotherapy followed by stem cell transplantation appeared to confer the best outcome. CONCLUSIONS Patients with CML-BP continue to pose a therapeutic challenge, have dismal outcomes, and require newer treatment approaches. Cancer 2017;123:4391-402. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Preetesh Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ahmad Ghorab
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias J Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ghayas C Issa
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Devendra Kc
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sara Dellasala
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zeev Estrov
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge E Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
Sun C, Luan S, Zhang G, Wang N, Shao H, Luan C. CEBPA-mediated upregulation of the lncRNA PLIN2 promotes the development of chronic myelogenous leukemia via the GSK3 and Wnt/β-catenin signaling pathways. Am J Cancer Res 2017; 7:1054-1067. [PMID: 28560057 PMCID: PMC5446474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 03/22/2017] [Indexed: 06/07/2023] Open
Abstract
Accumulating evidence has shown that long noncoding RNAs (lncRNAs) are significant regulators of multiple cellular processes, including the development of chronic myelocytic leukemia (CML). However, the mechanism of how the lncRNA PLIN2 affects CML development remains unclear. In this study, we aimed to investigate the potential roles of CEBPA-mediated upregulation of PLIN2 in the process of CML development by regulating the GSK3 and Wnt/β-catenin signaling pathways. We found that both CEBPA and PLIN2 were expressed at significantly higher levels in CML. Simultaneously, we found that CEBPA upregulated the expression of PLIN2 and that there was a positive correlation between CEBPA and PLIN2 in CML patients. CEBPA promoted the progression of CML by upregulating PLIN2. We also found that PLIN2 increased the expression levels of AKT, p-AKT, GSK-3β, β-catenin and Axin2/Conductin as well as promoted the progression of CML via the GSK3 and Wnt/β-catenin signaling pathways in vitro. Furthermore, we found that CEBPA-mediated upregulation of PLIN2 expression promotes tumor growth via GSK3 and Wnt/β-catenin signaling in vivo. Therefore, our study provided a new theoretical basis for CML treatment through the CEBPA/PLIN2 axis.
Collapse
Affiliation(s)
- Chengming Sun
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical CollegeYantai 264000, Shandong Province, China
| | - Shuping Luan
- Rongcheng Shidao People’s HospitalWeihai 264200, Shandong Province, China
| | - Guili Zhang
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical CollegeYantai 264000, Shandong Province, China
| | - Na Wang
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical CollegeYantai 264000, Shandong Province, China
| | - Huiyuan Shao
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical CollegeYantai 264000, Shandong Province, China
| | - Caifu Luan
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University Medical CollegeYantai 264000, Shandong Province, China
| |
Collapse
|
10
|
Enhanced targeting of CML stem and progenitor cells by inhibition of porcupine acyltransferase in combination with TKI. Blood 2016; 129:1008-1020. [PMID: 28011678 DOI: 10.1182/blood-2016-05-714089] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 12/16/2016] [Indexed: 12/22/2022] Open
Abstract
Tyrosine kinase inhibitor (TKI) treatment of chronic myeloid leukemia (CML) has limited efficacy against leukemia stem cells (LSC) responsible for disease propagation, and most CML patients require continued TKI treatment to maintain remission. LSC maintenance is related, at least in part, to signals from the bone marrow microenvironment (BMM). Our previous studies have shown that Wnt signaling from the BMM contributes to preservation of CML LSC following TKI treatment. Secretion of Wnt ligands requires their modification by the O-acyl transferase Porcupine (PORCN). Here we investigated the activity of a potent and selective PORCN inhibitor, WNT974, against CML stem and progenitor cells. WNT974 efficiently antagonized Wnt signaling in human CML CD34+ cells, and in combination with the TKI nilotinib (NIL) significantly enhanced inhibition of proliferation and colony-forming potential of CML stem and progenitor cells and reduced their growth in immunodeficient mice in vivo, in comparison with NIL alone. Treatment of transgenic CML mice in vivo with NIL in combination with WNT974 significantly reduced leukemic stem and progenitor cell numbers, reduced regeneration of leukemic long-term hematopoietic stem cells in secondary transplant recipients, and enhanced survival of mice after discontinuation of treatment, in comparison with NIL alone. CML progenitors demonstrated enhanced sensitivity to Wnt stimulation, associated with increased expression of the FZD4 receptor. FZD4 knockdown inhibited CML progenitor growth. These results support further investigation of PORCN targeting to inhibit Wnt secretion and signaling and enhance targeting of CML stem cells while sparing their normal counterparts.
Collapse
|
11
|
Xie H, Peng C, Huang J, Li BE, Kim W, Smith EC, Fujiwara Y, Qi J, Cheloni G, Das PP, Nguyen M, Li S, Bradner JE, Orkin SH. Chronic Myelogenous Leukemia- Initiating Cells Require Polycomb Group Protein EZH2. Cancer Discov 2016; 6:1237-1247. [PMID: 27630126 DOI: 10.1158/2159-8290.cd-15-1439] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 08/25/2016] [Indexed: 12/14/2022]
Abstract
Tyrosine kinase inhibitors (TKI) have revolutionized chronic myelogenous leukemia (CML) management. Disease eradication, however, is hampered by innate resistance of leukemia-initiating cells (LIC) to TKI-induced killing, which also provides the basis for subsequent emergence of TKI-resistant mutants. We report that EZH2, the catalytic subunit of Polycomb Repressive Complex 2 (PRC2), is overexpressed in CML LICs and required for colony formation and survival and cell-cycle progression of CML cell lines. A critical role for EZH2 is supported by genetic studies in a mouse CML model. Inactivation of Ezh2 in conventional conditional mice and through CRISPR/Cas9-mediated gene editing prevents initiation and maintenance of disease and survival of LICs, irrespective of BCR-ABL1 mutational status, and extends survival. Expression of the EZH2 homolog EZH1 is reduced in EZH2-deficient CML LICs, creating a scenario resembling complete loss of PRC2. EZH2 dependence of CML LICs raises prospects for improved therapy of TKI-resistant CML and/or eradication of disease by addition of EZH2 inhibitors. SIGNIFICANCE This work defines EZH2 as a selective vulnerability for CML cells and their LICs, regardless of BCR-ABL1 mutational status. Our findings provide an experimental rationale for improving disease eradication through judicious use of EZH2 inhibitors within the context of standard-of-care TKI therapy. Cancer Discov; 6(11); 1237-47. ©2016 AACR.See related article by Scott et al., p. 1248This article is highlighted in the In This Issue feature, p. 1197.
Collapse
Affiliation(s)
- Huafeng Xie
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Cong Peng
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Jialiang Huang
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts.,Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Heath, Boston, Massachusetts
| | - Bin E Li
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Woojin Kim
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Elenoe C Smith
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Yuko Fujiwara
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Jun Qi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Giulia Cheloni
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Partha P Das
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Minh Nguyen
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Shaoguang Li
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts. .,Howard Hughes Medical Institute, Boston, Massachusetts
| |
Collapse
|
12
|
Immunological Analyses of Leukemia Stem Cells. Methods Mol Biol 2016. [PMID: 27581137 DOI: 10.1007/978-1-4939-4011-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Traditionally, the intracellular localization and expression levels of specific proteins in CML Leukemia stem cells (LSCs) have been evaluated by fluorescence immunohistochemistry (FIHC). More recently, Duolink(®) in situ PLA technology has opened up a new and more quantitative way to evaluate signal transduction, posttranslational modification, and protein-protein interaction at the single-stem-cell level. This novel methodology, which employs two antibody-based probes, has already increased our understanding of the biology of the rare CML LSC population. In the future, the use of this approach may contribute to the development of novel therapeutics aimed at eradicating CML LSCs in CML patients.
Collapse
|
13
|
Efficacy of the polo-like kinase inhibitor rigosertib, alone or in combination with Abelson tyrosine kinase inhibitors, against break point cluster region-c-Abelson-positive leukemia cells. Oncotarget 2016; 6:20231-40. [PMID: 26008977 PMCID: PMC4653000 DOI: 10.18632/oncotarget.4047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/22/2015] [Indexed: 11/25/2022] Open
Abstract
The potency of Abelson (ABL) tyrosine kinase inhibitors (TKIs) against chronic myeloid leukemia (CML) has been demonstrated. However, ABL TKI resistance can develop. In this study, we investigated the efficacy of a combination therapy including rigosertib (ON 01910.Na), a polo-like kinase (PLK) and phosphoinositide 3-kinase (PI3K) inhibitor, and ABL TKIs. A 72-h rigosertib treatment was found to inhibit cell growth, induce apoptosis, reduce phosphorylation of the breakpoint cluster region-c (BCR)-ABL and its substrate Crk-L, and increase the activities of caspase 3 and poly (ADP-ribose) polymerase (PARP). This combination therapy also exerted a synergistic inhibitory effect on Philadelphia chromosome (Ph)-positive cell proliferation and reduced the phosphorylation of BCR-ABL and Crk-L while increasing that of cleaved PARP and the H2A.X histone. Rigosertib also potently inhibited the growth of ABL TKI-resistant cells, and cotreatment with ABL TKIs and rigosertib induced higher cytotoxicity. These results indicate that rigosertib treatment may be a powerful strategy against ABL TKI-resistant cells and could enhance the cytotoxic effects of ABL TKIs.
Collapse
|
14
|
Bhalla S, Tremblay D, Mascarenhas J. Discontinuing Tyrosine Kinase Inhibitor Therapy in Chronic Myelogenous Leukemia: Current Understanding and Future Directions. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 16:488-494. [PMID: 27406834 DOI: 10.1016/j.clml.2016.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/11/2016] [Accepted: 06/01/2016] [Indexed: 11/29/2022]
Abstract
BCR-ABL1 tyrosine kinase inhibitors (TKIs) have dramatically transformed the treatment of patients with chronic myelogenous leukemia (CML). Given the impressive and sustained response to TKI therapy that the majority of treated patients with CML enjoy, recent studies have explored the potential to achieve treatment-free remission in select patients, which may allow these patients to escape the adverse clinical and financial effects associated with life-long TKI therapy. The results of multiple prospective trials have demonstrated that patients who maintain a deep molecular response for at least 2 years with TKI treatment may be eligible for trial of TKI discontinuation. Mounting data indicates that approximately 40% of those who discontinue therapy on trial will remain in remission at least 1 year after TKI discontinuation; the majority of patients with molecular recurrence relapse within the first 6 months after TKI discontinuation, and TKI retreatment is highly effective in restoring response. Sokol score, duration of TKI therapy, depth of molecular response, and the presence of natural killer cells may all be associated with a higher probability of attaining treatment-free remission. Moving forward, emerging data from ongoing TKI discontinuation trials will allow for appropriate selection of patients with CML eligible for this approach, will expand our current understanding of the CML stem cell, and identify therapeutic interventions capable of effectively deleting the malignant hematopoietic stem cell.
Collapse
Affiliation(s)
- Sheena Bhalla
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Douglas Tremblay
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
15
|
Gugliotta G, Castagnetti F, Breccia M, Gozzini A, Usala E, Carella AM, Rege-Cambrin G, Martino B, Abruzzese E, Albano F, Stagno F, Luciano L, D'Adda M, Bocchia M, Cavazzini F, Tiribelli M, Lunghi M, Pia Falcone A, Musolino C, Levato L, Venturi C, Soverini S, Cavo M, Alimena G, Pane F, Martinelli G, Saglio G, Rosti G, Baccarani M. Rotation of nilotinib and imatinib for first-line treatment of chronic phase chronic myeloid leukemia. Am J Hematol 2016; 91:617-22. [PMID: 26971721 DOI: 10.1002/ajh.24362] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 12/16/2022]
Abstract
The introduction of second-generation tyrosine-kinase inhibitors (TKIs) has generated a lively debate on the choice of first-line TKI in chronic phase, chronic myeloid leukemia (CML). Despite the TKIs have different efficacy and toxicity profiles, the planned use of two TKIs has never been investigated. We report on a phase 2 study that was designed to evaluate efficacy and safety of a treatment alternating nilotinib and imatinib, in newly diagnosed BCR-ABL1 positive, chronic phase, CML patients. One hundred twenty-three patients were enrolled. Median age was 56 years. The probabilities of achieving a complete cytogenetic response, a major molecular response, and a deep molecular response (MR 4.0) by 2 years were 93%, 87%, and 61%, respectively. The 5-year overall survival and progression-free survival were 89%. Response rates and survival are in the range of those reported with nilotinib alone. Moreover, we observed a relatively low rate of cardiovascular adverse events (5%). These data show that the different efficacy and toxicity profiles of TKIs could be favorably exploited by alternating their use. Am. J. Hematol. 91:617-622, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gabriele Gugliotta
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | - Fausto Castagnetti
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | | | - Antonella Gozzini
- Chair of Hematology, “Careggi” Hospital, University of Florence; Florence Italy
| | - Emilio Usala
- Hematology Unit, “A. Businco” Hospital; Cagliari Italy
| | - Angelo M. Carella
- IRCCS AOU San Martino-IST, Hematology and Bone Marrow Transplantation Unit; Genova Italy
| | - Giovanna Rege-Cambrin
- Chair of Hematology; Department of Clinical and Biological Sciences; “S. Luigi Gonzaga” University Hospital, University of Torino; Orbassano (Torino) Italy
| | - Bruno Martino
- Hematology Unit, “Bianchi-Melacrino-Morelli” Hospital; Reggio Calabria Italy
| | | | - Francesco Albano
- Chair of Hematology; Department of Emergency and Organ Transplantation; University of Bari; Bari Italy
| | - Fabio Stagno
- Chair of Hematology, “Ferrarotto” Hospital, University of Catania; Catania Italy
| | - Luigia Luciano
- Chair of Hematology; Department of Biochemistry and Medical Biotechnologies; “Federico II” University; Naples Italy
| | | | - Monica Bocchia
- Chair of Hematology; “S.Maria alle Scotte” Hospital, Siena, Italy, University of Siena
| | - Francesco Cavazzini
- Chair of Hematology; “S. Anna” Hospital, University of Ferrara; Ferrara Italy
| | | | - Monia Lunghi
- Chair of Hematology; “A. Avogadro” University of Eastern Piedmont; Novara Italy
| | - Antonietta Pia Falcone
- Hematology Unit; IRCCS “Ospedale Casa Sollievo della Sofferenza”; S.Giovanni Rotondo Italy
| | | | - Luciano Levato
- Hematology Unit; “Pugliese-Ciaccio” Hospital; Catanzaro Italy
| | - Claudia Venturi
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | - Simona Soverini
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | - Michele Cavo
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | | | - Fabrizio Pane
- Chair of Hematology; Department of Biochemistry and Medical Biotechnologies; “Federico II” University; Naples Italy
| | - Giovanni Martinelli
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | - Giuseppe Saglio
- Chair of Hematology; Department of Clinical and Biological Sciences; “S. Luigi Gonzaga” University Hospital, University of Torino; Orbassano (Torino) Italy
| | - Gianantonio Rosti
- Institute of Hematology “L. and A. Seràgnoli”, Department of Experimental Diagnostic and Specialty Medicine, “S. Orsola-Malpighi” Hospital, University of Bologna; Bologna Italy
| | - Michele Baccarani
- Department of Hematology and Oncology “L. and A. Seràgnoli”; University of Bologna; Bologna Italy
| | | |
Collapse
|
16
|
Mughal TI, Radich JP, Deininger MW, Apperley JF, Hughes TP, Harrison CJ, Gambacorti-Passerini C, Saglio G, Cortes J, Daley GQ. Chronic myeloid leukemia: reminiscences and dreams. Haematologica 2016; 101:541-58. [PMID: 27132280 PMCID: PMC5004358 DOI: 10.3324/haematol.2015.139337] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/20/2016] [Indexed: 12/26/2022] Open
Abstract
With the deaths of Janet Rowley and John Goldman in December 2013, the world lost two pioneers in the field of chronic myeloid leukemia. In 1973, Janet Rowley, unraveled the cytogenetic anatomy of the Philadelphia chromosome, which subsequently led to the identification of the BCR-ABL1 fusion gene and its principal pathogenetic role in the development of chronic myeloid leukemia. This work was also of major importance to support the idea that cytogenetic changes were drivers of leukemogenesis. John Goldman originally made seminal contributions to the use of autologous and allogeneic stem cell transplantation from the late 1970s onwards. Then, in collaboration with Brian Druker, he led efforts to develop ABL1 tyrosine kinase inhibitors for the treatment of patients with chronic myeloid leukemia in the late 1990s. He also led the global efforts to develop and harmonize methodology for molecular monitoring, and was an indefatigable organizer of international conferences. These conferences brought together clinicians and scientists, and accelerated the adoption of new therapies. The abundance of praise, tributes and testimonies expressed by many serve to illustrate the indelible impressions these two passionate and affable scholars made on so many people's lives. This tribute provides an outline of the remarkable story of chronic myeloid leukemia, and in writing it, it is clear that the historical triumph of biomedical science over this leukemia cannot be considered without appreciating the work of both Janet Rowley and John Goldman.
Collapse
MESH Headings
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cytogenetic Analysis/history
- Cytogenetic Analysis/methods
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/chemistry
- Fusion Proteins, bcr-abl/genetics
- History, 20th Century
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Molecular Targeted Therapy/history
- Molecular Targeted Therapy/methods
- Mutation
- Philadelphia Chromosome
- Prognosis
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Research/history
Collapse
Affiliation(s)
| | - Jerald P Radich
- Fredrick Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | | | | - George Q Daley
- Boston Children's Hospital, Harvard Medicine, School, Boston, MA, USA
| |
Collapse
|
17
|
Naka K, Ishihara K, Jomen Y, Jin CH, Kim DH, Gu YK, Jeong ES, Li S, Krause DS, Kim DW, Bae E, Takihara Y, Hirao A, Oshima H, Oshima M, Ooshima A, Sheen YY, Kim SJ, Kim DK. Novel oral transforming growth factor-β signaling inhibitor EW-7197 eradicates CML-initiating cells. Cancer Sci 2016; 107:140-8. [PMID: 26583567 PMCID: PMC4768399 DOI: 10.1111/cas.12849] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/11/2015] [Accepted: 11/12/2015] [Indexed: 12/17/2022] Open
Abstract
Recent strategies for treating CML patients have focused on investigating new combinations of tyrosine kinase inhibitors (TKIs) as well as identifying novel translational research agents that can eradicate CML leukemia‐initiating cells (CML‐LICs). However, little is known about the therapeutic benefits such CML‐LIC targeting therapies might bring to CML patients. In this study, we investigated the therapeutic potential of EW‐7197, an orally bioavailable transforming growth factor‐β signaling inhibitor which has recently been approved as an Investigational New Drug (NIH, USA), to suppress CML‐LICs in vivo. Compared to TKI treatment alone, administration of TKI plus EW‐7197 to CML‐affected mice significantly delayed disease relapse and prolonged survival. Notably, combined treatment with EW‐7197 plus TKI was effective in eliminating CML‐LICs even if they expressed the TKI‐resistant T315I mutant BCR‐ABL1 oncogene. Collectively, these results indicate that EW‐7197 may be a promising candidate for a new therapeutic that can greatly benefit CML patients by working in combination with TKIs to eradicate CML‐LICs.
Collapse
Affiliation(s)
- Kazuhito Naka
- Exploratory Project on Cancer Stem Cells, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kaori Ishihara
- Exploratory Project on Cancer Stem Cells, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Yoshie Jomen
- Exploratory Project on Cancer Stem Cells, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Cheng Hua Jin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Dong-Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Korea
| | - Yoon-Kang Gu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Korea
| | - Eun-Sook Jeong
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Korea
| | - Shaoguang Li
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Daniela S Krause
- Institute for Tumor Biology and Experimental Therapy, Georg Speyer Haus, Goethe University, Frankfurt, Germany
| | - Dong-Wook Kim
- Department of Hematology, Seoul St. Mary's Hospital, Cancer Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Eunjin Bae
- Exploratory Project on Cancer Stem Cells, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,CHA Cancer Institute and Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Yoshihiro Takihara
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Akira Ooshima
- CHA Cancer Institute and Department of Biomedical Science, CHA University, Seongnam, Korea
| | | | - Seong-Jin Kim
- CHA Cancer Institute and Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Dae-Kee Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
18
|
Russo D, Malagola M, Skert C, Cancelli V, Turri D, Pregno P, Bergamaschi M, Fogli M, Testoni N, De Vivo A, Castagnetti F, Pungolino E, Stagno F, Breccia M, Martino B, Intermesoli T, Cambrin GR, Nicolini G, Abruzzese E, Tiribelli M, Bigazzi C, Usala E, Russo S, Russo-Rossi A, Lunghi M, Bocchia M, D'Emilio A, Santini V, Girasoli M, Lorenzo RD, Bernardi S, Palma AD, Cesana BM, Soverini S, Martinelli G, Rosti G, Baccarani M. Managing chronic myeloid leukaemia in the elderly with intermittent imatinib treatment. Blood Cancer J 2015; 5:e347. [PMID: 26383820 PMCID: PMC4648524 DOI: 10.1038/bcj.2015.75] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/21/2015] [Indexed: 01/18/2023] Open
Abstract
The aim of this study was to investigate the effects of a non-standard, intermittent imatinib treatment in elderly patients with Philadelphia-positive chronic myeloid leukaemia and to answer the question on which dose should be used once a stable optimal response has been achieved. Seventy-six patients aged ⩾65 years in optimal and stable response with ⩾2 years of standard imatinib treatment were enrolled in a study testing a regimen of intermittent imatinib (INTERIM; 1-month on and 1-month off). With a minimum follow-up of 6 years, 16/76 patients (21%) have lost complete cytogenetic response (CCyR) and major molecular response (MMR), and 16 patients (21%) have lost MMR only. All these patients were given imatinib again, the same dose, on the standard schedule and achieved again CCyR and MMR or an even deeper molecular response. The probability of remaining on INTERIM at 6 years was 48% (95% confidence interval 35-59%). Nine patients died in remission. No progressions were recorded. Side effects of continuous treatment were reduced by 50%. In optimal and stable responders, a policy of intermittent imatinib treatment is feasible, is successful in about 50% of patients and is safe, as all the patients who relapsed could be brought back to optimal response.
Collapse
Affiliation(s)
- D Russo
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - M Malagola
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - C Skert
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - V Cancelli
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - D Turri
- Ematologia 1-TMO, AOR Villa Sofia-Cervello, Palermo, Italy
| | - P Pregno
- S.C. Ematologia, Dipartimento di Oncologia ed Ematologia, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
| | - M Bergamaschi
- Dipartimento di Terapie Oncologiche Integrate, IRCCS AOU S. Martino-IST, Genova, Italy
| | - M Fogli
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - N Testoni
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - A De Vivo
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - F Castagnetti
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - E Pungolino
- Division of Hematology, Department of Oncology and Hematology, Niguarda Ca' Granda Hospital, Milan, Italy
| | - F Stagno
- Divisione Clinicizzata di Ematologia AOU Policlinico-V. Emanuele, University of Catania, Catania, Italy
| | - M Breccia
- Azienda Policlinico Umberto I, Sapienza Università, Roma, Italy
| | - B Martino
- Hematology Unit, ‘Bianchi-Melacrino-Morelli' Hospital, Reggio Calabria, Italy
| | - T Intermesoli
- Hematology and Bone Marrow Transplant Unit, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - G R Cambrin
- University of Turin, San Luigi Gonzaga Hospital, Turin, Italy
| | - G Nicolini
- Hematology and Hematopoietic Stem Cell Transplant Center, San Salvatore Hospital, Pesaro, Italy
| | - E Abruzzese
- Hematology, S Eugenio Hospital Tor Vergata University, Rome, Italy
| | - M Tiribelli
- Division of Hematology and BMT, Azienda Ospedaliero—Universitaria di Udine, Udine, Italy
| | - C Bigazzi
- Hematology, Mazzoni Hospital, Ascoli Piceno, Italy
| | - E Usala
- U O Ematologia e CTMO Ospedale A., Businco-Cagliari, Italy
| | - S Russo
- UOC Ematologia AOU 'G Martino' Policlinico Universitario di Messina, Messina, Italy
| | - A Russo-Rossi
- Division of Hematology, University of Bari, Bari, Italy
| | - M Lunghi
- Division of Hematology, Department of Clinical and Experimental Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - M Bocchia
- Hematology and Transplants, University of Siena and AOUS, Siena, Italy
| | - A D'Emilio
- Department of Cellular Therapies and Haematology, San Bortolo Hospital, Vicenza, Italy
| | - V Santini
- Unità di Ematologia, AOU Careggi, University of Florence, Florence, Italy
| | - M Girasoli
- Hematology Department, 'A. Perrino' Hospital, Brindisi, Italy
| | - R Di Lorenzo
- Division of Haematology, Spirito Santo Hospital, Pescara, Italy
| | - S Bernardi
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - A Di Palma
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - B M Cesana
- DMMT, Unit of Medical Statistics, University of Brescia, Brescia, Italy
| | - S Soverini
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - G Martinelli
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - G Rosti
- Institute of Hematology 'L. & A. Seràgnoli', DIMES, University of Bologna, Bologna, Italy
| | - M Baccarani
- Department of Haematology-Oncology 'L. and A. Seràgnoli' – S. Orsola Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Anti-leukemic activity of axitinib against cells harboring the BCR-ABL T315I point mutation. J Hematol Oncol 2015; 8:97. [PMID: 26239229 PMCID: PMC4523922 DOI: 10.1186/s13045-015-0190-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/17/2015] [Indexed: 01/29/2023] Open
Abstract
The BCR-ABL; breakpoint cluster region-Abelson point mutation T315I is resistant to ABL tyrosine kinase inhibitors. However, axitinib, a vascular endothelial growth factor receptor inhibitor, is effective against this mutation. In this study, we investigated axitinib activity against ponatinib-resistant cells and found that axitinib inhibited cellular growth and apoptosis in Ba/F3 T315I-mutant cells and T315I-mutant primary samples, but not in ponatinib-resistant Ba/F3 cells and primary samples. Thus, an alternative strategy may be required to improve the prognosis of Philadelphia-chromosome-positive leukemia patients harboring BCR-ABL point mutations.
Collapse
|
20
|
Fava C, Morotti A, Dogliotti I, Saglio G, Rege-Cambrin G. Update on emerging treatments for chronic myeloid leukemia. Expert Opin Emerg Drugs 2015; 20:183-96. [DOI: 10.1517/14728214.2015.1031217] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
21
|
Bartley PA, Latham S, Budgen B, Ross DM, Hughes E, Branford S, White D, Hughes TP, Morley AA. A DNA real-time quantitative PCR method suitable for routine monitoring of low levels of minimal residual disease in chronic myeloid leukemia. J Mol Diagn 2014; 17:185-92. [PMID: 25554588 DOI: 10.1016/j.jmoldx.2014.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/15/2014] [Accepted: 10/20/2014] [Indexed: 12/20/2022] Open
Abstract
The BCR-ABL1 sequence has advantages over the BCR-ABL1 transcript as a molecular marker in chronic myeloid leukemia and has been used in research studies. We developed a DNA real-time quantitative PCR (qPCR) method for quantification of BCR-ABL1 sequences, which is also potentially suitable for routine use. The BCR-ABL1 breakpoint was sequenced after isolation by nested short-range PCR of DNA from blood, marrow, and cells on slides, obtained either at diagnosis or during treatment, or from artificial mixtures. PCR primers were chosen from a library of presynthesized and pretested BCR (n = 19) and ABL1 (n = 568) primers. BCR-ABL1 sequences were quantified relative to BCR sequences in 521 assays on 266 samples from 92 patients. For minimal residual disease detectable by DNA qPCR and RT-qPCR, DNA qPCR gave similar minimal residual disease results as RT-qPCR but had better precision at low minimal residual disease levels. The limit of detection of DNA qPCR depended on the amount of DNA assayed, being 10(-5.8) when 5 μg was assayed and 10(-7.0) when 80 μg was assayed. DNA qPCR may be useful and practical for monitoring the increasing number of patients with minimal residual disease around or below the limit of detection of RT-qPCR as the assay itself is simple and the up-front costs will be amortized if sequential assays are performed.
Collapse
Affiliation(s)
- Paul A Bartley
- Department of Haematology and Genetic Pathology, School of Medicine, Flinders University and Medical Centre, Bedford Park, South Australia, Australia
| | - Susan Latham
- Department of Haematology and Genetic Pathology, School of Medicine, Flinders University and Medical Centre, Bedford Park, South Australia, Australia
| | - Bradley Budgen
- Department of Haematology and Genetic Pathology, School of Medicine, Flinders University and Medical Centre, Bedford Park, South Australia, Australia
| | - David M Ross
- Department of Haematology and Genetic Pathology, School of Medicine, Flinders University and Medical Centre, Bedford Park, South Australia, Australia; Haematology Division, SA Pathology, Adelaide, South Australia, Australia
| | - Elizabeth Hughes
- Department of Haematology and Genetic Pathology, School of Medicine, Flinders University and Medical Centre, Bedford Park, South Australia, Australia
| | - Susan Branford
- Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Deborah White
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Timothy P Hughes
- Haematology Division, SA Pathology, Adelaide, South Australia, Australia
| | - Alexander A Morley
- Department of Haematology and Genetic Pathology, School of Medicine, Flinders University and Medical Centre, Bedford Park, South Australia, Australia; Monoquant Pty. Ltd., Adelaide, South Australia, Australia.
| |
Collapse
|
22
|
Taverna S, Amodeo V, Saieva L, Russo A, Giallombardo M, De Leo G, Alessandro R. Exosomal shuttling of miR-126 in endothelial cells modulates adhesive and migratory abilities of chronic myelogenous leukemia cells. Mol Cancer 2014; 13:169. [PMID: 25015105 PMCID: PMC4105877 DOI: 10.1186/1476-4598-13-169] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/03/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Recent findings indicate that exosomes released from cancer cells contain microRNAs (miRNAs) that may be delivered to cells of tumor microenvironment. RESULTS To elucidate whether miRNAs secreted from chronic myelogenous leukemia cells (CML) are shuttled into endothelial cells thus affecting their phenotype, we first analysed miRNAs content in LAMA84 exosomes. Among the 124 miRNAs identified in LAMA84 exosomes, we focused our attention on miR-126 which was found to be over-overexpressed in exosomes compared with producing parental cells. Transfection of LAMA84 with Cy3-labelled miR-126 and co-culture of leukemia cells with endothelial cells (EC) confirmed that miR-126 is shuttled into HUVECs. The treatment of HUVECs with LAMA84 exosomes for 24 hours reduced CXCL12 and VCAM1 expression, both at the mRNA and protein level, and negatively modulated LAMA84 motility and cells adhesion. Transfection in HUVECs of miR-126 inhibitor reversed the decrease of CXCL12 and restored the motility and adhesion of LAMA84 cells while the over-expression of miR-126, showed opposite effects. CONCLUSION Our results show that the miR-126 shuttled by exosomes is biologically active in the target cells, and support the hypothesis that exosomal miRNAs have an important role in tumor-endothelial crosstalk occurring in the bone marrow microenvironment, potentially affecting disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Riccardo Alessandro
- Dipartimento di Biopatologia e Metodologie Biomediche, Sezione di Biologia e Genetica, Università di Palermo, Palermo, Italy.
| |
Collapse
|
23
|
Hsieh MY, Van Etten RA. IKK-dependent activation of NF-κB contributes to myeloid and lymphoid leukemogenesis by BCR-ABL1. Blood 2014; 123:2401-11. [PMID: 24464015 PMCID: PMC3983614 DOI: 10.1182/blood-2014-01-547943] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 01/17/2014] [Indexed: 11/20/2022] Open
Abstract
The product of the Ph chromosome, the BCR-ABL1 tyrosine kinase activates diverse signaling pathways in leukemic cells from patients with chronic myeloid leukemia (CML) and Ph(+) B-cell acute lymphoblastic leukemia (B-ALL). Previous studies showed that nuclear factor κB (NF-κB) is activated in BCR-ABL1-expressing cells, but the mechanism of activation and importance of NF-κB to the pathogenesis of BCR-ABL1-positive myeloid and lymphoid leukemias are unknown. Coexpression of BCR-ABL1 and a superrepressor mutant of inhibitory NF-κB α (IκBαSR) blocked nuclear p65/RelA expression and inhibited the proliferation of Ba/F3 cells and primary BCR-ABL1-transformed B lymphoblasts without affecting cell survival. In retroviral mouse models of CML and B-ALL, coexpression of IκBαSR attenuated leukemogenesis, prolonged survival, and reduced myeloid leukemic stem cells. Coexpression of dominant-negative mutants of IκB kinase α (IKKα)/IKK1 or IKKβ/IKK2 also inhibited lymphoid and myeloid leukemogenesis by BCR-ABL1. Blockade of NF-κB decreased expression of the NF-κB targets c-MYC and BCL-X and increased the sensitivity of BCR-ABL1-transformed lymphoblasts to ABL1 kinase inhibitors. These results demonstrate that NF-κB is activated through the canonical IKK pathway and plays distinct roles in the pathogenesis of myeloid and lymphoid leukemias induced by BCR-ABL1, validating NF-κB and IKKs as targets for therapy of Ph(+) leukemias.
Collapse
MESH Headings
- Animals
- Blotting, Southern
- Blotting, Western
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Enzyme Activation/physiology
- Fluorescent Antibody Technique
- Fusion Proteins, bcr-abl/genetics
- I-kappa B Kinase/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Mice
- Microscopy, Confocal
- NF-kappa B/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- Mo-Ying Hsieh
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA
| | | |
Collapse
|