1
|
De Santis S, Monaldi C, Mancini M, Bruno S, Cavo M, Soverini S. Overcoming Resistance to Kinase Inhibitors: The Paradigm of Chronic Myeloid Leukemia. Onco Targets Ther 2022; 15:103-116. [PMID: 35115784 PMCID: PMC8800859 DOI: 10.2147/ott.s289306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Abstract
Protein kinases (PKs) play crucial roles in cellular proliferation and survival, hence their deregulation is a common event in the pathogenesis of solid and hematologic malignancies. Targeting PKs has been a promising strategy in cancer treatment, and there are now a variety of approved anticancer drugs targeting PKs. However, the phenomenon of resistance remains an obstacle to be addressed and overcoming resistance is a goal to be achieved. Chronic myeloid leukemia (CML) is the first as well as one of the best examples of a cancer that can be targeted by molecular therapy; hence, it can be used as a model disease for other cancers. This review aims to summarize up-to-date knowledge on the main mechanisms implicated in resistance to PK inhibitory therapies and to outline the main strategies that are being explored to overcome resistance. The importance of molecular diagnostics and disease monitoring in counteracting resistance will also be discussed.
Collapse
Affiliation(s)
- Sara De Santis
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
- Correspondence: Sara De Santis Insitute of Hematology “Lorenzo e Ariosto Seràgnoli”, Via Massarenti 9, Bologna, 40138, ItalyTel +39 051 2143791Fax +39 051 2144037 Email
| | - Cecilia Monaldi
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
| | - Manuela Mancini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Samantha Bruno
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
| | - Michele Cavo
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Simona Soverini
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, UO Ematologia ‘Lorenzo e Ariosto Seràgnoli’, Università di Bologna, Bologna, Italy
| |
Collapse
|
2
|
Second-Generation Jak2 Inhibitors for Advanced Prostate Cancer: Are We Ready for Clinical Development? Cancers (Basel) 2021; 13:cancers13205204. [PMID: 34680353 PMCID: PMC8533841 DOI: 10.3390/cancers13205204] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Prostate Cancer (PC) is currently estimated to affect 1 in 9 men and is the second leading cause of cancer in men in the US. While androgen deprivation therapy, which targets the androgen receptor, is one of the front-line therapies for advanced PC and for recurrence of organ-confined PC treated with surgery, lethal castrate-resistant PC develops consistently in patients. PC is a multi-focal cancer with different grade carcinoma areas presenting simultaneously. Jak2-Stat5 signaling pathway has emerged as a potentially highly effective molecular target in PCs with positive areas for activated Stat5 protein. Activated Jak2-Stat5 signaling can be readily targeted by the second-generation Jak2-inhibitors that have been developed for myeloproliferative and autoimmune disorders and hematological malignancies. In this review, we analyze and summarize the Jak2 inhibitors that are currently in preclinical and clinical development. Abstract Androgen deprivation therapy (ADT) for metastatic and high-risk prostate cancer (PC) inhibits growth pathways driven by the androgen receptor (AR). Over time, ADT leads to the emergence of lethal castrate-resistant PC (CRPC), which is consistently caused by an acquired ability of tumors to re-activate AR. This has led to the development of second-generation anti-androgens that more effectively antagonize AR, such as enzalutamide (ENZ). However, the resistance of CRPC to ENZ develops rapidly. Studies utilizing preclinical models of PC have established that inhibition of the Jak2-Stat5 signaling leads to extensive PC cell apoptosis and decreased tumor growth. In large clinical cohorts, Jak2-Stat5 activity predicts PC progression and recurrence. Recently, Jak2-Stat5 signaling was demonstrated to induce ENZ-resistant PC growth in preclinical PC models, further emphasizing the importance of Jak2-Stat5 for therapeutic targeting for advanced PC. The discovery of the Jak2V617F somatic mutation in myeloproliferative disorders triggered the rapid development of Jak1/2-specific inhibitors for a variety of myeloproliferative and auto-immune disorders as well as hematological malignancies. Here, we review Jak2 inhibitors targeting the mutated Jak2V617F vs. wild type (WT)-Jak2 that are currently in the development pipeline. Among these 35 compounds with documented Jak2 inhibitory activity, those with potency against WT-Jak2 hold strong potential for advanced PC therapy.
Collapse
|
3
|
Zhao M, Yang C, Chai S, Yuan Y, Zhang J, Cao P, Wang Y, Xiao X, Wu K, Yan H, Liu J, Sun S. Curcumol and FTY720 synergistically induce apoptosis and differentiation in chronic myelomonocytic leukemia via multiple signaling pathways. Phytother Res 2020; 35:2157-2170. [PMID: 33274566 DOI: 10.1002/ptr.6968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Chronic myelomonocytic leukemia (CML) is a myeloid tumor characterized by MDS (myelodysplastic syndrome) and MPN (myeloproliferative neoplasms). Allogeneic hematopoietic stem cell transplantation, chemotherapy, interferon, and targeted therapy are the main treatment methods for CML. Tyrosine kinase inhibitors (TKIs) are also a treatment option, and patients are currently recommended to take these drugs throughout their lives to prevent CML recurrence. Therefore, there is a need to investigate and identify other potential chemotherapy drugs. Currently, research on CML treatment with a single drug has shown little progress. Fingolimod (FTY720), an FDA-approved drug used to treat relapsing multiple sclerosis, has also shown great potential in the treatment of lymphocytic leukemia. In our study, we find that FTY720 and curcumol have a significant inhibitory effect on K562 cells, K562/ADR cells, and CD34+ cells from CML patients. RNAseq data analysis shows that regulation of apoptosis and differentiation pathways are key pathways in this process. Besides, BCR/ABL-Jak2/STAT3 signaling, PI3K/Akt-Jnk signaling, and activation of BH3-only genes are involved in CML inhibition. In a K562 xenograft mouse model, therapy with curcumol and FTY720 led to significant inhibition of tumor growth and induction of apoptosis. To summarize, curcumol and FTY720 synergistically inhibit proliferation involved in differentiation and induce apoptosis in CML cells. Therefore, synergistic treatment with two drugs could be the next choice of treatment for CML.
Collapse
Affiliation(s)
- Mingri Zhao
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Chaoying Yang
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Siyu Chai
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Yijun Yuan
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Ji Zhang
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Pengfei Cao
- Xiangya Hospital, Central South University, Changsha, China
| | - Yanpeng Wang
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Kunlu Wu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Huiwen Yan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Shuming Sun
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
4
|
Perrotti D, Silvestri G, Stramucci L, Yu J, Trotta R. Cellular and Molecular Networks in Chronic Myeloid Leukemia: The Leukemic Stem, Progenitor and Stromal Cell Interplay. Curr Drug Targets 2017; 18:377-388. [PMID: 27307150 DOI: 10.2174/1389450117666160615074120] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 12/13/2022]
Abstract
The use of imatinib, second and third generation ABL tyrosine kinase inhibitors (TKI) (i.e. dasatinib, nilotinib, bosutinib and ponatinib) made CML a clinically manageable and, in a small percentage of cases, a cured disease. TKI therapy also turned CML blastic transformation into a rare event; however, disease progression still occurs in those patients who are refractory, not compliant with TKI therapy or develop resistance to multiple TKIs. In the past few years, it became clear that the BCRABL1 oncogene does not operate alone to drive disease emergence, maintenance and progression. Indeed, it seems that bone marrow (BM) microenvironment-generated signals and cell autonomous BCRABL1 kinase-independent genetic and epigenetic alterations all contribute to: i. persistence of a quiescent leukemic stem cell (LSC) reservoir, ii. innate or acquired resistance to TKIs, and iii. progression into the fatal blast crisis stage. Herein, we review the intricate leukemic network in which aberrant, but finely tuned, survival, mitogenic and self-renewal signals are generated by leukemic progenitors, stromal cells, immune cells and metabolic microenvironmental conditions (e.g. hypoxia) to promote LSC maintenance and blastic transformation.
Collapse
Affiliation(s)
- Danilo Perrotti
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | | | | | | | | |
Collapse
|
5
|
Rethinking JAK2 inhibition: towards novel strategies of more specific and versatile janus kinase inhibition. Leukemia 2017; 31:1023-1038. [DOI: 10.1038/leu.2017.43] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/16/2016] [Accepted: 01/10/2017] [Indexed: 12/19/2022]
|
6
|
Chakraborty SN, Leng X, Perazzona B, Sun X, Lin YH, Arlinghaus RB. Combination of JAK2 and HSP90 inhibitors: an effective therapeutic option in drug-resistant chronic myelogenous leukemia. Genes Cancer 2016; 7:201-208. [PMID: 27551334 PMCID: PMC4979592 DOI: 10.18632/genesandcancer.111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Recent studies suggest that JAK2 serves as a novel therapeutic target in Bcr-Abl+ chronic myelogenous leukemia (CML). We have reported the existence of an HSP90- associated high molecular weight network complex (HMWNC) that is composed of HSP90 client proteins BCR-ABL, JAK2, and STAT3 in wild type Bcr-Abl+ leukemic cells. Here we showed that the HSP90-HMWNC is present in leukemia cells from CML patients in blast stage, and in Imatinib (IM)-resistant 32Dp210 (T315I) leukemia cells. We found that the HSP90-HMWNC could be disassembled by depleting JAK2 with either Jak2-specific shRNA or treatment with JAK2 inhibitors (TG101209 or Ruxolitinib) and HSP90 inhibitor (AUY922). Combinational treatment with JAK2 and HSP90 inhibitors diminished the activation of BCR-ABL, JAK2 and its downstream targets. As a result, the IM-resistant 32Dp210 T315I cells underwent apoptosis. When administered in mice bearing 32Dp210 T315I leukemia, combinational therapy using Ruxolitinib and AUY922 prolonged the survival significantly. Thus, a combination of JAK2 and HSP90 inhibitors could be a powerful strategy for the treatment of CML, especially in IM-resistant patients.
Collapse
Affiliation(s)
- Sandip N Chakraborty
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Xiaohong Leng
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Bastianella Perazzona
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Xiaoping Sun
- Department of Laboratory Medicine, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Yu-Hsi Lin
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| |
Collapse
|
7
|
Li B, Zhang G, Li C, Li R, Lu J, He Z, Wang Q, Peng Z, Wang J, Dong Y, Zhang C, Tan JQ, Bahri N, Wang Y, Duan C. Lyn mediates FIP1L1-PDGFRA signal pathway facilitating IL-5RA intracellular signal through FIP1L1-PDGFRA/JAK2/Lyn/Akt network complex in CEL. Oncotarget 2016; 8:64984-64998. [PMID: 29029406 PMCID: PMC5630306 DOI: 10.18632/oncotarget.11401] [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: 10/04/2015] [Accepted: 07/26/2016] [Indexed: 11/25/2022] Open
Abstract
The Fip1-like1 (FIP1L1)–platelet-derived growth factor receptor alpha (PDGFRA) (F/P) oncogene can cause chronic eosinophilic leukemia (CEL), but requires IL-5 cytokine participation. In this study, we investigate the mechanism of F/P in collaboration with IL-5 in CEL. The results showed that Lyn, a key effector in the IL-5-motivated eosinophil production, is extensively activated in F/P-positive CEL cells. Lyn can associate and phosphorylate IL-5 receptor α (IL-5RA) in F/P-positive cells. Moreover, the activation of Lyn and IL-5R kinase were strengthened when the cells were stimulated by IL-5. Lyn inhibition in F/P-positive CEL cells attenuated cellular proliferation, induced apoptosis, and blocked cell migration and major basic protein (MBP) release. We identified the FIP1L1-PDGFRA/JAK2/Lyn/Akt complex in the F/P-expressing cells which can be disrupted by dual inhibition of JAK2 and Lyn, repressing cell proliferation in both EOL-1(F/P-positive human eosinophilic cell line) and imatinib-resistance (IR) cells. Altogether, our data demonstrate that Lyn is a vital downstream kinase activated by F/P converged with IL-5 signals in CEL cells. Lyn activate and expand IL-5RA intracellular signaling through FIP1L1-PDGFRA/JAK2/Lyn/Akt network complex, provoking eosinophils proliferation and exaggerated activation manifested as CEL.
Collapse
Affiliation(s)
- Bin Li
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Division of Hematology, Institute of Molecular Hematology, The Second Xiang Ya Hospital, Central South University, Changsha, People's Republic of China.,Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Guangsen Zhang
- Division of Hematology, Institute of Molecular Hematology, The Second Xiang Ya Hospital, Central South University, Changsha, People's Republic of China
| | - Cui Li
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ruijuan Li
- Division of Hematology, Institute of Molecular Hematology, The Second Xiang Ya Hospital, Central South University, Changsha, People's Republic of China
| | - Jingchen Lu
- Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhengxi He
- Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Quan Wang
- Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhenzi Peng
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jun Wang
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yeping Dong
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chunfang Zhang
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jie Qiong Tan
- State Key Laboratory of Medical Genetics, Xiangya Medical School, Central South University, Changsha, People's Republic of China
| | - Nacef Bahri
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuexiang Wang
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,The Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chaojun Duan
- Medical Research Center, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
8
|
Chiarenza A, Manetti F, Petricci E, Ruat M, Naldini A, Taddei M, Carraro F. Novel Acylguanidine Derivatives Targeting Smoothened Induce Antiproliferative and Pro-Apoptotic Effects in Chronic Myeloid Leukemia Cells. PLoS One 2016; 11:e0149919. [PMID: 26934052 PMCID: PMC4774938 DOI: 10.1371/journal.pone.0149919] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/05/2016] [Indexed: 11/18/2022] Open
Abstract
The most relevant therapeutic approaches to treat CML rely on the administration of tyrosine kinase inhibitors (TKIs) like Imatinib, which are able to counteract the activity of Bcr-Abl protein increasing patient's life expectancy and survival. Unfortunately, there are some issues TKIs are not able to address; first of all TKIs are not so effective in increasing survival of patients in blast crisis, second they are not able to eradicate leukemic stem cells (LSC) which represent the major cause of disease relapse, and third patients often develop resistance to TKIs due to mutations in the drug binding site. For all these reasons it's of primary interest to find alternative strategies to treat CML. Literature shows that Hedgehog signaling pathway is involved in LSC maintenance, and pharmacological inhibition of Smoothened (SMO), one of the key molecules of the pathway, has been demonstrated to reduce Bcr-Abl positive bone marrow cells and LSC. Consequently, targeting SMO could be a promising way to develop a new treatment strategy for CML overcoming the limitations of current therapies. In our work we have tested some compounds able to inhibit SMO, and among them MRT92 appears to be a very potent SMO antagonist. We found that almost all our compounds were able to reduce Gli1 protein levels in K-562 and in KU-812 CML cell lines. Furthermore, they were also able to increase Gli1 and SMO RNA levels, and to reduce cell proliferation and induce apoptosis/autophagy in both the tested cell lines. Finally, we demonstrated that our compounds were able to modulate the expression of some miRNAs related to Hedgehog pathway such as miR-324-5p and miR-326. Being Hedgehog pathway deeply implicated in the mechanisms of CML we may conclude that it could be a good therapeutic target for CML and our compounds seem to be promising antagonists of such pathway.
Collapse
MESH Headings
- Apoptosis/drug effects
- Autophagy/drug effects
- Blast Crisis/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Fusion Proteins, bcr-abl/metabolism
- Hedgehog Proteins/metabolism
- Humans
- Imatinib Mesylate/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- MicroRNAs/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction/drug effects
- Smoothened Receptor
Collapse
Affiliation(s)
- Alessandra Chiarenza
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Elena Petricci
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Martial Ruat
- CNRS, UMR-9197, Neuroscience Paris- Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France
| | - Antonella Naldini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Maurizio Taddei
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Fabio Carraro
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Istituto Toscano Tumori, Siena, Italy
- * E-mail:
| |
Collapse
|
9
|
Wang M, Shen A, Zhang C, Song Z, Ai J, Liu H, Sun L, Ding J, Geng M, Zhang A. Development of Heat Shock Protein (Hsp90) Inhibitors To Combat Resistance to Tyrosine Kinase Inhibitors through Hsp90-Kinase Interactions. J Med Chem 2016; 59:5563-86. [PMID: 26844689 DOI: 10.1021/acs.jmedchem.5b01106] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heat shock protein 90 (Hsp90) is a ubiquitous chaperone of all of the oncogenic tyrosine kinases. Many Hsp90 inhibitors, alone or in combination, have shown significant antitumor efficacy against the kinase-positive naïve and mutant models. However, clinical trials of these inhibitors are unsuccessful due to insufficient clinical benefits and nonoptimal safety profiles. Recently, much progress has been reported on the Hsp90-cochaperone-client complex, which will undoubtedly assist in the understanding of the interactions between Hsp90 and its clients. Meanwhile, Hsp90 inhibitors have shown promise against patients' resistance caused by early generation tyrosine kinase inhibitors (TKIs), and at least 13 Hsp90 inhibitors are being reevaluated in the clinic. In this regard, the objectives of the current perspective are to summarize the structure and function of the Hsp90-cochaperone-client complex, to analyze the structural and functional insights into the Hsp90-client interactions to address several existing unresolved problems with Hsp90 inhibitors, and to highlight the preclinical and clinical studies of Hsp90 inhibitors as an effective treatment against resistance to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Meining Wang
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| | - Aijun Shen
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Chi Zhang
- Department of Medicinal Chemistry, China Pharmaceutical University , Nanjing 210009, China
| | - Zilan Song
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| | - Jing Ai
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Hongchun Liu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Liping Sun
- Department of Medicinal Chemistry, China Pharmaceutical University , Nanjing 210009, China
| | - Jian Ding
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Meiyu Geng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| |
Collapse
|
10
|
Mu C, Wu X, Ma H, Tao W, Zhang G, Xia X, Shen J, Mai J, Sun T, Sun X, Arlinghaus RB, Shen H. Effective Concentration of a Multikinase Inhibitor within Bone Marrow Correlates with In Vitro Cell Killing in Therapy-Resistant Chronic Myeloid Leukemia. Mol Cancer Ther 2016; 15:899-910. [PMID: 26846820 DOI: 10.1158/1535-7163.mct-15-0577-t] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 01/25/2016] [Indexed: 12/12/2022]
Abstract
Leukemia cells escape BCR-ABL-targeted therapy by developing mutations, such as T315I, in the p210(BCR-ABL) fusion protein in Philadelphia chromosome-positive chronic myeloid leukemia (CML). Although most effort has been focused on development of new tyrosine kinase inhibitors, enrichment of these small-molecule inhibitors in the tumor tissue can also have a profound impact on treatment outcomes. Here, we report that a 2-hour exposure of the T315I-mutant CML cells to 10 μmol/L of the multikinase inhibitor TG101209 suppressed BCR-ABL-independent signaling and caused cell-cycle arrest at G2-M. Further increase in drug concentration to 17.5 μmol/L blocked phosphorylation of the mutant BCR-ABL kinase and its downstream JAK2 and STAT5. The effective dosage to overcome therapy resistance identified in an in vitro setting serves as a guidance to develop the proper drug formulation for in vivo efficacy. A targeted formulation was developed to achieve sustained bone marrow TG101209 concentration at or above 17.5 μmol/L for effective killing of CML cells in vivo Potent inhibition of leukemia cell growth and extended survival were observed in two murine models of CML treated with 40 mg/kg intravenously administered targeted TG101209, but not with the untargeted drug at the same dosage. Our finding provides a unique approach to develop treatments for therapy-resistant CML. Mol Cancer Ther; 15(5); 899-910. ©2016 AACR.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Aurora Kinase B/antagonists & inhibitors
- Bone Marrow/drug effects
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- 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
- Mice
- Mutation
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- Signal Transduction/drug effects
- Sulfonamides/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chaofeng Mu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Xiaoyan Wu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Helen Ma
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Wenjing Tao
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Guodong Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Xiaojun Xia
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Jianliang Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Tong Sun
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Xiaoping Sun
- Department of Laboratory Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas. Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
11
|
Huo X, Liao Y, Tian Y, Gao L, Cao L. Zeylenone promotes apoptosis in chronic myelogenous leukemia-derived K562 cells by a mechanism involving Jak2 and Src kinase. RSC Adv 2016. [DOI: 10.1039/c6ra23443g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is a hematopoietic malignancy caused by the constitutive activation of BCR–ABL tyrosine kinase.
Collapse
Affiliation(s)
- Xiaowei Huo
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
- China
| | - Yonghong Liao
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
- China
| | - Yu Tian
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
- China
| | - Li Gao
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
- China
| | - Li Cao
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100193
- China
| |
Collapse
|
12
|
Abstract
p53 plays a key role in regulating DNA damage response by suppressing cell cycle progression or inducing apoptosis depending on extent of DNA damage. However, it is not clear why mild genotoxic stress favors growth arrest, whereas excessive lesions signal cells to die. Here we showed that TAp73, a p53 homologue thought to have a similar function as p53, restrains the transcriptional activity of p53 and prevents excessive activation of its downstream targets upon low levels of DNA damage, which results in cell cycle arrest. Extensive DNA damage triggers TAp73 depletion through ubiquitin/proteasome-mediated degradation of E2F1, leading to enhanced transcriptional activation by p53 and subsequent induction of apoptosis. These findings provide novel insights into the regulation of p53 function and suggest that TAp73 keeps p53 activity in check in regulating cell fate decisions upon genotoxic stress.
Collapse
|
13
|
Tao W, Chakraborty SN, Leng X, Ma H, Arlinghaus RB. HSP90 inhibitor AUY922 induces cell death by disruption of the Bcr-Abl, Jak2 and HSP90 signaling network complex in leukemia cells. Genes Cancer 2015; 6:19-29. [PMID: 25821558 PMCID: PMC4362481 DOI: 10.18632/genesandcancer.49] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/28/2015] [Indexed: 11/25/2022] Open
Abstract
The Bcr-Abl protein is an important client protein of heat shock protein 90 (HSP90). We evaluated the inhibitory effects of the HSP90 ATPase inhibitor AUY922 on 32D mouse hematopoietic cells expressing wild-type Bcr-Abl (b3a2, 32Dp210) and mutant Bcr-Abl imatinib (IM)-resistant cell lines. Western blotting results of fractions from gel filtration column chromatography of 32Dp210 cells showed that HSP90 together with Bcr-Abl, Jak2 Stat3 and several other proteins co-eluted in peak column fractions of a high molecular weight network complex (HMWNC). Co-IP results showed that HSP90 directly bound to Bcr-Abl, Jak2, Stat 3 and Akt. The associations between HSP90 and Bcr-Abl or Bcr-Abl kinase domain mutants (T315I and E255K) were interrupted by AUY922 treatment. Tyrosine phosphorylation of Bcr-Abl showed a dose-dependent decrease in 32Dp210T315I following AUY922 treatment for 16h. AUY922 also markedly inhibited cell proliferation of both IM-sensitive 32Dp210 (IC50 =6 nM) and IM-resistant 32Dp210T315I cells (IC50 ≈6 nM) and human KBM-5R/KBM-7R cell lines (IC50 =50 nM). AUY922 caused significant G1 arrest in 32Dp210 cells but not in T315I or E255K cells. AUY922 efficiently induced apoptosis in 32Dp210 (IC50 =10 nM) and T315I or E255K lines with IC50 around 20 to 50 nM. Our results showed that Bcr-Abl and Jak2 form HMWNC with HSP90 in CML cells. Inhibition of HSP90 by AUY922 disrupted the structure of HMWNC, leading to Bcr-Abl degradation, nhibiting cell proliferation and inducing apoptosis. Thus, inhibition of HSP90 is a powerful way to inhibit not only IM-sensitive CML cells but also IM-resistant CML cells.
Collapse
Affiliation(s)
- Wenjing Tao
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Sandip N Chakraborty
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Xiaohong Leng
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Helen Ma
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Okabe S, Tauchi T, Katagiri S, Tanaka Y, Ohyashiki K. Combination of the ABL kinase inhibitor imatinib with the Janus kinase 2 inhibitor TG101348 for targeting residual BCR-ABL-positive cells. J Hematol Oncol 2014; 7:37. [PMID: 24775308 PMCID: PMC4012544 DOI: 10.1186/1756-8722-7-37] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/22/2014] [Indexed: 01/08/2023] Open
Abstract
Background The ABL kinase inhibitor imatinib is highly effective in treating most, but not all, patients with chronic myeloid leukemia (CML). This is because residual CML cells are generally present in the bone marrow microenvironment and are refractory to imatinib. Hematopoietic cytokine receptor signaling is mediated by Janus kinases (JAKs) and their downstream transcription factor, signal transducer and activator of transcription (STAT). TG101348 (SAR302503) is an oral inhibitor of JAK2. Methods We investigated the efficacy of imatinib and TG101348 using the break point cluster region-c-Abelson (BCR-ABL)-positive cell line and primary CML samples wherein leukemia cells were protected by a feeder cell line (HS-5). Results Imatinib treatment resulted in partial inhibition of cell growth in HS-5-conditioned medium. Furthermore, combined treatment with imatinib and TG101348 abrogated the protective effects of HS-5-conditioned medium on K562 cells. Phosphorylation of Crk-L, a BCR-ABL substrate, decreased considerably, while apoptosis increased. In addition, the combined treatment of CD34-positive primary samples resulted in considerably increased cytotoxicity, decreased Crk-L phosphorylation, and increased apoptosis. We also investigated TG101348 activity against feeder cells and observed that STAT5 phosphorylation, granulocyte macrophage colony-stimulating factor, and interleukin 6 levels decreased, indicating reduced cytokine production in HS-5 cells treated with TG101348. Conclusions These results showed that JAK inhibitors may enhance the cytotoxic effect of imatinib against residual CML cells and that a combined approach may be a powerful strategy against the stroma-associated drug resistance of Philadelphia chromosome-positive cells.
Collapse
Affiliation(s)
- Seiichi Okabe
- First Department of Internal Medicine, Tokyo Medical University, Shinjuku-ku, Tokyo 160-0023, Japan.
| | | | | | | | | |
Collapse
|
15
|
Bernt KM, Hunger SP. Current concepts in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia. Front Oncol 2014; 4:54. [PMID: 24724051 PMCID: PMC3971203 DOI: 10.3389/fonc.2014.00054] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/06/2014] [Indexed: 12/22/2022] Open
Abstract
The t(9;22)(q34;q11) or Philadelphia chromosome creates a BCR-ABL1 fusion gene encoding for a chimeric BCR-ABL1 protein. It is present in 3-4% of pediatric acute lymphoblastic leukemia (Ph(+) ALL), and about 25% of adult ALL cases. Prior to the advent of tyrosine kinase inhibitors (TKI), Ph(+) ALL was associated with a very poor prognosis despite the use of intensive chemotherapy and frequently hematopoietic stem-cell transplantation (HSCT) in first remission. The development of TKIs revolutionized the therapy of Ph(+) ALL. Addition of the first generation ABL1 class TKI imatinib to intensive chemotherapy dramatically increased the survival for children with Ph(+) ALL and established that many patients can be cured without HSCT. In parallel, the mechanistic understanding of Ph(+) ALL expanded exponentially through careful mapping of pathways downstream of BCR-ABL1, the discovery of mutations in master regulators of B-cell development such as IKZF1 (Ikaros), PAX5, and early B-cell factor (EBF), the recognition of the complex clonal architecture of Ph(+) ALL, and the delineation of genomic, epigenetic, and signaling abnormalities contributing to relapse and resistance. Still, many important basic and clinical questions remain unanswered. Current clinical trials are testing second generation TKIs in patients with newly diagnosed Ph(+) ALL. Neither the optimal duration of therapy nor the optimal chemotherapy backbone are currently defined. The role of HSCT in first remission and post-transplant TKI therapy also require further study. In addition, it will be crucial to continue to dig deeper into understanding Ph(+) ALL at a mechanistic level, and translate findings into complementary targeted approaches. Expanding targeted therapies hold great promise to decrease toxicity and improve survival in this high-risk disease, which provides a paradigm for how targeted therapies can be incorporated into treatment of other high-risk leukemias.
Collapse
Affiliation(s)
- Kathrin M Bernt
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado , Aurora, CO , USA
| | - Stephen P Hunger
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado , Aurora, CO , USA
| |
Collapse
|
16
|
Ringel F, Kaeda J, Schwarz M, Oberender C, Grille P, Dörken B, Marque F, Manley PW, Radimerski T, le Coutre P. Effects of Jak2 type 1 inhibitors NVP-BSK805 and NVP-BVB808 on Jak2 mutation-positive and Bcr-Abl-positive cell lines. Acta Haematol 2014; 132:75-86. [PMID: 24504330 DOI: 10.1159/000356784] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 10/15/2013] [Indexed: 01/14/2023]
Abstract
Janus kinases are critical components of signaling pathways that regulate hematopoiesis. Mutations of the non-receptor tyrosine kinase JAK2 are found in many BCR-ABL-negative myeloproliferative neoplasms. Preclinical results support that JAK2 inhibitors could show efficacy in treating chronic myeloproliferative neoplasms. JAK2 has also been postulated to play a role in BCR-ABL signal transduction. Therefore, inhibitors of JAK2 kinases are turning into therapeutic strategies for treatment of chronic myelogenous leukemia (CML). In this study, the effects of two novel JAK2 inhibitors, NVP-BSK805 and NVP-BVB808, have been investigated in cell lines expressing either BCR-ABL or mutant JAK2. Possible synergies between NVP-BSK805/NVP-BVB808 and the kinase inhibitors imatinib and nilotinib were assessed. Proliferation and apoptosis tests with both substances showed response in the following cell lines: CHRF-288-11, SET-2 and UKE-1. All BCR-ABL-positive cell lines showed some reduction in proliferation, but with half-maximal growth-inhibitory values >1 µM. Combination of the JAK2 inhibitors with imatinib and nilotinib showed no significant additive or synergistic effects, although all BCR-ABL-positive cell lines responded well to both CML therapeutic agents. Interestingly, it seemed that the combination of imatinib with NVP-BSK805 had a protective effect on the cells. Combination treatment with nilotinib did not show this effect.
Collapse
MESH Headings
- Apoptosis/drug effects
- Benzamides/administration & dosage
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Imatinib Mesylate
- Janus Kinase 2/antagonists & inhibitors
- Janus Kinase 2/genetics
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Mutation
- Myeloproliferative Disorders/drug therapy
- Myeloproliferative Disorders/enzymology
- Myeloproliferative Disorders/genetics
- Phosphorylation/drug effects
- Piperazines/administration & dosage
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/administration & dosage
- Quinoxalines/administration & dosage
- Quinoxalines/pharmacology
- STAT5 Transcription Factor/metabolism
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Frauke Ringel
- Medizinische Klinik m.S. Hämatologie und Onkologie, Campus Virchow Klinikum, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Aggoune D, Tosca L, Sorel N, Bonnet ML, Dkhissi F, Tachdjian G, Bennaceur-Griscelli A, Chomel JC, Turhan AG. Modeling the influence of stromal microenvironment in the selection of ENU-induced BCR-ABL1 mutants by tyrosine kinase inhibitors. Oncoscience 2014; 1:57-68. [PMID: 25593988 PMCID: PMC4295758 DOI: 10.18632/oncoscience.9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 01/20/2014] [Indexed: 01/04/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have profoundly changed the natural history of chronic myeloid leukemia (CML). However, acquired resistance to imatinib, dasatinib or nilotinib (1st and 2nd generation TKIs), due in part to BCR-ABL1 kinase mutations, has been largely described. These drugs are ineffective on the T315I gatekeeper substitution, which remains sensitive to 3rd generation TKI ponatinib. It has recently been suggested that the hematopoietic niche could protect leukemic cells from targeted therapy. In order to investigate the role of a stromal niche in mutation-related resistance, we developed a niche-based cell mutagenesis assay. For this purpose, ENU (N-ethyl-N-nitrosourea)-exposed UT-7 cells expressing non-mutated or T315I-mutated BCR-ABL1 were cultured with or without murine MS-5 stromal cells and in the presence of imatinib, dasatinib, nilotinib, or ponatinib. In the assays relative to 1st and 2nd generation TKIs, which were performed on non-mutated BCR-ABL1 cells, our data highlighted the increasing efficacy of the latter, but did not reveal any substantial effect of the niche. In ponatinib assays performed on both non-mutated and T315I–mutated BCR-ABL1 cells, an increased number of resistant clones were observed in the presence of MS-5. Present data suggested that T315I mutants need either compound mutations (e.g. E255K/T315I) or a stromal niche to escape from ponatinib. Using array-comparative genomic hybridization experiments, we found an increased number of variations (involving some recurrent chromosome regions) in clones cultured on MS-5 feeder. Overall, our study suggests that the hematopoietic niche could play a crucial role in conferring resistance to ponatinib, by providing survival signals and favoring genetic instability.
Collapse
Affiliation(s)
| | - Lucie Tosca
- INSERM, U935, F-94800, Villejuif, France ; Université Paris-Sud 11, F-94270 Le Kremlin-Bicêtre, France ; Hôpital Antoine Béclère, Service d'Histologie-Embryologie-Cytogénétique, F-92140 Clamart, France
| | - Nathalie Sorel
- INSERM, U935, F-86000 Poitiers, France ; CHU de Poitiers, Service de Cancérologie Biologique, F-86000 Poitiers, France
| | | | | | - Gérard Tachdjian
- INSERM, U935, F-94800, Villejuif, France ; Université Paris-Sud 11, F-94270 Le Kremlin-Bicêtre, France ; Hôpital Antoine Béclère, Service d'Histologie-Embryologie-Cytogénétique, F-92140 Clamart, France
| | - Annelise Bennaceur-Griscelli
- INSERM, U935, F-94800, Villejuif, France ; Université Paris-Sud 11, F-94270 Le Kremlin-Bicêtre, France ; Hôpital Paul Brousse, Service d'Hématologie Biologique, F-94800 Villejuif, France
| | - Jean-Claude Chomel
- INSERM, U935, F-86000 Poitiers, France ; CHU de Poitiers, Service de Cancérologie Biologique, F-86000 Poitiers, France
| | - Ali G Turhan
- INSERM, U935, F-86000 Poitiers, France ; INSERM, U935, F-94800, Villejuif, France ; Université Paris-Sud 11, F-94270 Le Kremlin-Bicêtre, France ; Hôpital Bicêtre, Service d'Hématologie Biologique, F-94270 Le Kremlin Bicêtre, France
| |
Collapse
|
18
|
Sinclair A, Latif AL, Holyoake TL. Targeting survival pathways in chronic myeloid leukaemia stem cells. Br J Pharmacol 2014; 169:1693-707. [PMID: 23517124 DOI: 10.1111/bph.12183] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/11/2013] [Accepted: 02/19/2013] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Chronic myeloid leukaemia (CML) is a clonal myeloproliferative disorder characterized by the presence of a fusion oncogene BCR-ABL, which encodes a protein with constitutive TK activity. The implementation of tyrosine kinase inhibitors (TKIs) marked a major advance in CML therapy; however, there are problems with current treatment. For example, relapse occurs when these drugs are discontinued in the majority of patients who have achieved a complete molecular response on TKI and these agents are less effective in patients with mutations in the BCR-ABL kinase domain. Importantly, TKI can effectively target proliferating mature cells, but do not eradicate quiescent leukaemic stem cells (LSCs), therefore allowing disease persistence despite treatment. It is essential that alternative strategies are used to target the LSC population. BCR-ABL activation is responsible for the modulation of different signalling pathways, which allows the LSC fraction to evade cell death. Several pathways have been shown to be modulated by BCR-ABL, including PI3K/AKT/mTOR, JAK-STAT and autophagy signalling pathways. Targeting components of these survival pathways, alone or in combination with TKI, therefore represents an attractive potential therapeutic approach for targeting the LSC. However, many pathways are also active in normal stem cells. Therefore, potential targets must be validated to effectively eradicate CML stem cells while sparing normal counterparts. This review summarizes the main pathways modulated in CML stem cells, the recent developments and the use of novel drugs to target components in these pathways which may be used to target the LSC population. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- A Sinclair
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | | | | |
Collapse
|
19
|
Neviani P, Harb JG, Oaks JJ, Santhanam R, Walker CJ, Ellis JJ, Ferenchak G, Dorrance AM, Paisie CA, Eiring AM, Ma Y, Mao HC, Zhang B, Wunderlich M, May PC, Sun C, Saddoughi SA, Bielawski J, Blum W, Klisovic RB, Solt JA, Byrd JC, Volinia S, Cortes J, Huettner CS, Koschmieder S, Holyoake TL, Devine S, Caligiuri MA, Croce CM, Garzon R, Ogretmen B, Arlinghaus RB, Chen CS, Bittman R, Hokland P, Roy DC, Milojkovic D, Apperley J, Goldman JM, Reid A, Mulloy JC, Bhatia R, Marcucci G, Perrotti D. PP2A-activating drugs selectively eradicate TKI-resistant chronic myeloid leukemic stem cells. J Clin Invest 2013; 123:4144-57. [PMID: 23999433 DOI: 10.1172/jci68951] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 07/03/2013] [Indexed: 12/26/2022] Open
Abstract
The success of tyrosine kinase inhibitors (TKIs) in treating chronic myeloid leukemia (CML) depends on the requirement for BCR-ABL1 kinase activity in CML progenitors. However, CML quiescent HSCs are TKI resistant and represent a BCR-ABL1 kinase-independent disease reservoir. Here we have shown that persistence of leukemic HSCs in BM requires inhibition of the tumor suppressor protein phosphatase 2A (PP2A) and expression--but not activity--of the BCR-ABL1 oncogene. Examination of HSCs from CML patients and healthy individuals revealed that PP2A activity was suppressed in CML compared with normal HSCs. TKI-resistant CML quiescent HSCs showed increased levels of BCR-ABL1, but very low kinase activity. BCR-ABL1 expression, but not kinase function, was required for recruitment of JAK2, activation of a JAK2/β-catenin survival/self-renewal pathway, and inhibition of PP2A. PP2A-activating drugs (PADs) markedly reduced survival and self-renewal of CML quiescent HSCs, but not normal quiescent HSCs, through BCR-ABL1 kinase-independent and PP2A-mediated inhibition of JAK2 and β-catenin. This led to suppression of human leukemic, but not normal, HSC/progenitor survival in BM xenografts and interference with long-term maintenance of BCR-ABL1-positive HSCs in serial transplantation assays. Targeting the JAK2/PP2A/β-catenin network in quiescent HSCs with PADs (e.g., FTY720) has the potential to treat TKI-refractory CML and relieve lifelong patient dependence on TKIs.
Collapse
|
20
|
Seke Etet PF, Vecchio L, Nwabo Kamdje AH. Signaling pathways in chronic myeloid leukemia and leukemic stem cell maintenance: key role of stromal microenvironment. Cell Signal 2012; 24:1883-1888. [PMID: 22659137 DOI: 10.1016/j.cellsig.2012.05.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 05/15/2012] [Accepted: 05/24/2012] [Indexed: 12/11/2022]
Abstract
Chronic myeloid leukemia (CML) is caused by the malignant transformation of hematopoietic stem cells in leukemic stem cells. From the introduction of the anti-cancer drug imatinib, the therapy of CML has been positively transformed. However, following treatment most patients display a residual CML disease attributed to the presence of quiescent leukemic stem cells intrinsically resistant to imatinib. Considering that the later cancer cells lose their chemoresistance in vitro, it appears that the stromal microenvironment plays a crucial role in CML-affected cell chemoresistance. In the present review, we summarize and discuss the recent findings on signaling pathways through which stromal cells sustain CML leukemogenesis, as well as leukemic stem cell maintenance and chemoresistance.
Collapse
Affiliation(s)
- P F Seke Etet
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, 51452 Al-Qaseem, Saudi Arabia
| | | | | |
Collapse
|
21
|
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that control the expression of around 60% of the human protein-coding genes. In the past decade, deregulation of miRNAs (by expression and/or function) has been associated with the pathogenesis, progression and prognosis of different diseases, including leukemia. The number of discovered genes encoding miRNAs has risen exponentially in this period, but the numbers of miRNA-target genes discovered and validated lag far behind. Scientists have gained more in-depth knowledge of the basic mechanism of action of miRNAs, but the main challenge still remaining is the identification of direct targets of these important 'micro-players', to understand how they fine-tune so many biological processes in both healthy and diseased tissue. Many technologies have been developed in the past few years, some with more potential than others, but all with their own pros and cons. Here, we review the most common and most potent computational and experimental approaches for miRNA-target gene discovery and discuss how the hunting of targets is challenging but possible by taking the experimental limitations in consideration and choosing the correct cellular context for identifying relevant target genes.
Collapse
|
22
|
Esmailzadeh S, Jiang X. AHI-1: a novel signaling protein and potential therapeutic target in human leukemia and brain disorders. Oncotarget 2012; 2:918-34. [PMID: 22248740 PMCID: PMC3282096 DOI: 10.18632/oncotarget.405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Progress in the understanding of the molecular and cellular mechanisms of human cancer, including human leukemia and lymphomas, has been spurred by cloning of fusion genes created by chromosomal translocations or by retroviral insertional mutagenesis; a number of oncogenes and tumor suppressors involved in development of a number of malignancies have been identified in this manner. The BCR-ABL fusion gene, originating in a multipotent hematopoietic stem cell, is the molecular signature of chronic myeloid leukemia (CML). Discovery of this fusion gene has led to the development of one of the first successful targeted molecular therapies for cancer (Imatinib). It illustrates the advances that can result from an understanding of the molecular basis of disease. However, there still remain many as yet unidentified mutations that may influence the initiation or progression of human diseases. Thus, identification and characterization of the mechanism of action of genes that contribute to human diseases is an important and opportune area of current research. One promising candidate as a potential therapeutic target is Abelson helper integration site-1(Ahi-1/AHI-1) that was identified by retroviral insertional mutagenesis in murine models of leukemia/lymphomas and is highly elevated in certain human lymphoma and leukemia stem/progenitor cells. It encodes a unique protein with a SH3 domain, multiple SH3 binding sites and a WD40-repeat domain, suggesting that the normal protein has novel signaling activities. A new AHI-1-BCR-ABL-JAK2 interaction complex has recently been identified and this complex regulates transforming activities and drug resistance in CML stem/progenitor cells. Importantly, AHI-1 has recently been identified as a susceptibility gene involved in a number of brain disorders, including Joubert syndrome. Therefore, understanding molecular functions of the AHI-1 gene could lead to important and novel insights into disease processes involved in specific types of diseases. Ultimately, this knowledge will set the stage for translation into new and more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Sharmin Esmailzadeh
- Terry Fox Laboratory, British Columbia Cancer Agency and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
23
|
Abstract
Blast crisis (BC) remains the major challenge in the management of chronic myeloid leukemia (CML). It is now generally accepted that BC is the consequence of continued BCR-ABL activity leading to genetic instability, DNA damage, and impaired DNA repair. Most patients with BC carry multiple mutations, and up to 80% show additional chromosomal aberrations in a nonrandom pattern. Treatment with tyrosine kinase inhibitors has improved survival in BC modestly, but most long-term survivors are those who have been transplanted. Patients in BC should be treated with a tyrosine kinase inhibitor according to mutation profile, with or without chemotherapy, with the goal of achieving a second chronic phase and proceeding to allogeneic stem cell transplantation as quickly as possible. Although long-term remissions are rare, allogeneic stem cell transplantation provides the best chance of a cure in BC. Investigational agents are not likely to provide an alternative in the near future. In view of these limited options, prevention of BC by a rigorous and early elimination of BCR-ABL is recommended. Early response indicators should be used to select patients for alternative therapies and early transplantation. Every attempt should be made to reduce or eliminate BCR-ABL consistent with good patient care as far as possible.
Collapse
|
24
|
Li B, Zhang G, Li C, He D, Li X, Zhang C, Tang F, Deng X, Lu J, Tang Y, Li R, Chen Z, Duan C. Identification of JAK2 as a mediator of FIP1L1-PDGFRA-induced eosinophil growth and function in CEL. PLoS One 2012; 7:e34912. [PMID: 22523564 PMCID: PMC3327703 DOI: 10.1371/journal.pone.0034912] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 03/08/2012] [Indexed: 12/23/2022] Open
Abstract
The Fip1-like1 (FIP1L1)-platelet-derived growth factor receptor alpha fusion gene (F/P) arising in the pluripotent hematopoietic stem cell (HSC),causes 14% to 60% of patients with hypereosinophilia syndrome (HES). These patients, classified as having F/P (+) chronic eosinophilic leukemia (CEL), present with clonal eosinophilia and display a more aggressive disease phenotype than patients with F/P (–) HES patients. The mechanisms underlying predominant eosinophil lineage targeting and the cytotoxicity of eosinophils in this leukemia remain unclear. Given that the Janus tyrosine kinase (JAK)/signal transducers and activators of transcription (Stat) signaling pathway is key to cytokine receptor-mediated eosinophil development and activated Stat3 and Stat5 regulate the expression of genes involved in F/P malignant transformation, we investigated whether and how JAK proteins were involved in the pathogenesis of F/P-induced CEL. F/P activation of JAK2, Stat3 and Stat5, were confirmed in all the 11 F/P (+) CEL patients examined. In vitro inhibition of JAK2 in EOL-1, primary F/P(+) CEL cells (PC) and T674I F/P Imatinib resistant cells(IR) by either JAK2-specific short interfering RNA (siRNA) or the tryphostin derivative AG490(AG490), significantly reduced cellular proliferation and induced cellular apoptosis. The F/P can enhance the IL-5-induced JAK2 activation, and further results indicated that JAK2 inhibition blocked IL-5-induced cellular migration and activation of the EOL-1 and PC cells in vitro. F/P-stimulation of the JAK2 suppressed cells led to a significantly reduction in Stat3 activation, but relatively normal induction of Stat5 activation. Interestingly, JAK2 inhibition also reduced PI3K, Akt and NF-κB activity in a dose-dependent manner, and suppressed expression levels of c-Myc and Survivin. These results strongly suggest that JAK2 is activated by F/P and is required for F/P stimulation of cellular proliferation and infiltration, possibly through induction of c-Myc and Survivin expression via activation of multiple signaling pathways, including NF-κB, Stat3, and PI3K/Akt.
Collapse
Affiliation(s)
- Bin Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Division of Hematology, Institute of Molecular Hematology, the Second Xiang Ya Hospital, Central South University, Changsha City, Hunan, People's Republic of China
- Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Guangsen Zhang
- Division of Hematology, Institute of Molecular Hematology, the Second Xiang Ya Hospital, Central South University, Changsha City, Hunan, People's Republic of China
| | - Cui Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Dan He
- Medical Research Center, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xinying Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chunfang Zhang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Faqing Tang
- Clinical Laboratory, Zhuhai Hospital, Jinan University, Zhuhai, People's Republic of China
| | - Xiyun Deng
- Department of Surgery, the University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Jingchen Lu
- Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Youhong Tang
- Division of Oncology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ruijuan Li
- Division of Hematology, Institute of Molecular Hematology, the Second Xiang Ya Hospital, Central South University, Changsha City, Hunan, People's Republic of China
| | - Zhuchu Chen
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chaojun Duan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Medical Research Center, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- * E-mail:
| |
Collapse
|
25
|
Novel Combination Treatments Targeting Chronic Myeloid Leukemia Stem Cells. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:94-105. [DOI: 10.1016/j.clml.2011.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/18/2011] [Accepted: 10/27/2011] [Indexed: 11/23/2022]
|
26
|
Gnanasambandan K, Sayeski PP. A structure-function perspective of Jak2 mutations and implications for alternate drug design strategies: the road not taken. Curr Med Chem 2012; 18:4659-73. [PMID: 21864276 DOI: 10.2174/092986711797379267] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 01/13/2023]
Abstract
Jak2 is a non-receptor tyrosine kinase that is involved in the control of cellular growth and proliferation. Due to its significant role in hematopoiesis, Jak2 is a frequent target for mutations in cancer, especially myeloid leukemia, lymphoid leukemia and the myeloproliferative neoplasms (MPN). These mutations are common amongst different populations all over the world and there is a great deal of effort to develop therapeutic drugs for the affected patients. Jak2 mutations, whether they are point, deletion, or gene fusion, most commonly result in constitutive kinase activation. Here, we explore the structure-function relation of various Jak2 mutations identified in cancer and understand how they disrupt Jak2 regulation. Current Jak2 inhibitors target the highly conserved active site in the kinase domain and therefore, these inhibitors may lack specificity. Based on our knowledge regarding structure-function correlations as they pertain to regulation of Jak2 kinase activity, an alternative approach for specific Jak2 targeting could be via allosteric inhibitor design. Successful reports of allosteric inhibitors developed against other kinases provide precedent for the development of Jak2 allosteric inhibitors. Here, we suggest plausible target sites in the Jak2 structure for allosteric inhibition. Such targets include the type II inhibitor pocket and substrate binding site in the kinase domain, the kinase-pseudokinase domain interface, SH2-JH2 linker region and the FERM domain. Thus, future Jak2 inhibitors that target these sites via allosteric mechanisms may provide alternative therapeutic strategies to existing ATP competitive inhibitors.
Collapse
Affiliation(s)
- K Gnanasambandan
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, USA
| | | |
Collapse
|
27
|
Katayama H, Wang J, Treekitkarnmongkol W, Kawai H, Sasai K, Zhang H, Wang H, Adams HP, Jiang S, Chakraborty SN, Suzuki F, Arlinghaus RB, Liu J, Mobley JA, Grizzle WE, Wang H, Sen S. Aurora kinase-A inactivates DNA damage-induced apoptosis and spindle assembly checkpoint response functions of p73. Cancer Cell 2012; 21:196-211. [PMID: 22340593 PMCID: PMC3760020 DOI: 10.1016/j.ccr.2011.12.025] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 09/21/2011] [Accepted: 12/23/2011] [Indexed: 11/15/2022]
Abstract
Elevated Aurora kinase-A expression is correlated with abrogation of DNA damage-induced apoptotic response and mitotic spindle assembly checkpoint (SAC) override in human tumor cells. We report that Aurora-A phosphorylation of p73 at serine235 abrogates its transactivation function and causes cytoplasmic sequestration in a complex with the chaperon protein mortalin. Aurora-A phosphorylated p73 also facilitates inactivation of SAC through dissociation of the MAD2-CDC20 complex in cells undergoing mitosis. Cells expressing phosphor-mimetic mutant (S235D) of p73 manifest altered growth properties, resistance to cisplatin- induced apoptosis, as well as premature dissociation of the MAD2-CDC20 complex, and accelerated mitotic exit with SAC override in the presence of spindle damage. Elevated cytoplasmic p73 in Aurora-A overexpressing primary human tumors corroborates the experimental findings.
Collapse
Affiliation(s)
- Hiroshi Katayama
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jin Wang
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Warapen Treekitkarnmongkol
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hidehiko Kawai
- Department of Molecular Radiobiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kaori Sasai
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hui Zhang
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hua Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Henry P. Adams
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shoulei Jiang
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Sandip N. Chakraborty
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Fumio Suzuki
- Department of Molecular Radiobiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| | - Ralph B. Arlinghaus
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - James A. Mobley
- Department of Surgery and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - William E. Grizzle
- Department of Pathology and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Subrata Sen
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
28
|
Blockade of JAK2-mediated extrinsic survival signals restores sensitivity of CML cells to ABL inhibitors. Leukemia 2011; 26:1140-3. [PMID: 22094585 DOI: 10.1038/leu.2011.325] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder that is characterized by the presence of a fusion oncogene, BCR-ABL, which encodes a protein with constitutive tyrosine kinase activity. This activity causes excessive production of myeloid cells and their premature release into the circulation. The discovery of tyrosine kinase inhibitors marked a major advance in CML therapy, but these drugs cannot eradicate the disease because they are unable to kill the most primitive, quiescent leukemic stem cells. This review discusses current research in CML and attractive targets that have emerged with potential for eradicating the disease. Several new targets have recently been investigated as potential modulators in myeloid leukemia pathogenesis, including the multiple gene regulators miRNAs, the apparently leukemia-specific cell surface marker IL1RAP, transcription factors such as BMI1 and FOXOs, the tumor suppressors PML and PP2A, and the tyrosine kinase JAK2.
Collapse
MESH Headings
- Forkhead Box Protein O1
- Forkhead Transcription Factors/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Humans
- Interleukin-1 Receptor Accessory Protein/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- MicroRNAs/genetics
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Nuclear Proteins/genetics
- Polycomb Repressive Complex 1
- Protein Kinase Inhibitors/therapeutic use
- Proto-Oncogene Proteins/genetics
- Repressor Proteins/genetics
Collapse
Affiliation(s)
- Francesca Pellicano
- Paul O'Gorman Leukaemia Research Centre, Institute for Cancer Sciences, University of Glasgow, 21 Shelley Road, G12 0ZD Glasgow, UK.
| | | | | |
Collapse
|
30
|
Abstract
Despite the success of imatinib mesylate (IM) in the early chronic phase of chronic myeloid leukemia (CML), patients are resistant to IM and other kinase inhibitors in the later stages of CML. Our findings indicate that inhibition of Janus kinase 2 (Jak2) in Bcr–Abl+ cells overcomes IM resistance although the precise mechanism of Jak2 action is unknown. Knocking down Jak2 in Bcr–Abl+ cells reduced levels of the Bcr–Abl protein and also the phosphorylation of Tyr177 of Bcr–Abl, and Jak2 overexpression rescued these knockdown effects. Treatment of Bcr–Abl+ cells with Jak2 inhibitors for 4–6 h but not with IM also reduced Bcr–Abl protein and pTyr177 levels. In vitro kinase experiments performed with recombinant Jak2 showed that Jak2 readily phosphorylated Tyr177 of Bcr–Abl (a Jak2 consensus site, YvnV) whereas c-Abl did not. Importantly, Jak2 inhibition decreased pTyr177 Bcr–Abl in immune complexes but did not reduce levels of Bcr–Abl, suggesting that the reduction of Bcr–Abl by Jak2 inhibition is a separate event from phosphorylation of Tyr177. Jak2 inhibition by chemical inhibitors (TG101209/WP1193) and Jak2 knockdown diminished the activation of Ras, PI-3 kinase pathways and reduced levels of pTyrSTAT5. These findings suggest that Bcr–Abl stability and oncogenic signaling in CML cells are under the control of Jak2.
Collapse
|