1
|
Benjamin ESB, Vinod E, Illangeswaran RSS, Rajamani BM, Vidhyadharan RT, Bagchi A, Maity A, Mohan A, Parasuraman G, Amirtham SM, Abraham A, Velayudhan SR, Balasubramanian P. Immortalised chronic myeloid leukemia (CML) derived mesenchymal stromal cells (MSCs) line retains the immunomodulatory and chemoprotective properties of CML patient-derived MSCs. Cell Signal 2024; 116:111067. [PMID: 38281615 DOI: 10.1016/j.cellsig.2024.111067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 01/30/2024]
Abstract
Despite the success of Tyrosine kinase inhibitors (TKIs) in treating chronic myeloid leukemia (CML), leukemic stem cells (LSCs) persist, contributing to relapse and resistance. CML Mesenchymal Stromal Cells (MSCs) help in LSC maintenance and protection from TKIs. However, the limited passage and self-differentiation abilities of primary CML MSCs hinder extensive research. To overcome this, we generated and characterized an immortalised CML patient-derived MSC (iCML MSC) line and assessed its role in LSC maintenance. We also compared the immunophenotype and differentiation potential between primary CML MSCs at diagnosis, post-treatment, and with normal bone marrow MSCs. Notably, CML MSCs exhibited enhanced chondrogenic differentiation potential compared to normal MSCs. The iCML MSC line retained the trilineage differentiation potential and was genetically stable, enabling long-term investigations. Functional studies demonstrated that iCML MSCs protected CML CD34+ cells from imatinib-induced apoptosis, recapitulating the bone marrow microenvironment-mediated resistance observed in patients. iCML MSC-conditioned media enabled CML CD34+ and AML blast cells to proliferate rapidly, with no impact on healthy donor CD34+ cells. Gene expression profiling revealed dysregulated genes associated with calcium metabolism in CML CD34+ cells cocultured with iCML MSCs, providing insights into potential therapeutic targets. Further, cytokine profiling revealed that the primary CML MSC lines abundantly secreted 25 cytokines involved in immune regulation, supporting the hypothesis that CML MSCs create an immune modulatory microenvironment that promotes growth and protects against TKIs. Our study establishes the utility of iCML MSCs as a valuable model to investigate leukemic-stromal interactions and study candidate genes involved in mediating TKI resistance in CML LSCs.
Collapse
Affiliation(s)
- Esther Sathya Bama Benjamin
- Department of Haematology, Christian Medical College, Ranipet campus, India; Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, India
| | - Elizabeth Vinod
- Department of Physiology, Christain Medical College, Vellore, India; Centre for Stem Cell Research (A Unit of inStem, Bengaluru), CMC Campus, Vellore, India
| | | | | | | | - Abhirup Bagchi
- Centre for Stem Cell Research (A Unit of inStem, Bengaluru), CMC Campus, Vellore, India
| | - Arnab Maity
- Department of Haematology, Christian Medical College, Ranipet campus, India
| | - Ajith Mohan
- Department of Haematology, Christian Medical College, Ranipet campus, India
| | | | | | - Aby Abraham
- Department of Haematology, Christian Medical College, Ranipet campus, India
| | - Shaji R Velayudhan
- Department of Haematology, Christian Medical College, Ranipet campus, India; Centre for Stem Cell Research (A Unit of inStem, Bengaluru), CMC Campus, Vellore, India
| | | |
Collapse
|
2
|
Wang W, Sun Y, Liu X, Kumar SK, Jin F, Dai Y. Dual-Targeted Therapy Circumvents Non-Genetic Drug Resistance to Targeted Therapy. Front Oncol 2022; 12:859455. [PMID: 35574302 PMCID: PMC9093074 DOI: 10.3389/fonc.2022.859455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/14/2022] [Indexed: 02/05/2023] Open
Abstract
The introduction of various targeted agents into the armamentarium of cancer treatment has revolutionized the standard care of patients with cancer. However, like conventional chemotherapy, drug resistance, either preexisting (primary or intrinsic resistance) or developed following treatment (secondary or acquired resistance), remains the Achilles heel of all targeted agents with no exception, via either genetic or non-genetic mechanisms. In the latter, emerging evidence supports the notion that intracellular signaling pathways for tumor cell survival act as a mutually interdependent network via extensive cross-talks and feedback loops. Thus, dysregulations of multiple signaling pathways usually join forces to drive oncogenesis, tumor progression, invasion, metastasis, and drug resistance, thereby providing a basis for so-called "bypass" mechanisms underlying non-genetic resistance in response to targeted agents. In this context, simultaneous interruption of two or more related targets or pathways (an approach called dual-targeted therapy, DTT), via either linear or parallel inhibition, is required to deal with such a form of drug resistance to targeted agents that specifically inhibit a single oncoprotein or oncogenic pathway. Together, while most types of tumor cells are often addicted to two or more targets or pathways or can switch their dependency between them, DTT targeting either intrinsically activated or drug-induced compensatory targets/pathways would efficiently overcome drug resistance caused by non-genetic events, with a great opportunity that those resistant cells might be particularly more vulnerable. In this review article, we discuss, with our experience, diverse mechanisms for non-genetic resistance to targeted agents and the rationales to circumvent them in the treatment of cancer, emphasizing hematologic malignancies.
Collapse
Affiliation(s)
- Wei Wang
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yue Sun
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaobo Liu
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Shaji K. Kumar
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Fengyan Jin
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Agarwal P, Li H, Choi K, Hueneman K, He J, Welner RS, Starczynowski DT, Bhatia R. TNF-α-induced alterations in stromal progenitors enhance leukemic stem cell growth via CXCR2 signaling. Cell Rep 2021; 36:109386. [PMID: 34260914 PMCID: PMC8292106 DOI: 10.1016/j.celrep.2021.109386] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/30/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic myeloid leukemia (CML) is propagated by leukemia stem cells (LSCs) that are not eradicated by tyrosine kinase inhibitor (TKI) treatment and persist as a source of disease recurrence. Bone marrow (BM) mesenchymal niches play an essential role in hematopoietic stem cell (HSC) and LSC maintenance. Using a murine CML model, we examine leukemia-induced alterations in mesenchymal cell populations. We show that 6C3+ stromal progenitors expand in CML BM and exhibit increased LSC but reduced HSC supportive capacity. Tumor necrosis factor alpha (TNF-α) signaling mediates expansion and higher expression of CXCL1 in CML BM 6C3+ cells and higher expression of the CXCL1 receptor CXCR2 in LSCs. CXCL1 enhances LSC proliferation and self-renewal, whereas CXCR2 inhibition reduces LSC growth and enhances LSC targeting in combination with tyrosine kinase inhibitors (TKIs). We find that TNF-α-mediated alterations in CML BM stromal niches enhance support of LSC maintenance and growth via CXCL1-CXCR2 signaling and that CXCR2 inhibition effectively depletes CML LSCs.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Bone Marrow/pathology
- Cell Cycle/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Chemokines/metabolism
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Inflammation/genetics
- Inflammation/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Middle Aged
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptors, Interleukin-8B/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Puneet Agarwal
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA; Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hui Li
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Kwangmin Choi
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathleen Hueneman
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jianbo He
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Robert S Welner
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ravi Bhatia
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA.
| |
Collapse
|
4
|
Plasma imatinib levels and ABCB1 polymorphism influences early molecular response and failure-free survival in newly diagnosed chronic phase CML patients. Sci Rep 2020; 10:20640. [PMID: 33244077 PMCID: PMC7691501 DOI: 10.1038/s41598-020-77140-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/02/2020] [Indexed: 11/09/2022] Open
Abstract
Achieving early molecular response (EMR) has been shown to be associated with better event free survival in patients with chronic phase chronic myeloid leukemia (CP-CML) on Imatinib therapy. We prospectively evaluated the factors influencing the 2-year failure free survival (FFS) and EMR to imatinib therapy in these patients including day29 plasma Imatinib levels, genetic variants and the gene expression of target genes in imatinib transport and biotransformation. Patients with low and intermediate Sokal score had better 2-year FFS compared to those with high Sokal Score (p = 0.02). Patients carrying ABCB1-C1236T variants had high day29 plasma imatinib levels (P = 0.005), increased EMR at 3 months (P = 0.044) and a better 2 year FFS (P = 0.003) when compared to those with wild type genotype. This translates to patients with lower ABCB1 mRNA expression having a significantly higher intracellular imatinib levels (P = 0.029). Higher day29 plasma imatinib levels was found to be strongly associated with patients achieving EMR at 3 months (P = 0.022), MMR at 12 months (P = 0.041) which essentially resulted in better 2-year FFS (p = 0.05). Also, patients who achieved EMR at 3 months, 6 months and MMR at 12 months had better FFS when compared to those who did not. This study suggests the incorporation of these variables in to the imatinib dosing algorithm as predictive biomarkers of response to Imatinib therapy.
Collapse
|
5
|
Polymethine Dye-Functionalized Nanoparticles for Targeting CML Stem Cells. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:372-381. [PMID: 32913887 PMCID: PMC7452122 DOI: 10.1016/j.omto.2020.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/22/2020] [Indexed: 11/22/2022]
Abstract
In chronic myelogenous leukemia (CML), treatment with tyrosine kinase inhibitors (TKI) is unable to eradicate leukemic stem cells (LSC). Polymethine dye-functionalized nanoparticles can be internalized by specific cell types using transmembrane carrier proteins. In this study we investigated the uptake behavior of various polymethine dyes on leukemia cell lines and searched for carrier proteins that guide dye transport using RNA interference. The results show that the uptake of DY-635 is dependent on organic anion transport protein 1B3 (OATP1B3) in CML cells and immature myeloid precursor cells of CML patients. In contrast to nonspecific poly(lactide-co-glycolic acid) (PLGA) nanoparticle constructs, DY-635-functionalization of nanoparticles led to an uptake in CML cells. Investigation of these nanoparticles on bone marrow of CML patients showed a preferred uptake in LSC. The transcription of OATP1B3 is known to be induced under hypoxic conditions via the hypoxia-inducing factor 1 alpha (HIF1α), thus also in the stem cells niche. Since these cells have the potential to repopulate the bone marrow after CML treatment discontinuation, eliminating them by means of drug-loaded DY-635-functionalized PLGA nanoparticles deployed as a selective delivery system to LSC is highly relevant to the ongoing search for curative treatment options for CML patients.
Collapse
|
6
|
Spiess B, Kleiner H, Flach J, Fabarius A, Saussele S, Hofmann WK, Seifarth W. Separase activity distribution can be a marker of major molecular response and proliferation of CD34 + cells in TKI-treated chronic myeloid leukemia patients. Ann Hematol 2020; 99:991-1006. [PMID: 32253454 PMCID: PMC7196950 DOI: 10.1007/s00277-020-04007-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/18/2020] [Indexed: 11/28/2022]
Abstract
Separase, a cysteine endopeptidase, is a key player in mitotic sister chromatid separation, replication fork dynamics, and DNA repair. Aberrant expression and/or altered separase proteolytic activity are associated with aneuploidy, tumorigenesis, and disease progression. Since genomic instability and clonal evolution are hallmarks of progressing chronic myeloid leukemia (CML), we have comparatively examined separase proteolytic activity in TKI-treated chronic phase CML. Separase proteolytic activity was analyzed on single cell level in 88 clinical samples and in 14 healthy controls by a flow cytometric assay. In parallel, BCR-ABL1 gene expression and replication fork velocity were measured by qRT-PCR and DNA fiber assays, respectively. The separase activity distribution (SAD) value indicating the occurrence of MNCs with elevated separase proteolytic activity within samples was found to positively correlate with BCR-ABL1 gene expression levels and loss of MMR (relapse) throughout routine BCR-ABL1 monitoring. Analyses of CD34+ cells and MNCs fractionized by flow cytometric cell sorting according to their separase activity levels (H- and L-fractions) revealed that CD34+ cells with elevated separase activity levels (H-fractions) displayed enhanced proliferation/viability when compared with cells with regular (L-fraction) separase activity (mean 3.3-fold, p = 0.0011). BCR-ABL1 gene expression positivity prevailed in MNC H-fractions over L-fractions (42% vs. 8%, respectively). Moreover, expanding CD34+ cells of H-fractions showed decreased replication fork velocity compared with cells of L-fractions (p < 0.0001). Our data suggests an association between high separase activity, residual BCR-ABL1 gene expression, and enhanced proliferative capacity in hematopoietic cells within the leukemic niche of TKI-treated chronic phase CML.
Collapse
Affiliation(s)
- Birgit Spiess
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany. .,Hämatologie und Onkologie, III. Medizinische Klinik, Wissenschaftliches Labor, Universitätsklinikum Mannheim GmbH, Pettenkoferstraße 22, 68169, Mannheim, Germany.
| | - Helga Kleiner
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johanna Flach
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alice Fabarius
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Susanne Saussele
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang Seifarth
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
7
|
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
|
8
|
Long MJC, Hnedzko D, Kim BK, Aye Y. Breaking the Fourth Wall: Modulating Quaternary Associations for Protein Regulation and Drug Discovery. Chembiochem 2019; 20:1091-1104. [PMID: 30589188 PMCID: PMC6499692 DOI: 10.1002/cbic.201800716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Indexed: 12/13/2022]
Abstract
Protein-protein interactions (PPIs) are an effective means to orchestrate intricate biological processes required to sustain life. Approximately 650 000 PPIs underlie the human interactome; thus underscoring its complexity and the manifold signaling outputs altered in response to changes in specific PPIs. This minireview illustrates the growing arsenal of PPI assemblies and offers insights into how these varied PPI regulatory modalities are relevant to customized drug discovery, with a focus on cancer. First, known and emerging PPIs and PPI-targeted drugs of both natural and synthetic origin are categorized. Building on these discussions, the merits of PPI-guided therapeutics over traditional drug design are discussed. Finally, a compare-and-contrast section for different PPI blockers, with gain-of-function PPI interventions, such as PROTACS, is provided.
Collapse
Affiliation(s)
- Marcus J. C. Long
- 47 Pudding Gate, Bishop Burton, Beverley East Riding of Yorkshire, HU17 8QH, UK
| | - Dziyana Hnedzko
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA
| | - Bo Kyoung Kim
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
| | - Yimon Aye
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- 47 Pudding Gate, Bishop Burton, Beverley East Riding of Yorkshire, HU17 8QH, UK
| |
Collapse
|
9
|
Agarwal P, Isringhausen S, Li H, Paterson AJ, He J, Gomariz Á, Nagasawa T, Nombela-Arrieta C, Bhatia R. Mesenchymal Niche-Specific Expression of Cxcl12 Controls Quiescence of Treatment-Resistant Leukemia Stem Cells. Cell Stem Cell 2019; 24:769-784.e6. [PMID: 30905620 PMCID: PMC6499704 DOI: 10.1016/j.stem.2019.02.018] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 10/01/2018] [Accepted: 02/25/2019] [Indexed: 12/25/2022]
Abstract
Chronic myeloid leukemia (CML) originates in a hematopoietic stem cell (HSC) transformed by the breakpoint cluster region (BCR)-abelson (ABL) oncogene and is effectively treated with tyrosine kinase inhibitors (TKIs). TKIs do not eliminate disease-propagating leukemic stem cells (LSCs), suggesting a deeper understanding of niche-dependent regulation of CML LSCs is required to eradicate disease. Cxcl12 is expressed in bone marrow niches and controls HSC maintenance, and here, we show that targeted deletion of Cxcl12 from mesenchymal stromal cells (MSCs) reduces normal HSC numbers but promotes LSC expansion by increasing self-renewing cell divisions, possibly through enhanced Ezh2 activity. In contrast, endothelial cell-specific Cxcl12 deletion decreases LSC proliferation, suggesting niche-specific effects. During CML development, abnormal clusters of colocalized MSCs and LSCs form but disappear upon Cxcl12 deletion. Moreover, MSC-specific deletion of Cxcl12 increases LSC elimination by TKI treatment. These findings highlight a critical role of niche-specific effects of Cxcl12 expression in maintaining quiescence of TKI-resistant LSC populations.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Line, Tumor
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Drug Resistance, Neoplasm
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Hematopoietic Stem Cells/physiology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Mesenchymal Stem Cells/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplastic Stem Cells/physiology
- Organ Specificity
- Protein Kinase Inhibitors/therapeutic use
- Stem Cell Niche/physiology
Collapse
Affiliation(s)
- Puneet Agarwal
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Stephan Isringhausen
- Department of Hematology and Oncology, Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Hui Li
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Andrew J Paterson
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Jianbo He
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Álvaro Gomariz
- Department of Hematology and Oncology, Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology & Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - César Nombela-Arrieta
- Department of Hematology and Oncology, Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Ravi Bhatia
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA.
| |
Collapse
|
10
|
Tissue "Hypoxia" and the Maintenance of Leukemia Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:129-145. [PMID: 31338818 DOI: 10.1007/978-981-13-7342-8_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The relationship of the homing of normal hematopoietic stem cells (HSC) in the bone marrow to specific environmental conditions, referred to as the stem cell niche (SCN), has been intensively studied over the last three decades. These conditions include the action of a number of molecular and cellular players, as well as critical levels of nutrients, oxygen and glucose in particular, involved in energy production. These factors are likely to act also in leukemias, due to the strict analogy between the hierarchical structure of normal hematopoietic cell populations and that of leukemia cell populations. This led to propose that leukemic growth is fostered by cells endowed with stem cell properties, the leukemia stem cells (LSC), a concept readily extended to comprise the cancer stem cells (CSC) of solid tumors. Two alternative routes have been proposed for CSC generation, that is, the oncogenic staminalization (acquisition of self-renewal) of a normal progenitor cell (the "CSC in normal progenitor cell" model) and the oncogenic transformation of a normal (self-renewing) stem cell (the "CSC in normal stem cell" model). The latter mechanism, in the hematological context, makes LSC derive from HSC, suggesting that LSC share SCN homing with HSC. This chapter is focused on the availability of oxygen and glucose in the regulation of LSC maintenance within the SCN. In this respect, the most critical aspect in view of the outcome of therapy is the long-term maintenance of the LSC subset capable to sustain minimal residual disease and the related risk of relapse of disease.
Collapse
|
11
|
Mazumder A, Lee JY, Talhi O, Cerella C, Chateauvieux S, Gaigneaux A, Hong CR, Kang HJ, Lee Y, Kim KW, Kim DW, Shin HY, Dicato M, Bachari K, Silva AM, Orlikova-Boyer B, Diederich M. Hydroxycoumarin OT-55 kills CML cells alone or in synergy with imatinib or Synribo: Involvement of ER stress and DAMP release. Cancer Lett 2018; 438:197-218. [DOI: 10.1016/j.canlet.2018.07.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/21/2018] [Accepted: 07/14/2018] [Indexed: 01/08/2023]
|
12
|
Yang H, Chennamaneni LR, Ho MWT, Ang SH, Tan ESW, Jeyaraj DA, Yeap YS, Liu B, Ong EH, Joy JK, Wee JLK, Kwek P, Retna P, Dinie N, Nguyen TTH, Tai SJ, Manoharan V, Pendharkar V, Low CB, Chew YS, Vuddagiri S, Sangthongpitag K, Choong ML, Lee MA, Kannan S, Verma CS, Poulsen A, Lim S, Chuah C, Ong TS, Hill J, Matter A, Nacro K. Optimization of Selective Mitogen-Activated Protein Kinase Interacting Kinases 1 and 2 Inhibitors for the Treatment of Blast Crisis Leukemia. J Med Chem 2018; 61:4348-4369. [PMID: 29683667 DOI: 10.1021/acs.jmedchem.7b01714] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by bcr-abl1, a constitutively active tyrosine kinase fusion gene responsible for an abnormal proliferation of leukemic stem cells (LSCs). Inhibition of BCR-ABL1 kinase activity offers long-term relief to CML patients. However, for a proportion of them, BCR-ABL1 inhibition will become ineffective at treating the disease, and CML will progress to blast crisis (BC) CML with poor prognosis. BC-CML is often associated with excessive phosphorylated eukaryotic translation initiation factor 4E (eIF4E), which renders LSCs capable of proliferating via self-renewal, oblivious to BCR-ABL1 inhibition. In vivo, eIF4E is exclusively phosphorylated on Ser209 by MNK1/2. Consequently, a selective inhibitor of MNK1/2 should reduce the level of phosphorylated eIF4E and re-sensitize LSCs to BCR-ABL1 inhibition, thus hindering the proliferation of BC LSCs. We report herein the structure-activity relationships and pharmacokinetic properties of a selective MNK1/2 inhibitor clinical candidate, ETC-206, which in combination with dasatinib prevents BC-CML LSC self-renewal in vitro and enhances dasatinib antitumor activity in vivo.
Collapse
Affiliation(s)
- Haiyan Yang
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Lohitha Rao Chennamaneni
- Organic Chemistry, Institute of Chemical and Engineering Sciences (ICES), A*STAR , 8 Biomedical Grove, Neuros, #07-01 , 138665 Singapore
| | - Melvyn Wai Tuck Ho
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Shi Hua Ang
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Eldwin Sum Wai Tan
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | | | - Yoon Sheng Yeap
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Boping Liu
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Esther Hq Ong
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Joma Kanikadu Joy
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - John Liang Kuan Wee
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Perlyn Kwek
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Priya Retna
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Nurul Dinie
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Thuy Thi Hanh Nguyen
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Shi Jing Tai
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Vithya Manoharan
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Vishal Pendharkar
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Choon Bing Low
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Yun Shan Chew
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Susmitha Vuddagiri
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Kanda Sangthongpitag
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Meng Ling Choong
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - May Ann Lee
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | | | - Chandra S Verma
- Bioinformatics Institute (BII) , A*STAR , 30 Biopolis Street, #07-01 Matrix , 138671 Singapore.,School of Biological Sciences , Nanyang Technological University , 60 Nanyang Drive , 637551 Singapore.,Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , 117543 Singapore
| | - Anders Poulsen
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Sharon Lim
- Duke-NUS Medical School , 8 College Road , 169857 Singapore
| | - Charles Chuah
- Duke-NUS Medical School , 8 College Road , 169857 Singapore
| | - Tiong Sin Ong
- Duke-NUS Medical School , 8 College Road , 169857 Singapore.,Department of Medicine , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Jeffrey Hill
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Alex Matter
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Kassoum Nacro
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| |
Collapse
|
13
|
Costa FB, Cortez AP, de Ávila RI, de Carvalho FS, Andrade WM, da Cruz AF, Reis KB, Menegatti R, Lião LM, Romeiro LAS, Noël F, Fraga CAM, Barreiro EJ, Sanz G, Rodrigues MF, Vaz BG, Valadares MC. The novel piperazine-containing compound LQFM018: Necroptosis cell death mechanisms, dopamine D 4 receptor binding and toxicological assessment. Biomed Pharmacother 2018; 102:481-493. [PMID: 29579709 DOI: 10.1016/j.biopha.2018.02.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 01/12/2023] Open
Abstract
Piperazine is a promising scaffold for drug development due to its broad spectrum of biological activities. Based on this, the new piperazine-containing compound LQFM018 (2) [ethyl 4-((1-(4-chlorophenyl)-1H-pyrazol-4-yl)methyl)piperazine-1-carboxylate] was synthetized and some biological activities investigated. In this work, we described its ability to bind aminergic receptors, antiproliferative effects as well as the LQFM018 (2)-triggered cell death mechanisms, in K562 leukemic cells, by flow cytometric analyses. Furthermore, acute oral systemic toxicity and potential myelotoxicity assessments of LQFM018 (2) were carried out. LQFM018 (2) was originally obtained by molecular simplification from LASSBio579 (1), an analogue compound of clozapine, with 33% of global yield. Binding profile assay to aminergic receptors showed that LQFM018 (2) has affinity for the dopamine D4 receptor (Ki = 0.26 μM). Moreover, it showed cytotoxicity in K562 cells, in a concentration and time-dependent manner; IC50 values obtained were 399, 242 and 119 μM for trypan blue assay and 427, 259 and 50 μM for MTT method at 24, 48 or 72 h, respectively. This compound (427 μM) also promoted increase in LDH release and cell cycle arrest in G2/M phase. Furthermore, it triggered necrotic morphologies in K562 cells associated with intense cell membrane rupture as confirmed by Annexin V/propidium iodide double-staining. LQFM018 (2) also triggered mitochondrial disturb through loss of ΔΨm associated with increase of ROS production. No significant accumulation of cytosolic cytochrome c was verified in treated cells. Furthermore, it was verified an increase of expression of TNF-R1 and mRNA levels of CYLD with no involviment in caspase-3 and -8 activation and NF-κB in K562 cells. LQFM018 (2) showed in vitro myelotoxicity potential, but it was orally well tolerated and classified as UN GHS category 5 (LD50 > 2000-5000 mg/Kg). Thus, LQFM018 (2) seems to have a non-selective action considering hematopoietic cells. In conclusion, it is suggested LQFM018 (2) promotes cell death in K562 cells via necroptotic signaling, probably with involvement of dopamine D4 receptor. These findings open new perspectives in cancer therapy by use of necroptosis inducing agents as a strategy of reverse cancer cell chemoresistance.
Collapse
Affiliation(s)
- Fabiana Bettanin Costa
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Alane P Cortez
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Renato Ivan de Ávila
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Flávio S de Carvalho
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Wanessa M Andrade
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Andrezza F da Cruz
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Karinna B Reis
- Laboratório de Química Farmacêutica Medicinal (LQFM), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Ricardo Menegatti
- Laboratório de Química Farmacêutica Medicinal (LQFM), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Luciano M Lião
- Laboratório de Ressonância Magnética Nuclear, Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Luiz Antônio S Romeiro
- Universidade Católica de Brasília, Brasília, DF, Brazil; Universidade de Brasília, Brasília, DF, Brazil
| | - François Noël
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biológicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carlos Alberto M Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Eliezer J Barreiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Germán Sanz
- Laboratório de Cromatografia e Espectrometria de Massas (LaCEM), Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Marcella F Rodrigues
- Laboratório de Cromatografia e Espectrometria de Massas (LaCEM), Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Boniek G Vaz
- Laboratório de Cromatografia e Espectrometria de Massas (LaCEM), Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Marize Campos Valadares
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
14
|
Arrigoni E, Del Re M, Galimberti S, Restante G, Rofi E, Crucitta S, Baratè C, Petrini M, Danesi R, Di Paolo A. Concise Review: Chronic Myeloid Leukemia: Stem Cell Niche and Response to Pharmacologic Treatment. Stem Cells Transl Med 2018; 7:305-314. [PMID: 29418079 PMCID: PMC5827745 DOI: 10.1002/sctm.17-0175] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/09/2018] [Indexed: 12/27/2022] Open
Abstract
Nowadays, more than 90% of patients affected by chronic myeloid leukemia (CML) survive with a good quality of life, thanks to the clinical efficacy of tyrosine kinase inhibitors (TKIs). Nevertheless, point mutations of the ABL1 pocket occurring during treatment may reduce binding of TKIs, being responsible of about 20% of cases of resistance among CML patients. In addition, the presence of leukemic stem cells (LSCs) represents the most important event in leukemia progression related to TKI resistance. LSCs express stem cell markers, including active efflux pumps and genetic and epigenetic alterations together with deregulated cell signaling pathways involved in self-renewal, such as Wnt/β-catenin, Notch, and Hedgehog. Moreover, the interaction with the bone marrow microenvironment, also known as hematopoietic niche, may influence the phenotype of surrounding cells, which evade mechanisms controlling cell proliferation and are less sensitive or frankly resistant to TKIs. This Review focuses on the role of LSCs and stem cell niche in relation to response to pharmacological treatments. A literature search from PubMed database was performed until April 30, 2017, and it has been analyzed according to keywords such as chronic myeloid leukemia, stem cell, leukemic stem cells, hematopoietic niche, tyrosine kinase inhibitors, and drug resistance. Stem Cells Translational Medicine 2018;7:305-314.
Collapse
Affiliation(s)
- Elena Arrigoni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Sara Galimberti
- Unit of Hematology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giuliana Restante
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Eleonora Rofi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Claudia Baratè
- Unit of Hematology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Mario Petrini
- Unit of Hematology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Antonello Di Paolo
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
15
|
Chen C, Zhuang Y, Chen X, Chen X, Li D, Fan Y, Xu J, Chen Y, Wu L. Hsp90 N- and C-terminal double inhibition synergistically suppresses Bcr-Abl-positive human leukemia cells. Oncotarget 2018; 8:10025-10036. [PMID: 28036294 PMCID: PMC5354638 DOI: 10.18632/oncotarget.14324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/15/2016] [Indexed: 12/23/2022] Open
Abstract
Heat shock protein 90 (Hsp90) contains amino (N)–terminal domain, carboxyl(C)-terminal domain, and middle domains, which activate Hsp90 chaperone function cooperatively in tumor cells. One terminal occupancy might influence another terminal binding with inhibitor. The Bcr-Abl kinase is one of the Hsp90 clients implicated in the pathogenesis of chronic myeloid leukemia (CML). Present studies demonstrate that double inhibition of the N- and C-terminal termini can disrupt Hsp90 chaperone function synergistically, but not antagonistically, in Bcr-Abl-positive human leukemia cells. Furthermore, both the N-terminal inhibitor 17-AAG and the C-terminal inhibitor cisplatin (CP) have the capacity to suppress progenitor cells; however, only CP is able to inhibit leukemia stem cells (LSCs) significantly, which implies that the combinational treatment is able to suppress human leukemia in different mature states.
Collapse
Affiliation(s)
- Chun Chen
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Yingting Zhuang
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Xianling Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, China
| | - Xiaole Chen
- Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Deptartment of Biopharmaceutics, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Ding Li
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Yingjuan Fan
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Jianhua Xu
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Yuanzhong Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, China
| | - Lixian Wu
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| |
Collapse
|
16
|
Carrà G, Torti D, Crivellaro S, Panuzzo C, Taulli R, Cilloni D, Guerrasio A, Saglio G, Morotti A. The BCR-ABL/NF-κB signal transduction network: a long lasting relationship in Philadelphia positive Leukemias. Oncotarget 2018; 7:66287-66298. [PMID: 27563822 PMCID: PMC5323234 DOI: 10.18632/oncotarget.11507] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 08/10/2016] [Indexed: 12/23/2022] Open
Abstract
The Nuclear Factor-kappa B (NF-κB) family of transcription factors plays a key role in cancer pathogenesis due to the ability to promote cellular proliferation and survival, to induce resistance to chemotherapy and to mediate invasion and metastasis. NF-κB is recruited through different mechanisms involving either canonical (RelA/p50) or non-canonical pathways (RelB/p50 or RelB/p52), which transduce the signals originated from growth-factors, cytokines, oncogenic stress and DNA damage, bacterial and viral products or other stimuli. The pharmacological inhibition of the NF-κB pathway has clearly been associated with significant clinical activity in different cancers. Almost 20 years ago, NF-κB was described as an essential modulator of BCR-ABL signaling in Chronic Myeloid Leukemia and Philadelphia-positive Acute Lymphoblastic Leukemia. This review summarizes the role of NF-κB in BCR-ABL-mediated leukemogenesis and provides new insights on the long lasting BCR-ABL/NF-κB connection.
Collapse
Affiliation(s)
- Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Davide Torti
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Sabrina Crivellaro
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Riccardo Taulli
- Department of Oncology, University of Turin, Orbassano, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Angelo Guerrasio
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| |
Collapse
|
17
|
Raghavendra NM, Pingili D, Kadasi S, Mettu A, Prasad SVUM. Dual or multi-targeting inhibitors: The next generation anticancer agents. Eur J Med Chem 2017; 143:1277-1300. [PMID: 29126724 DOI: 10.1016/j.ejmech.2017.10.021] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 12/17/2022]
Abstract
Dual-targeting/Multi-targeting of oncoproteins by a single drug molecule represents an efficient, logical and alternative approach to drug combinations. An increasing interest in this approach is indicated by a steady upsurge in the number of articles on targeting dual/multi proteins published in the last 5 years. Combining different inhibitors that destiny specific single target is the standard treatment for cancer. A new generation of dual or multi-targeting drugs is emerging, where a single chemical entity can act on multiple molecular targets. Dual/Multi-targeting agents are beneficial for solving limited efficiencies, poor safety and resistant profiles of an individual target. Designing dual/multi-target inhibitors with predefined biological profiles present a challenge. The latest advances in bioinformatic tools and the availability of detailed structural information of target proteins have shown a way of discovering multi-targeting molecules. This neoteric artifice that amalgamates the molecular docking of small molecules with protein-based common pharmacophore to design multi-targeting inhibitors is gaining great importance in anticancer drug discovery. Current review focus on the discoveries of dual targeting agents in cancer therapy using rational, computational, proteomic, bioinformatics and polypharmacological approach that enables the discovery and rational design of effective and safe multi-target anticancer agents.
Collapse
Affiliation(s)
- Nulgumnalli Manjunathaiah Raghavendra
- Center for Technological Development in Health, National Institute of Science and Technology on Innovation on Neglected Diseases, Fiocruz, Rio de Janeiro, Brazil.
| | - Divya Pingili
- Sri Venkateshwara College of Pharmacy, Osmania University, Hyderabad, India; Department of Pharmacy, Jawaharlal Nehru Technological University, Kakinada, India
| | - Sundeep Kadasi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Osmania University, Hyderabad, India
| | - Akhila Mettu
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - S V U M Prasad
- Department of Pharmacy, Jawaharlal Nehru Technological University, Kakinada, India
| |
Collapse
|
18
|
Yuda J, Miyamoto T, Odawara J, Ohkawa Y, Semba Y, Hayashi M, Miyamura K, Tanimoto M, Yamamoto K, Taniwaki M, Akashi K. Persistent detection of alternatively spliced BCR-ABL variant results in a failure to achieve deep molecular response. Cancer Sci 2017; 108:2204-2212. [PMID: 28801986 PMCID: PMC5666036 DOI: 10.1111/cas.13353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 01/04/2023] Open
Abstract
Treatment with tyrosine kinase inhibitors (TKI) may sequentially induce TKI‐resistant BCR‐ABL mutants in chronic myeloid leukemia (CML). Conventional PCR monitoring of BCR‐ABL is an important indicator to determine therapeutic intervention for preventing disease progression. However, PCR cannot separately quantify amounts of BCR‐ABL and its mutants, including alternatively spliced BCR‐ABL with an insertion of 35 intronic nucleotides (BCR‐ABLIns35bp) between ABL exons 8 and 9, which introduces the premature termination and loss of kinase activity. To assess the clinical impact of BCR‐ABL mutants, we performed deep sequencing analysis of BCR‐ABL transcripts of 409 samples from 37 patients with suboptimal response to frontline imatinib who were switched to nilotinib. At baseline, TKI‐resistant mutations were documented in 3 patients, whereas BCR‐ABLIns35bp was detected in all patients. After switching to nilotinib, both BCR‐ABL and BCR‐ABLIns35bp became undetectable in 3 patients who attained complete molecular response (CMR), whereas in the remaining all 34 patients, BCR‐ABLIns35bp was persistently detected, and minimal residual disease (MRD) fluctuated at low but detectable levels. PCR monitoring underestimated molecular response in 5 patients whose BCR‐ABLIns35bp was persisted, although BCR‐ABLIns35bp does not definitively mark TKI resistance. Therefore, quantification of BCR‐ABLIns35bp is useful for evaluating “functional” MRD and determining the effectiveness of TKI with accuracy.
Collapse
Affiliation(s)
- Junichiro Yuda
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Jun Odawara
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan.,Department of Advanced Medical Initiatives, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Department of Advanced Medical Initiatives, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichiro Semba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan.,Department of Advanced Medical Initiatives, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masayasu Hayashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan.,Department of Advanced Medical Initiatives, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Miyamura
- Department of Hematology, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Japan
| | - Mitsune Tanimoto
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Kazuhito Yamamoto
- Department of Clinical Research and Department of Hematology and Cell Therapy, Aichi Cancer Center, Nagoya, Japan
| | - Masafumi Taniwaki
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| |
Collapse
|
19
|
Prinzhorn W, Stehle M, Kleiner H, Ruppenthal S, Müller MC, Hofmann WK, Fabarius A, Seifarth W. c-MYB is a transcriptional regulator of ESPL1/Separase in BCR-ABL-positive chronic myeloid leukemia. Biomark Res 2016; 4:5. [PMID: 26937281 PMCID: PMC4774018 DOI: 10.1186/s40364-016-0059-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/24/2016] [Indexed: 01/05/2023] Open
Abstract
Background Genomic instability and clonal evolution are hallmarks of progressing chronic myeloid leukemia (CML). Recently, we have shown that clonal evolution and blast crisis correlate with altered expression and activity of Separase, a cysteine endopeptidase that is a mitotic key player in chromosomal segregation and centriole duplication. Hyperactivation of Separase in human hematopoietic cells has been linked to a feedback mechanism that posttranslationally stimulates Separase proteolytic activity after imatinib therapy-induced reduction of Separase protein levels. Methods and Results In search for potential therapy-responsive transcriptional mechanisms we have investigated the role of the transcription factor c-MYB for Separase expression in CML cell lines (LAMA-84, K562, BV-173) and in clinical samples. Quantitative RT-PCR and Western blot immunostaining experiments revealed that c-MYB expression levels are decreased in an imatinib-dependent manner and positively correlate with Separase expression levels in cell lines and in clinical CML samples. RNA silencing of c-MYB expression in CML cell lines resulted in reduced Separase protein levels. Gelshift and ChIP assays confirmed that c-MYB binds to a putative c-MYB binding sequence located within the ESPL1 promoter. Conclusions Our data suggest that ESPL1/Separase is a regulatory target of c-MYB. Therefore, c-MYB, known to be required for BCR-ABL-dependent transformation of hematopoietic progenitors and leukemogenesis, may also control the Separase-dependent fidelity of mitotic chromosomal segregation and centriole duplication essential for maintenance of genomic stability.
Collapse
Affiliation(s)
- Wiltrud Prinzhorn
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Michael Stehle
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Helga Kleiner
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Sabrina Ruppenthal
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Martin C Müller
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Wolf-Karsten Hofmann
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Alice Fabarius
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| | - Wolfgang Seifarth
- III. Medizinische Klinik (Hämatologie und Onkologie), Wissenschaftliches Labor, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkofer Str. 22, 68169 Mannheim, Germany
| |
Collapse
|
20
|
Zhang N, Huang L, Tian J, Chen X, Ke F, Zheng M, Xu J, Wu L. A novel synthetic novobiocin analog, FM-Nov17, induces DNA damage in CML cells through generation of reactive oxygen species. Pharmacol Rep 2015; 68:423-8. [PMID: 26922548 DOI: 10.1016/j.pharep.2015.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/14/2015] [Accepted: 11/02/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVES To investigate the cytotoxicity of FM-Nov17 against chronic myeloid leukemia (CML) cells, we explored its underlying mechanisms mediating the induction of DNA damage and apoptotic cell death by reactive oxygen species (ROS). METHODS MTT assays were used to measure the proliferation-inhibition ratio of K562 and K562/G01 cells. Flow cytometry (FCM) was used to test the level of extracellular ROS, DNA damage, cell cycle progression and apoptosis. Western blotting was used to verify the amount of protein. RESULTS FM-Nov17 significantly inhibited the proliferation of K562 cells, with an IC50 of 58.28±0.304μM, and K562/G01 cells, with an IC50 of 62.36±0.136μM. FM-Nov17 significantly stimulated the generation of intracellular ROS, followed by the induction of DNA damage and the activation of the ATM-p53-r-H2AX pathway and checkpoint-related signals Chk1/Chk2, which led to increased numbers of cells in the S and G2/M phases of the cell cycle. Furthermore, FM-Nov17 induced apoptotic cell death by decreasing mitochondrial membrane potential and activating caspase-3 and PARP. The above effects were all prevented by the ROS scavenger N-acetylcysteine. CONCLUSIONS FM-Nov17-induces DNA damage and mitochondria-dependent cellular apoptosis in CML cells. The process is mediated by the generation of ROS.
Collapse
Affiliation(s)
- Nanwen Zhang
- Dept. of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China
| | - Lisen Huang
- Dept. of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China
| | - Jue Tian
- Dept. of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China
| | - Xianling Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, PR China
| | - Fang Ke
- Department of Pharmacochemistry, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China
| | - Ming Zheng
- Department of Anatomy, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China
| | - Jianhua Xu
- Dept. of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China
| | - Lixian Wu
- Dept. of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, PR China.
| |
Collapse
|
21
|
Yokoo M, Kubota Y, Motoyama K, Higashi T, Taniyoshi M, Tokumaru H, Nishiyama R, Tabe Y, Mochinaga S, Sato A, Sueoka-Aragane N, Sueoka E, Arima H, Irie T, Kimura S. 2-Hydroxypropyl-β-Cyclodextrin Acts as a Novel Anticancer Agent. PLoS One 2015; 10:e0141946. [PMID: 26535909 PMCID: PMC4633159 DOI: 10.1371/journal.pone.0141946] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022] Open
Abstract
2-Hydroxypropyl-β-cyclodextrin (HP-β-CyD) is a cyclic oligosaccharide that is widely used as an enabling excipient in pharmaceutical formulations, but also as a cholesterol modifier. HP-β-CyD has recently been approved for the treatment of Niemann-Pick Type C disease, a lysosomal lipid storage disorder, and is used in clinical practice. Since cholesterol accumulation and/or dysregulated cholesterol metabolism has been described in various malignancies, including leukemia, we hypothesized that HP-β-CyD itself might have anticancer effects. This study provides evidence that HP-β-CyD inhibits leukemic cell proliferation at physiologically available doses. First, we identified the potency of HP-β-CyD in vitro against various leukemic cell lines derived from acute myeloid leukemia (AML), acute lymphoblastic leukemia and chronic myeloid leukemia (CML). HP-β-CyD treatment reduced intracellular cholesterol resulting in significant leukemic cell growth inhibition through G2/M cell-cycle arrest and apoptosis. Intraperitoneal injection of HP-β-CyD significantly improved survival in leukemia mouse models. Importantly, HP-β-CyD also showed anticancer effects against CML cells expressing a T315I BCR-ABL mutation (that confers resistance to most ABL tyrosine kinase inhibitors), and hypoxia-adapted CML cells that have characteristics of leukemic stem cells. In addition, colony forming ability of human primary AML and CML cells was inhibited by HP-β-CyD. Systemic administration of HP-β-CyD to mice had no significant adverse effects. These data suggest that HP-β-CyD is a promising anticancer agent regardless of disease or cellular characteristics.
Collapse
MESH Headings
- 2-Hydroxypropyl-beta-cyclodextrin
- Animals
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/toxicity
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cholesterol/analysis
- Cholesterol/metabolism
- Colorimetry
- Drug Resistance, Neoplasm/drug effects
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- G2 Phase Cell Cycle Checkpoints/drug effects
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myeloid, Acute/drug therapy
- Lung/pathology
- M Phase Cell Cycle Checkpoints/drug effects
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Nude
- Mice, SCID
- Signal Transduction/drug effects
- Transplantation, Heterologous
- beta-Cyclodextrins/therapeutic use
- beta-Cyclodextrins/toxicity
Collapse
Affiliation(s)
- Masako Yokoo
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasushi Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
- * E-mail:
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Taniyoshi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroko Tokumaru
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rena Nishiyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Akemi Sato
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Eisaburo Sueoka
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hidetoshi Arima
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Global Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | - Tetsumi Irie
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Global Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
22
|
Rovida E, Peppicelli S, Bono S, Bianchini F, Tusa I, Cheloni G, Marzi I, Cipolleschi MG, Calorini L, Sbarba PD. The metabolically-modulated stem cell niche: a dynamic scenario regulating cancer cell phenotype and resistance to therapy. Cell Cycle 2015; 13:3169-75. [PMID: 25485495 PMCID: PMC4612663 DOI: 10.4161/15384101.2014.964107] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
This Perspective addresses the interactions of cancer stem cells (CSC) with environment which result in the modulation of CSC metabolism, and thereby of CSC phenotype and resistance to therapy. We considered first as a model disease chronic myeloid leukemia (CML), which is triggered by a well-identified oncogenetic protein (BCR/Abl) and brilliantly treated with tyrosine kinase inhibitors (TKi). However, TKi are extremely effective in inducing remission of disease, but unable, in most cases, to prevent relapse. We demonstrated that the interference with cell metabolism (oxygen/glucose shortage) enriches cells exhibiting the leukemia stem cell (LSC) phenotype and, at the same time, suppresses BCR/Abl protein expression. These LSC are therefore refractory to the TKi Imatinib-mesylate, pointing to cell metabolism as an important factor controlling the onset of TKi-resistant minimal residual disease (MRD) of CML and the related relapse. Studies of solid neoplasias brought another player into the control of MRD, low tissue pH, which often parallels cancer growth and progression. Thus, a 3-party scenario emerged for the regulation of CSC/LSC maintenance, MRD induction and disease relapse: the "hypoxic" versus the "ischemic" vs. the "acidic" environment. As these environments are unlikely constrained within rigid borders, we named this model the "metabolically-modulated stem cell niche."
Collapse
Affiliation(s)
- Elisabetta Rovida
- a Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio" ; Università degli Studi di Firenze & Istituto Toscano Tumori ; Firenze , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kobayashi M, Chen S, Gao R, Bai Y, Zhang ZY, Liu Y. Phosphatase of regenerating liver in hematopoietic stem cells and hematological malignancies. Cell Cycle 2015; 13:2827-35. [PMID: 25486470 DOI: 10.4161/15384101.2014.954448] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The phosphatases of regenerating liver (PRLs), consisting PRL1, PRL2 and PRL3, are dual-specificity protein phosphatases that have been implicated as biomarkers and therapeutic targets in several solid tumors. However, their roles in hematological malignancies are largely unknown. Recent findings demonstrate that PRL2 is important for hematopoietic stem cell self-renewal and proliferation. In addition, both PRL2 and PRL3 are highly expressed in some hematological malignancies, including acute myeloid leukemia (AML), chronic myeloid leukemia (CML), multiple myeloma (MM) and acute lymphoblastic leukemia (ALL). Moreover, PRL deficiency impairs the proliferation and survival of leukemia cells through regulating oncogenic signaling pathways. While PRLs are potential novel therapeutic targets in hematological malignancies, their exact biological function and cellular substrates remain unclear. This review will discuss how PRLs regulate hematopoietic stem cell behavior, what signaling pathways are regulated by PRLs, and how to target PRLs in hematological malignancies. An improved understanding of how PRLs function and how they are regulated may facilitate the development of PRL inhibitors that are effective in cancer treatment.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- a Department of Pediatrics, Herman B Wells Center for Pediatric Research; Department of Biochemistry and Molecular Biology , Indiana University School of Medicine ; Indianapolis , IN USA
| | | | | | | | | | | |
Collapse
|
24
|
Dagrada GP, Spagnuolo RD, Mauro V, Tamborini E, Cesana L, Gronchi A, Stacchiotti S, Pierotti MA, Negri T, Pilotti S. Solitary fibrous tumors: loss of chimeric protein expression and genomic instability mark dedifferentiation. Mod Pathol 2015; 28:1074-83. [PMID: 26022454 DOI: 10.1038/modpathol.2015.70] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/25/2015] [Indexed: 01/21/2023]
Abstract
Solitary fibrous tumors, which are characterized by their broad morphological spectrum and unpredictable behavior, are rare mesenchymal neoplasias that are currently divided into three main variants that have the NAB2-STAT6 gene fusion as their unifying molecular lesion: usual, malignant and dedifferentiated solitary fibrous tumors. The aims of this study were to validate molecular and immunohistochemical/biochemical approaches to diagnose the range of solitary fibrous tumors by focusing on the dedifferentiated variant, and to reveal the genetic events associated with dedifferentiation by integrating the findings of array comparative genomic hybridization. We studied 29 usual, malignant and dedifferentiated solitary fibrous tumors from 24 patients (including paired samples from five patients whose tumors progressed to the dedifferentiated form) by means of STAT6 immunohistochemistry and (when frozen material was available) reverse-transcriptase polymerase chain reaction and biochemistry. In addition, the array comparative genomic hybridization findings were used to profile 12 tumors from nine patients. The NAB2/STAT6 fusion was detected in all of the tumors, but immunohistochemistry and western blotting indicated that chimeric protein expression was atypical or absent in 9 out of 11 dedifferentiated tumors. The comparative genomic hybridization results revealed that the usual and malignant solitary fibrous tumors had a simple profile, whereas the genome of the dedifferentiated tumors was complex and unstable, and suggested that 13q and 17p deletions and TP53 mutations may be present in malignant lesions before the full expression of a dedifferentiated phenotype. Solitary fibrous tumor dedifferentiation is associated with the loss of chimeric oncoprotein expression, genomic instability, and cell decommitment and reprogramming. The assessment of dedifferentiated solitary fibrous tumors is based on the presence of the fusion transcripts and, in principle, negative STAT6 immunohistochemistry should not rule out a diagnosis of solitary fibrous tumor.
Collapse
Affiliation(s)
- Gian P Dagrada
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosalin D Spagnuolo
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Mauro
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Tamborini
- Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Cesana
- Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco A Pierotti
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tiziana Negri
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
25
|
Czader M, Orazi A. Acute Myeloid Leukemia and Other Types of Disease Progression in Myeloproliferative Neoplasms. Am J Clin Pathol 2015; 144:188-206. [PMID: 26185305 DOI: 10.1309/ajcpzqk40jozzzcc] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES This session of the Society for Hematopathology/European Association for Haematopathology workshop focused on disease progression in myeloproliferative neoplasms (MPNs). METHODS The session included typical and unusual presentations of chronic myelogenous leukemia (CML), BCR-ABL1 positive; Philadelphia chromosome-negative (Ph-neg) MPNs; and mastocytosis. RESULTS Cases of CML illustrated various manifestations of progression, with emphasis on criteria defining stages of the disease. Issues were discussed related to the patterns of recurrence in patients receiving tyrosine kinase inhibitor therapy, including leukemic transformation occurring in a Ph-neg clone. Ph-neg MPN cases highlighted diagnostic approaches used to establish accelerated and blast phases, including cases with significant myelofibrosis and when an adequate bone marrow aspirate smear is not available. The session also included rare cases of aggressive mastocytosis. CONCLUSIONS There was agreement that a definitive diagnosis can be challenging in the absence of documented review of prior diagnostic material and clinical history.
Collapse
|
26
|
Huang Y, Kumazoe M, Bae J, Yamada S, Takai M, Hidaka S, Yamashita S, Kim Y, Won Y, Murata M, Tsukamoto S, Tachibana H. Green tea polyphenol epigallocatechin-O-gallate induces cell death by acid sphingomyelinase activation in chronic myeloid leukemia cells. Oncol Rep 2015; 34:1162-8. [PMID: 26135316 PMCID: PMC4530928 DOI: 10.3892/or.2015.4086] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 05/29/2015] [Indexed: 12/14/2022] Open
Abstract
An epidemiological study showed that green tea consumption is associated with a reduced risk of hematopoietic malignancy. The major green tea polyphenol epigallocatechin-3-O-gallate (EGCG) is reported to have anticancer effects. Chronic myeloid leukemia (CML) is a major hematopoietic malignancy characterized by expansion of myeloid cells. In the present study, we showed EGCG-induced acid sphingomyelinase (ASM) activation and lipid raft clustering in CML cells. The ASM inhibitor desipramine significantly reduced EGCG-induced cell death. Protein kinase Cδ is a well-known kinase that plays an important role in ASM activation. We observed EGCG-induced phos-phorylation of protein kinase Cδ at Ser664. Importantly, EGCG-induced ASM activation was significantly reduced by pretreatment of CML cells with the soluble guanylate cyclase inhibitor NS2028, suggesting that EGCG induced ASM activation through the cyclic guanosine monophosphate (cGMP)-dependent pathway. Indeed, pharmacological inhibition of a cGMP-negative regulator enhanced the anti-CML effect of EGCG. These results indicate that EGCG-induced cell death via the cGMP/ASM pathway in CML cells.
Collapse
Affiliation(s)
- Yuhui Huang
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Jaehoon Bae
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Shuhei Yamada
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Mika Takai
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Shiori Hidaka
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Shuya Yamashita
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Yoonhee Kim
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Yeongseon Won
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Motoki Murata
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Shuntaro Tsukamoto
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| |
Collapse
|
27
|
Wu L, Chen X, Huang L, Tian J, Ke F, Xu J, Chen Y, Zheng M. A Novobiocin Derivative, XN4, Inhibits the Proliferation of Chronic Myeloid Leukemia Cells by Inducing Oxidative DNA Damage. PLoS One 2015; 10:e0123314. [PMID: 25928540 PMCID: PMC4415758 DOI: 10.1371/journal.pone.0123314] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Abstract
XN4 might induce DNA damage and apoptotic cell death through reactive oxygen species (ROS). The inhibition of proliferation of K562 and K562/G01 cells was measured by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide). The mRNA levels of NADPH oxidase 1-5 (Nox1-5) genes were evaluated by qRT-PCR. The levels of extracellular reactive oxygen species (ROS), DNA damage, apoptosis, and cell cycle progression were examined by flow cytometry (FCM). Protein levels were analyzed by immunoblotting. XN4 significantly inhibited the proliferation of K562 and K562/G01 cells, with IC50 values of 3.75±0.07 µM and 2.63±0.43 µM, respectively. XN4 significantly increased the levels of Nox4 and Nox5 mRNA, stimulating the generation of intracellular ROS, inducing DNA damage and activating ATM-γ-H2AX signaling, which increased the number of cells in the S and G2/M phase of the cell cycle. Subsequently, XN4 induced apoptotic cell death by activating caspase-3 and PARP. Moreover, the above effects were all reversed by the ROS scavenger N-acetylcysteine (NAC). Additionally, XN4 can induce apoptosis in progenitor/stem cells isolated from CML patients’ bone marrow. In conclusion, XN4-induced DNA damage and cell apoptosis in CML cells is mediated by the generation of ROS.
Collapse
Affiliation(s)
- Lixian Wu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
- * E-mail: (LW); (MZ)
| | - Xianling Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, P. R.China
| | - Lisen Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
| | - Jue Tian
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
| | - Fang Ke
- Department of Pharmacochemistry, School of Pharmacy, FMU, Fuzhou, P. R.China
| | - Jianhua Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
| | - Yuanzhong Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, P. R.China
| | - Ming Zheng
- Department of Anatomy, School of Basic Medicine, FMU, Fuzhou, P. R.China
- * E-mail: (LW); (MZ)
| |
Collapse
|
28
|
Schulenburg A, Blatt K, Cerny-Reiterer S, Sadovnik I, Herrmann H, Marian B, Grunt TW, Zielinski CC, Valent P. Cancer stem cells in basic science and in translational oncology: can we translate into clinical application? J Hematol Oncol 2015; 8:16. [PMID: 25886184 PMCID: PMC4345016 DOI: 10.1186/s13045-015-0113-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/14/2015] [Indexed: 02/08/2023] Open
Abstract
Since their description and identification in leukemias and solid tumors, cancer stem cells (CSC) have been the subject of intensive research in translational oncology. Indeed, recent advances have led to the identification of CSC markers, CSC targets, and the preclinical and clinical evaluation of the CSC-eradicating (curative) potential of various drugs. However, although diverse CSC markers and targets have been identified, several questions remain, such as the origin and evolution of CSC, mechanisms underlying resistance of CSC against various targeted drugs, and the biochemical basis and function of stroma cell-CSC interactions in the so-called ‘stem cell niche.’ Additional aspects that have to be taken into account when considering CSC elimination as primary treatment-goal are the genomic plasticity and extensive subclone formation of CSC. Notably, various cell fractions with different combinations of molecular aberrations and varying proliferative potential may display CSC function in a given neoplasm, and the related molecular complexity of the genome in CSC subsets is considered to contribute essentially to disease evolution and acquired drug resistance. In the current article, we discuss new developments in the field of CSC research and whether these new concepts can be exploited in clinical practice in the future.
Collapse
Affiliation(s)
- Axel Schulenburg
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, A-1090, Wien, Austria. .,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Wien, Austria.
| | - Katharina Blatt
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Sabine Cerny-Reiterer
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Irina Sadovnik
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Radiation Therapy, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria.
| | - Brigitte Marian
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Institute for Cancer Research, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Thomas W Grunt
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Clinical Oncology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Christoph C Zielinski
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Clinical Oncology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| |
Collapse
|
29
|
Morgana acts as an oncosuppressor in chronic myeloid leukemia. Blood 2015; 125:2245-53. [PMID: 25678499 DOI: 10.1182/blood-2014-05-575001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 02/09/2015] [Indexed: 01/07/2023] Open
Abstract
We recently described morgana as an essential protein able to regulate centrosome duplication and genomic stability, by inhibiting ROCK. Here we show that morgana (+/-) mice spontaneously develop a lethal myeloproliferative disease resembling human atypical chronic myeloid leukemia (aCML), preceded by ROCK hyperactivation, centrosome amplification, and cytogenetic abnormalities in the bone marrow (BM). Moreover, we found that morgana is underexpressed in the BM of patients affected by atypical CML, a disorder of poorly understood molecular basis, characterized by nonrecurrent cytogenetic abnormalities. Morgana is also underexpressed in the BM of a portion of patients affected by Philadelphia-positive CML (Ph(+) CML) caused by the BCR-ABL oncogene, and in this condition, morgana underexpression predicts a worse response to imatinib, the standard treatment for Ph(+) CML. Thus, morgana acts as an oncosuppressor with different modalities: (1) Morgana underexpression induces centrosome amplification and cytogenetic abnormalities, and (2) in Ph(+) CML, it synergizes with BCR-ABL signaling, reducing the efficacy of imatinib treatment. Importantly, ROCK inhibition in the BM of patients underexpressing morgana restored the efficacy of imatinib to induce apoptosis, suggesting that ROCK inhibitors, combined with imatinib treatment, can overcome suboptimal responses in patients in which morgana is underexpressed.
Collapse
|
30
|
Wu L, Yu J, Chen R, Liu Y, Lou L, Wu Y, Huang L, Fan Y, Gao P, Huang M, Wu Y, Chen Y, Xu J. Dual Inhibition of Bcr-Abl and Hsp90 by C086 Potently Inhibits the Proliferation of Imatinib-Resistant CML Cells. Clin Cancer Res 2014; 21:833-43. [DOI: 10.1158/1078-0432.ccr-13-3317] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
31
|
Wu LX, Wu Y, Chen RJ, Liu Y, Huang LS, Lou LG, Zheng ZH, Chen YZ, Xu JH. Curcumin derivative C817 inhibits proliferation of imatinib-resistant chronic myeloid leukemia cells with wild-type or mutant Bcr-Abl in vitro. Acta Pharmacol Sin 2014; 35:401-9. [PMID: 24487968 DOI: 10.1038/aps.2013.180] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 11/15/2013] [Indexed: 01/21/2023] Open
Abstract
AIM To find new kinase inhibitors that overcome the imatinib resistance in treatment of chronic myeloid leukemia (CML), we synthesized C817, a novel derivative of curcumin, and tested its activities against wild-type (WT) and imatinib-resistant mutant Abl kinases, as well as in imatinib-sensitive and resistant CML cells in vitro. METHODS 32D cells harboring WT or mutant Abl kinases (nucleotide binding P-loop mutants Q252H, Y253F, and imatinib contact residue mutant T315I), as well as K562/G01 cells (with whole Bcr-Abl gene amplication) were tested. Kinase activity was measured using Kinase-Glo Luminescent Kinase Assay Platform in recombinant WT and mutant (Q252H, Y253F, and T315I) Abl kinases. Cell proliferation and apoptosis were examined using MTT assay and flow cytometry, respectively. The phosphorylation levels of Bcr-Abl initiated signaling proteins were analyzed using Western blotting. Colony forming units (CFU) growth and long term culture-initiating cells (LTC-ICs) were used to test the effects of C817 on human leukemia progenitor/stem cells. RESULTS C817 potently inhibited both WT and mutant (Q252H, Y253F, and T315I) Abl kinase activities in a non-ATP competitive manner with the values of IC₅₀ at low nanomole levels. In consistent with above results, C817 suppressed the growth of both imatinib-sensitive and resistant CML cells, including wild-type K562, K562/G01, 32D-T315I, 32D-Q252H, and 32D-Y253F cells with the values of IC₅₀ at low micromole levels. C817 (0.5 or 1 μmol/L) dose-dependently inhibited the phosphorylation of Bcr-Abl and downstream proteins STAT-5 and CrkL in imatinib-resistant K562/G01 cells. Furthermore, C817 significantly suppressed CFU growth and LTC-ICs, implicating that C817 could eradiate human leukemia progenitor/stem cells. CONCLUSION C817 is a promising compound for treatment of CML patients with Bcr-Abl kinase domain mutations that confer imatinib resistance.
Collapse
|
32
|
Rovida E, Marzi I, Cipolleschi MG, Dello Sbarba P. One more stem cell niche: how the sensitivity of chronic myeloid leukemia cells to imatinib mesylate is modulated within a "hypoxic" environment. HYPOXIA 2014; 2:1-10. [PMID: 27774462 PMCID: PMC5045050 DOI: 10.2147/hp.s51812] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This is a review (by no means comprehensive) of how the stem cell niche evolved from an abstract concept to a complex system, implemented with a number of experimental data at the cellular and molecular levels, including metabolic cues, on which we focused in particular. The concept was introduced in 1978 to model bone marrow sites suited to host hematopoietic stem cells (HSCs) and favor their self-renewal, while restraining clonal expansion and commitment to differentiation. Studies of the effects of low oxygen tension on HSC maintenance in vitro led us to hypothesize niches were located within bone marrow areas where oxygen tension is lower than elsewhere. We named these areas hypoxic stem cell niches, although a low oxygen tension is to be considered physiological for the environment where HSCs are maintained. HSCs were later shown to have the option of cycling in low oxygen, which steers this cycling to the maintenance of stem cell potential. Cell subsets capable of withstanding incubation in very low oxygen were also detected within leukemia cell populations, including chronic myeloid leukemia (CML). The oncogenetic Bcr/Abl protein is completely suppressed in these subsets, whereas Bcr/Abl messenger ribonucleic acid is not, indicating that CML cells resistant to low oxygen are independent of Bcr/Abl for persistence in culture but remain genetically leukemic. Accordingly, leukemia stem cells of CML selected in low oxygen are refractory to the Bcr/Abl inhibitor imatinib mesylate. Bcr/Abl protein suppression turned out to be actually determined when glucose shortage complicated the effects of low oxygen, indicating that ischemia-like conditions are the driving force of leukemia stem cell refractoriness to imatinib mesylate. These studies pointed to “ischemic” stem cell niches as a novel scenario for the maintenance of minimal residual disease of CML. A possible functional relationship of the “ischemic” with the “hypoxic” stem cell niche is discussed.
Collapse
Affiliation(s)
- Elisabetta Rovida
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università degli Studi di Firenze, Florence, Italy; Istituto Toscano Tumori, Florence, Italy
| | - Ilaria Marzi
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università degli Studi di Firenze, Florence, Italy; Istituto Toscano Tumori, Florence, Italy
| | - Maria Grazia Cipolleschi
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università degli Studi di Firenze, Florence, Italy; Istituto Toscano Tumori, Florence, Italy
| | - Persio Dello Sbarba
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università degli Studi di Firenze, Florence, Italy; Istituto Toscano Tumori, Florence, Italy
| |
Collapse
|
33
|
Morotti A, Panuzzo C, Fava C, Saglio G. Kinase-inhibitor-insensitive cancer stem cells in chronic myeloid leukemia. Expert Opin Biol Ther 2014; 14:287-99. [PMID: 24387320 DOI: 10.1517/14712598.2014.867323] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Chronic myeloid leukemia (CML) is a myeloproliferative disorder characterized by the translocation t(9;22), coding for the chimeric protein BCR-ABL. The development of BCR-ABL tyrosine kinase inhibitors (TKIs) has dramatically revolutionized and improved CML therapy. However, TKI-based therapy faces a major challenge: the insensitivity of CML leukemic stem cells (LSCs) to TKIs. In particular, while CML progenitor cells and differentiated cells are oncogene addicted, BCR-ABL tyrosine kinase is dispensable for CML LSC survival and maintenance. Notably, in CML, additional cellular mechanisms promote LSC survival and maintenance, rendering these cells able to survive even in the presence of TKI and to eventually promote relapse. AREAS COVERED This review will focus on the mechanisms of LSC insensitivity to TKI and on the strategies to obtain synthetic lethality with combination therapies. EXPERT OPINION Several pathways have been proposed to promote LSC maintenance and described as ideal targets to induce CML LSC exhaustion in combination with TKI. Ongoing clinical trials designed to target some of these pathways will assess which molecular target is relevant for in vivo human LSC survival in a new 'stem-cell targeting' perspective.
Collapse
Affiliation(s)
- Alessandro Morotti
- University Turin, San Luigi Hospital, Division of Hematology and Internal Medicine, Department of Oncology , Orbassano - Turin , Italy
| | | | | | | |
Collapse
|
34
|
Suresh S, McCallum L, Crawford LJ, Lu WH, Sharpe DJ, Irvine AE. The matricellular protein CCN3 regulates NOTCH1 signalling in chronic myeloid leukaemia. J Pathol 2013; 231:378-87. [PMID: 24308033 PMCID: PMC4314772 DOI: 10.1002/path.4246] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Deregulated NOTCH1 has been reported in lymphoid leukaemia, although its role in chronic myeloid leukaemia (CML) is not well established. We previously reported BCR-ABL down-regulation of a novel haematopoietic regulator, CCN3, in CML; CCN3 is a non-canonical NOTCH1 ligand. This study characterizes the NOTCH1–CCN3 signalling axis in CML. In K562 cells, BCR-ABL silencing reduced full-length NOTCH1 (NOTCH1-FL) and inhibited the cleavage of NOTCH1 intracellular domain (NOTCH1-ICD), resulting in decreased expression of the NOTCH1 targets c-MYC and HES1. K562 cells stably overexpressing CCN3 (K562/CCN3) or treated with recombinant CCN3 (rCCN3) showed a significant reduction in NOTCH1 signalling (> 50% reduction in NOTCH1-ICD, p < 0.05). Gamma secretase inhibitor (GSI), which blocks NOTCH1 signalling, reduced K562/CCN3 colony formation but increased that of K562/control cells. GSI combined with either rCCN3 or imatinib reduced K562 colony formation with enhanced reduction of NOTCH1 signalling observed with combination treatments. We demonstrate an oncogenic role for NOTCH1 in CML and suggest that BCR-ABL disruption of NOTCH1–CCN3 signalling contributes to the pathogenesis of CML.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Benzamides/pharmacology
- Flow Cytometry
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Imatinib Mesylate
- K562 Cells/drug effects
- K562 Cells/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Nephroblastoma Overexpressed Protein/metabolism
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- RNA, Small Interfering
- Real-Time Polymerase Chain Reaction
- Receptor, Notch1/metabolism
- Signal Transduction/drug effects
- Transfection
Collapse
|
35
|
Morotti A, Panuzzo C, Crivellaro S, Pergolizzi B, Familiari U, Berger AH, Saglio G, Pandolfi PP. BCR-ABL disrupts PTEN nuclear-cytoplasmic shuttling through phosphorylation-dependent activation of HAUSP. Leukemia 2013; 28:1326-33. [DOI: 10.1038/leu.2013.370] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/30/2022]
|
36
|
Ramsay AG. Immune checkpoint blockade immunotherapy to activate anti-tumour T-cell immunity. Br J Haematol 2013; 162:313-25. [PMID: 23691926 DOI: 10.1111/bjh.12380] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/03/2013] [Indexed: 01/21/2023]
Abstract
The tumour microenvironment plays a dual role in cancer: it can promote tumour progression by establishing pro-tumour survival conditions but can also suppress tumour progression by killing cancer cells or inhibiting their outgrowth. These dynamically interconnected processes are under intense investigation to better understand cancer pathophysiology and allow identification of new therapeutic approaches. The ability of cancer cells to evade anti-tumour T-cell activity in the microenvironment has recently been accepted as a hallmark of cancer progression. This review will highlight the most promising therapeutic approach aimed at activating anti-tumour T-cell immunity in the cancer microenvironment: blocking inhibitory immune regulatory proteins (immune checkpoint ligands and receptors). There is emerging evidence that haematological tumours co-opt immune checkpoints as a major immune resistance mechanism. Pre-clinical findings indicate that targeted therapies and blockade of immune checkpoints could be combined to promote therapeutic synergy and long-term anti-tumour immunity to improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Alan G Ramsay
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
37
|
Tumor dormancy: long-term survival in a hostile environment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:181-200. [PMID: 23143980 DOI: 10.1007/978-1-4614-1445-2_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor dormancy occurs when cancer cells are present but the tumor does not grow. Following treatment, patients may enter complete remission in which persistent cells represent the minimal residual disease (MRD). Experimental models and clinical data suggest that the absolute quantity of this MRD is extremely low. Very few cancer cells can persist for years or decades under these hostile conditions that include continuous exposure to maintenance treatment, autologous anti-tumor immune response, and a nonpermissive microenvironment. Dormant tumor cells may survive despite these destruction factors if they adapt and develop strategies to escape from cell death. Escape may result in a state of equilibrium between MRD and the patient. Equilibrium between the immune response and tumor cells can result in long-term tumor dormancy; however, after variable lengths of time, tumor dormancy ends, and the disease progresses. Experimental models have shown that dormant tumor cells may over-express B7-H1 and B7.1 and inhibit cytotoxic T-cell mediated lysis. This resistance could be therapeutically targeted using drugs like MEK inhibitors that modulate pathways involved in B7-H1 expression. Dormant tumor cells may also develop nonspecific resistance mechanisms to cell death, such as deregulation of JAK/STAT and mTORC2/AKT pathways or autocrine and paracrine production of cytokines. This deregulation leads to cross-resistance between the immune response and cytotoxic drugs, indicating that the long-term selection that occurs in vivo during tumor dormancy may ultimately result in resistant relapse. Long-term selection of cancer cells in vitro using tyrosine kinase inhibitors selects cells that harbor the same resistance mechanisms as dormant tumor cells. Elucidating the mechanisms underlying the equilibrium that allows for the persistence of dormant tumor cells presents a novel strategy for targeted drug treatment in the context of maintenance therapy.
Collapse
|
38
|
|
39
|
Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood 2012; 120:1412-21. [PMID: 22547582 DOI: 10.1182/blood-2012-02-411678] [Citation(s) in RCA: 286] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer immune evasion is an emerging hallmark of disease progression. We have demonstrated previously that impaired actin polymerization at the T-cell immunologic synapse is a global immune dysfunction in chronic lymphocytic leukemia (CLL). Direct contact with tumor cells induces defective actin polarization at the synapse in previously healthy T cells, but the molecules mediating this dysfunction were not known. In the present study, we show via functional screening assays that CD200, CD270, CD274, and CD276 are coopted by CLL cells to induce impaired actin synapse formation in both allogeneic and autologous T cells. We also show that inhibitory ligand-induced impairment of T-cell actin dynamics is a common immunosuppressive strategy used by both hematologic (including lymphoma) and solid carcinoma cells. This immunosuppressive signaling targets T-cell Rho-GTPase activation. Of clinical relevance, the immunomodulatory drug lenalidomide prevented the induction of these defects by down-regulating tumor cell-inhibitory molecule expression. These results using human CLL as a model cancer establish a novel evasion mechanism whereby malignant cells exploit multiple inhibitory ligand signaling to down-regulate small GTPases and lytic synapse function in global T-cell populations. These findings should contribute to the design of immunotherapeutic strategies to reverse T-cell tolerance in cancer.
Collapse
|
40
|
Barth BM, I Altinoğlu E, Shanmugavelandy SS, Kaiser JM, Crespo-Gonzalez D, DiVittore NA, McGovern C, Goff TM, Keasey NR, Adair JH, Loughran TP, Claxton DF, Kester M. Targeted indocyanine-green-loaded calcium phosphosilicate nanoparticles for in vivo photodynamic therapy of leukemia. ACS NANO 2011; 5:5325-5337. [PMID: 21675727 DOI: 10.1021/nn2005766] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Leukemia is one of the most common and aggressive adult cancers, as well as the most prevalent childhood cancer. Leukemia is a cancer of the hematological system and can be divided into a diversity of unique malignancies based on the onset of the disease as well as the specific cell lineages involved. Cancer stem cells, including recently identified leukemia stem cells (LSCs), are hypothesized to be responsible for cancer development, relapse, and resistance to treatment, and new therapeutics targeting these cellular populations are urgently needed. Nontoxic and nonaggregating calcium phosphosilicate nanoparticles (CPSNPs) encapsulating the near-infrared fluoroprobe indocyanine green (ICG) were recently developed for diagnostic imaging and drug delivery as well as for photodynamic therapy (PDT) of solid tumors. Prior studies revealed that specific targeting of CPSNPs allowed for enhanced accumulation within breast cancer tumors, via CD71 targeting, or pancreatic cancer tumors, via gastrin receptor targeting. In the present study, ICG-loaded CPSNPs were evaluated as photosensitizers for PDT of leukemia. Using a novel bioconjugation approach to specifically target CD117 or CD96, surface features enhanced on leukemia stem cells, in vitro ICG-CPSNP PDT of a murine leukemia cell line and human leukemia samples were dramatically improved. Furthermore, the in vivo efficacy of PDT was dramatically enhanced in a murine leukemia model by utilizing CD117-targeted ICG-CPSNPs, resulting in 29% disease-free survival. Altogether, this study demonstrates that leukemia-targeted ICG-loaded CPSNPs offer the promise to effectively treat relapsing and multidrug-resistant leukemia and to improve the life of leukemia patients.
Collapse
Affiliation(s)
- Brian M Barth
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|