Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Cancers (Basel) ; 14(21)2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36358672

ABSTRACT

Tyrosine kinase inhibitors (TKI) have revolutionised the treatment of CML. However, TKI do not eliminate the leukaemia stem cells (LSC), which can re-initiate the disease. Thus, finding new therapeutic targets in CML LSC is key to finding a curative treatment. Using microarray datasets, we defined a list of 227 genes that were differentially expressed in CML LSC compared to the healthy controls but were not affected by TKI in vitro. Two of them, CD33 and PPIF, are targeted by gemtuzumab-ozogamicin and cyclosporin A, respectively. We treated CML and the control CD34+ cells with either drug with or without imatinib to investigate the therapeutic potential of the TKI-independent gene expression programme. Cyclosporine A, in combination with imatinib, reduced the number of CML CFC compared with non-CML controls, but only at supra-therapeutic concentrations. Gemtuzumab-ozogamicin showed an EC50 of 146 ng/mL, below the plasma peak concentration of 630 ng/mL observed in the AML patients and below the EC50 of 3247 ng/mL observed in the non-CML cells. Interestingly, gemtuzumab-ozogamicin seems to promote cell cycle progression in CML CD34+ cells and demonstrated activation of the RUNX1 pathway in an RNAseq experiment. This suggests that targeting the TKI-independent genes in CML LSC could be exploited for the development of new therapies in CML.

2.
Nat Commun ; 12(1): 241, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431824

ABSTRACT

Acute myeloid leukemia (AML) is a typically lethal molecularly heterogeneous disease, with few broad-spectrum therapeutic targets. Unusually, most AML retain wild-type TP53, encoding the pro-apoptotic tumor suppressor p53. MDM2 inhibitors (MDM2i), which activate wild-type p53, and BET inhibitors (BETi), targeting the BET-family co-activator BRD4, both show encouraging pre-clinical activity, but limited clinical activity as single agents. Here, we report enhanced toxicity of combined MDM2i and BETi towards AML cell lines, primary human blasts and mouse models, resulting from BETi's ability to evict an unexpected repressive form of BRD4 from p53 target genes, and hence potentiate MDM2i-induced p53 activation. These results indicate that wild-type TP53 and a transcriptional repressor function of BRD4 together represent a potential broad-spectrum synthetic therapeutic vulnerability for AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blast Crisis/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Leukemic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Mice , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/antagonists & inhibitors
4.
Leukemia ; 34(6): 1613-1625, 2020 06.
Article in English | MEDLINE | ID: mdl-31896780

ABSTRACT

The introduction of BCR-ABL tyrosine kinase inhibitors has revolutionized the treatment of chronic myeloid leukemia (CML). A major clinical aim remains the identification and elimination of low-level disease persistence, termed "minimal residual disease". The phenomenon of disease persistence suggests that despite targeted therapeutic approaches, BCR-ABL-independent mechanisms exist which sustain the survival of leukemic stem cells (LSCs). Although other markers of a primitive CML LSC population have been identified in the preclinical setting, only CD26 appears to offer clinical utility. Here we demonstrate consistent and selective expression of CD93 on a lin-CD34+CD38-CD90+ CML LSC population and show in vitro and in vivo data to suggest increased stem cell characteristics, as well as robust engraftment in patient-derived xenograft models in comparison with a CD93- CML stem/progenitor cell population, which fails to engraft. Through bulk and single-cell analyses of selected stem cell and cell survival-specific genes, we confirmed the quiescent character and demonstrate their persistence in a population of CML patient samples who demonstrate molecular relapse on TKI withdrawal. Taken together, our results identify that CD93 is consistently and selectively expressed on a lin-CD34+CD38-CD90+ CML LSC population with stem cell characteristics and may be an important indicator in determining poor TKI responders.


Subject(s)
Biomarkers, Tumor/analysis , Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Glycoproteins/metabolism , Neoplastic Stem Cells/pathology , Receptors, Complement/metabolism , Animals , Drug Resistance, Neoplasm/physiology , Heterografts , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology
6.
Leukemia ; 33(4): 981-994, 2019 04.
Article in English | MEDLINE | ID: mdl-30185934

ABSTRACT

In chronic myeloid leukemia (CML), tyrosine kinase inhibitor (TKI) treatment induces autophagy that promotes survival and TKI-resistance in leukemic stem cells (LSCs). In clinical studies hydroxychloroquine (HCQ), the only clinically approved autophagy inhibitor, does not consistently inhibit autophagy in cancer patients, so more potent autophagy inhibitors are needed. We generated a murine model of CML in which autophagic flux can be measured in bone marrow-located LSCs. In parallel, we use cell division tracing, phenotyping of primary CML cells, and a robust xenotransplantation model of human CML, to investigate the effect of Lys05, a highly potent lysosomotropic agent, and PIK-III, a selective inhibitor of VPS34, on the survival and function of LSCs. We demonstrate that long-term haematopoietic stem cells (LT-HSCs: Lin-Sca-1+c-kit+CD48-CD150+) isolated from leukemic mice have higher basal autophagy levels compared with non-leukemic LT-HSCs and more mature leukemic cells. Additionally, we present that while HCQ is ineffective, Lys05-mediated autophagy inhibition reduces LSCs quiescence and drives myeloid cell expansion. Furthermore, Lys05 and PIK-III reduced the number of primary CML LSCs and target xenografted LSCs when used in combination with TKI treatment, providing a strong rationale for clinical use of second generation autophagy inhibitors as a novel treatment for CML patients with LSC persistence.


Subject(s)
Aminoquinolines/pharmacology , Autophagy , Drug Resistance, Neoplasm/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Polyamines/pharmacology , Animals , Apoptosis , Cell Proliferation , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured
7.
Nat Med ; 24(4): 450-462, 2018 05.
Article in English | MEDLINE | ID: mdl-29505034

ABSTRACT

Leukemia stem cells (LSCs) in individuals with chronic myelogenous leukemia (CML) (hereafter referred to as CML LSCs) are responsible for initiating and maintaining clonal hematopoiesis. These cells persist in the bone marrow (BM) despite effective inhibition of BCR-ABL kinase activity by tyrosine kinase inhibitors (TKIs). Here we show that although the microRNA (miRNA) miR-126 supported the quiescence, self-renewal and engraftment capacity of CML LSCs, miR-126 levels were lower in CML LSCs than in long-term hematopoietic stem cells (LT-HSCs) from healthy individuals. Downregulation of miR-126 levels in CML LSCs was due to phosphorylation of Sprouty-related EVH1-domain-containing 1 (SPRED1) by BCR-ABL, which led to inhibition of the RAN-exportin-5-RCC1 complex that mediates miRNA maturation. Endothelial cells (ECs) in the BM supply miR-126 to CML LSCs to support quiescence and leukemia growth, as shown using mouse models of CML in which Mir126a (encoding miR-126) was conditionally knocked out in ECs and/or LSCs. Inhibition of BCR-ABL by TKI treatment caused an undesired increase in endogenous miR-126 levels, which enhanced LSC quiescence and persistence. Mir126a knockout in LSCs and/or ECs, or treatment with a miR-126 inhibitor that targets miR-126 expression in both LSCs and ECs, enhanced the in vivo anti-leukemic effects of TKI treatment and strongly diminished LSC leukemia-initiating capacity, providing a new strategy for the elimination of LSCs in individuals with CML.


Subject(s)
Bone Marrow/pathology , Cell Self Renewal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Stem Cell Niche , Animals , Down-Regulation/genetics , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Gene Silencing , Hematopoietic Stem Cells/metabolism , Humans , Mice , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/ultrastructure , Protein Kinase Inhibitors/pharmacology
8.
Blood ; 131(14): 1532-1544, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29437554

ABSTRACT

Chronic myeloid leukemia (CML) stem/progenitor cells (SPCs) express a transcriptional program characteristic of proliferation, yet can achieve and maintain quiescence. Understanding the mechanisms by which leukemic SPCs maintain quiescence will help to clarify how they persist during long-term targeted treatment. We have identified a novel BCR-ABL1 protein kinase-dependent pathway mediated by the upregulation of hsa-mir183, the downregulation of its direct target early growth response 1 (EGR1), and, as a consequence, upregulation of E2F1. We show here that inhibition of hsa-mir183 reduced proliferation and impaired colony formation of CML SPCs. Downstream of this, inhibition of E2F1 also reduced proliferation of CML SPCs, leading to p53-mediated apoptosis. In addition, we demonstrate that E2F1 plays a pivotal role in regulating CML SPC proliferation status. Thus, for the first time, we highlight the mechanism of hsa-mir183/EGR1-mediated E2F1 regulation and demonstrate this axis as a novel, critical factor for CML SPC survival, offering new insights into leukemic stem cell eradication.


Subject(s)
E2F1 Transcription Factor/biosynthesis , Early Growth Response Protein 1/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , RNA, Neoplasm/metabolism , Up-Regulation , Animals , Cell Proliferation , Cell Survival , E2F1 Transcription Factor/genetics , Early Growth Response Protein 1/genetics , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice, Knockout , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , RNA, Neoplasm/genetics , Signal Transduction
10.
J Natl Cancer Inst ; 110(5): 467-478, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29165716

ABSTRACT

Background: Imatinib and second-generation tyrosine kinase inhibitors (TKIs) nilotinib and dasatinib have statistically significantly improved the life expectancy of chronic myeloid leukemia (CML) patients; however, resistance to TKIs remains a major clinical challenge. Although ponatinib, a third-generation TKI, improves outcomes for patients with BCR-ABL-dependent mechanisms of resistance, including the T315I mutation, a proportion of patients may have or develop BCR-ABL-independent resistance and fail ponatinib treatment. By modeling ponatinib resistance and testing samples from these CML patients, it is hoped that an alternative drug target can be identified and inhibited with a novel compound. Methods: Two CML cell lines with acquired BCR-ABL-independent resistance were generated following culture in ponatinib. RNA sequencing and gene ontology (GO) enrichment were used to detect aberrant transcriptional response in ponatinib-resistant cells. A validated oncogene drug library was used to identify US Food and Drug Administration-approved drugs with activity against TKI-resistant cells. Validation was performed using bone marrow (BM)-derived cells from TKI-resistant patients (n = 4) and a human xenograft mouse model (n = 4-6 mice per group). All statistical tests were two-sided. Results: We show that ponatinib-resistant CML cells can acquire BCR-ABL-independent resistance mediated through alternative activation of mTOR. Following transcriptomic analysis and drug screening, we highlight mTOR inhibition as an alternative therapeutic approach in TKI-resistant CML cells. Additionally, we show that catalytic mTOR inhibitors induce autophagy and demonstrate that genetic or pharmacological inhibition of autophagy sensitizes ponatinib-resistant CML cells to death induced by mTOR inhibition in vitro (% number of colonies of control[SD], NVP-BEZ235 vs NVP-BEZ235+HCQ: 45.0[17.9]% vs 24.0[8.4]%, P = .002) and in vivo (median survival of NVP-BEZ235- vs NVP-BEZ235+HCQ-treated mice: 38.5 days vs 47.0 days, P = .04). Conclusion: Combined mTOR and autophagy inhibition may provide an attractive approach to target BCR-ABL-independent mechanism of resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate/administration & dosage , Imidazoles/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Molecular Targeted Therapy/methods , Pyridazines/administration & dosage , Pyrimidines/administration & dosage , Quinolines/administration & dosage , Xenograft Model Antitumor Assays
11.
Nat Med ; 23(10): 1234-1240, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28920959

ABSTRACT

Treatment of chronic myeloid leukemia (CML) with imatinib mesylate and other second- and/or third-generation c-Abl-specific tyrosine kinase inhibitors (TKIs) has substantially extended patient survival. However, TKIs primarily target differentiated cells and do not eliminate leukemic stem cells (LSCs). Therefore, targeting minimal residual disease to prevent acquired resistance and/or disease relapse requires identification of new LSC-selective target(s) that can be exploited therapeutically. Considering that malignant transformation involves cellular metabolic changes, which may in turn render the transformed cells susceptible to specific assaults in a selective manner, we searched for such vulnerabilities in CML LSCs. We performed metabolic analyses on both stem cell-enriched (CD34+ and CD34+CD38-) and differentiated (CD34-) cells derived from individuals with CML, and we compared the signature of these cells with that of their normal counterparts. Through combination of stable isotope-assisted metabolomics with functional assays, we demonstrate that primitive CML cells rely on upregulated oxidative metabolism for their survival. We also show that combination treatment with imatinib and tigecycline, an antibiotic that inhibits mitochondrial protein translation, selectively eradicates CML LSCs both in vitro and in a xenotransplantation model of human CML. Our findings provide a strong rationale for investigation of the use of TKIs in combination with tigecycline to treat patients with CML with minimal residual disease.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Minocycline/analogs & derivatives , Mitochondria/drug effects , Neoplastic Stem Cells/drug effects , Oxidative Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Blotting, Western , Cell Survival/drug effects , Chromatography, Liquid , Drug Therapy, Combination , Female , Humans , Hypoglycemic Agents/pharmacology , Imatinib Mesylate/therapeutic use , In Vitro Techniques , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred NOD , Minocycline/pharmacology , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Phenformin/pharmacology , Protein Kinase Inhibitors/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Tigecycline , Tumor Cells, Cultured , Tumor Stem Cell Assay , Up-Regulation , Xenograft Model Antitumor Assays
12.
J Hematol Oncol ; 10(1): 129, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28637510

ABSTRACT

BACKGROUND: Protein tyrosine phosphatase receptor gamma (PTPRG) is a ubiquitously expressed member of the protein tyrosine phosphatase family known to act as a tumor suppressor gene in many different neoplasms with mechanisms of inactivation including mutations and methylation of CpG islands in the promoter region. Although a critical role in human hematopoiesis and an oncosuppressor role in chronic myeloid leukemia (CML) have been reported, only one polyclonal antibody (named chPTPRG) has been described as capable of recognizing the native antigen of this phosphatase by flow cytometry. Protein biomarkers of CML have not yet found applications in the clinic, and in this study, we have analyzed a group of newly diagnosed CML patients before and after treatment. The aim of this work was to characterize and exploit a newly developed murine monoclonal antibody specific for the PTPRG extracellular domain (named TPγ B9-2) to better define PTPRG protein downregulation in CML patients. METHODS: TPγ B9-2 specifically recognizes PTPRG (both human and murine) by flow cytometry, western blotting, immunoprecipitation, and immunohistochemistry. RESULTS: Co-localization experiments performed with both anti-PTPRG antibodies identified the presence of isoforms and confirmed protein downregulation at diagnosis in the Philadelphia-positive myeloid lineage (including CD34+/CD38bright/dim cells). After effective tyrosine kinase inhibitor (TKI) treatment, its expression recovered in tandem with the return of Philadelphia-negative hematopoiesis. Of note, PTPRG mRNA levels remain unchanged in tyrosine kinase inhibitors (TKI) non-responder patients, confirming that downregulation selectively occurs in primary CML cells. CONCLUSIONS: The availability of this unique antibody permits its evaluation for clinical application including the support for diagnosis and follow-up of these disorders. Evaluation of PTPRG as a potential therapeutic target is also facilitated by the availability of a specific reagent capable to specifically detect its target in various experimental conditions.


Subject(s)
Antibodies, Monoclonal/analysis , Immunohistochemistry/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/analysis , Animals , Antibodies, Monoclonal/immunology , Blotting, Western , Down-Regulation , Gene Expression Regulation, Leukemic , Humans , Immunoprecipitation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred BALB C , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/immunology , Tumor Cells, Cultured
13.
Oncoscience ; 4(1-2): 3-4, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28484726
14.
Blood ; 129(12): 1595-1606, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28159740

ABSTRACT

Chronic myeloid leukemia (CML) is caused by the acquisition of the tyrosine kinase BCR-ABL1 in a hemopoietic stem cell, transforming it into a leukemic stem cell (LSC) that self-renews, proliferates, and differentiates to give rise to a myeloproliferative disease. Although tyrosine kinase inhibitors (TKIs) that target the kinase activity of BCR-ABL1 have transformed CML from a once-fatal disease to a manageable one for the vast majority of patients, only ∼10% of those who present in chronic phase (CP) can discontinue TKI treatment and maintain a therapy-free remission. Strong evidence now shows that CML LSCs are resistant to the effects of TKIs and persist in all patients on long-term therapy, where they may promote acquired TKI resistance, drive relapse or disease progression, and inevitably represent a bottleneck to cure. Since their discovery in patients almost 2 decades ago, CML LSCs have become a well-recognized exemplar of the cancer stem cell and have been characterized extensively, with the aim of developing new curative therapeutic approaches based on LSC eradication. This review summarizes our current understanding of many of the pathways and mechanisms that promote the survival of the CP CML LSCs and how they can be a source of new gene coding mutations that impact in the clinic. We also review recent preclinical approaches that show promise to eradicate the LSC, and future challenges on the path to cure.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myeloid, Chronic-Phase/drug therapy , Neoplastic Stem Cells/drug effects , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Chronic-Phase/pathology , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/therapeutic use
15.
Exp Hematol ; 47: 13-23, 2017 03.
Article in English | MEDLINE | ID: mdl-28017647

ABSTRACT

Advances in the design of targeted therapies for the treatment of chronic myeloid leukemia (CML) have transformed the prognosis for patients diagnosed with this disease. However, leukemic stem cell persistence, drug intolerance, drug resistance, and advanced-phase disease represent unmet clinical needs demanding the attention of CML investigators worldwide. The availability of appropriate preclinical models is essential to efficiently translate findings from the bench to the clinic. Here we review the current approaches taken to preclinical work in the CML field, including examples of commonly used in vivo models and recent successes from systems biology-based methodologies.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Animals , Animals, Genetically Modified , Cell Line, Transformed , Cell Transplantation , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Transduction, Genetic , Transgenes , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Curr Drug Targets ; 18(4): 405-413, 2017.
Article in English | MEDLINE | ID: mdl-27411711

ABSTRACT

The introduction of tyrosine kinase inhibitors in chronic myeloid leukaemia (CML) has revolutionised disease outcome. However, despite this, progression to blast phase disease is high in those that do not achieve complete cytogenetic and major molecular response on standard therapy. As well as BCR-ABL-dependent mechanisms, disease persistence has been shown to play a key role. Disease persistence suggests that, despite a targeted therapeutic approach, BCR-ABL-independent mechanisms are being exploited to sustain the survival of a small population of cells termed leukaemic stem cells (LSCs). Increasing evidence highlights the importance of self-renewal and survival pathways in this process. This review will focus on the role of stem-cell restricted self-renewal pathways, namely Hedgehog, Notch, and Bone Morphogenic Pathway (BMP). Wingless-Int/ß-Catenin (Wnt/ß-Catenin) signalling will be discussed within a further review in this series in view of its regulatory role in GSK3ß. Further to this, we will highlight the role of key transcriptional regulators, namely p53 and c- MYC, in targeting wider deregulated networks.


Subject(s)
Blast Crisis/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Blast Crisis/drug therapy , Blast Crisis/metabolism , Cell Self Renewal/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
17.
Clin Cancer Res ; 23(9): 2289-2300, 2017 May 01.
Article in English | MEDLINE | ID: mdl-27856601

ABSTRACT

Purpose: BCR-ABL kinase inhibitors are employed successfully for chronic myeloid leukemia (CML) treatment. However, resistant disease and persistence of BCR-ABL1-independent leukemia stem and progenitor cells (LSPC) remain clinical challenges. The receptor tyrosine kinase Axl can mediate survival and therapy resistance of different cancer cells. We investigated the therapeutic potential of Axl inhibition in CML.Experimental Design: We used primary cells from patients with CML and TKI-sensitive and -resistant BCR-ABL1+ CML cell lines and a novel ponatinib-resistant cell line KCL-22 PonR. We analyzed the effects of genetic and pharmacologic Axl blockade by the small-molecule Axl inhibitor BGB324 in vitro and in vivo In BCR-ABL1-unmutated cells, we also investigated BGB324 in combination with imatinib.Results: We demonstrate overexpression of Axl receptor tyrosine kinase in primary cells of patients with CML compared with healthy individuals and a further increase of Axl expression in BCR-ABL TKI-resistant patients. We show that Axl blockage decreased growth of BCR-ABL TKI-sensitive CML cells including CD34+ cells and exerts additive effects with imatinib via inhibition of Stat5 activation. BGB324 also inhibits BCR-ABL TKI-resistant cells, including T315I-mutated and ponatinib-resistant primary cells. BGB324 exerted therapeutic effects in BCR-ABL1 T315I-mutated and ponatinib-resistant preclinical mouse models. Notably, BGB324 does not inhibit BCR-ABL1 and consequently inhibits CML independent of BCR-ABL1 mutational status.Conclusions: Our data show that Axl inhibition has therapeutic potential in BCR-ABL TKI-sensitive as well as -resistant CML and support the need for clinical trials. Clin Cancer Res; 23(9); 2289-300. ©2016 AACR.


Subject(s)
Benzocycloheptenes/administration & dosage , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Triazoles/administration & dosage , Animals , Apoptosis/drug effects , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate/administration & dosage , Imidazoles/administration & dosage , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mutation , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridazines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Small Molecule Libraries/administration & dosage , Axl Receptor Tyrosine Kinase
18.
Blood ; 129(2): 199-208, 2017 01 12.
Article in English | MEDLINE | ID: mdl-27793879

ABSTRACT

Targeting the fusion oncoprotein BCR-ABL with tyrosine kinase inhibitors has significantly affected chronic myeloid leukemia (CML) treatment, transforming the life expectancy of patients; however the risk for relapse remains, due to persistence of leukemic stem cells (LSCs). Therefore it is imperative to explore the mechanisms that result in LSC survival and develop new therapeutic approaches. We now show that major histocompatibility complex (MHC)-II and its master regulator class II transactivator (CIITA) are downregulated in CML compared with non-CML stem/progenitor cells in a BCR-ABL kinase-independent manner. Interferon γ (IFN-γ) stimulation resulted in an upregulation of CIITA and MHC-II in CML stem/progenitor cells; however, the extent of IFN-γ-induced MHC-II upregulation was significantly lower than when compared with non-CML CD34+ cells. Interestingly, the expression levels of CIITA and MHC-II significantly increased when CML stem/progenitor cells were treated with the JAK1/2 inhibitor ruxolitinib (RUX). Moreover, mixed lymphocyte reactions revealed that exposure of CD34+ CML cells to IFN-γ or RUX significantly enhanced proliferation of the responder CD4+CD69+ T cells. Taken together, these data suggest that cytokine-driven JAK-mediated signals, provided by CML cells and/or the microenvironment, antagonize MHC-II expression, highlighting the potential for developing novel immunomodulatory-based therapies to enable host-mediated immunity to assist in the detection and eradication of CML stem/progenitor cells.


Subject(s)
Histocompatibility Antigens Class II/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Neoplastic Stem Cells/immunology , Tumor Escape/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Down-Regulation , Female , Flow Cytometry , Gene Expression Regulation/immunology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Lymphocyte Culture Test, Mixed , Male , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology
19.
Cell ; 167(1): 45-46, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662083

ABSTRACT

Effective differentiation therapy for acute myeloid leukemia (AML) has been restricted to a small subset of patients with one defined genetic abnormality. Using an unbiased small molecule screen, Sykes et al. now identify a mechanism of de-repression of differentiation in several models of AML driven by distinct genetic drivers.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Lifting , Cell Differentiation , Humans
20.
Cancer Discov ; 6(11): 1248-1257, 2016 11.
Article in English | MEDLINE | ID: mdl-27630125

ABSTRACT

A major obstacle to curing chronic myeloid leukemia (CML) is residual disease maintained by tyrosine kinase inhibitor (TKI)-persistent leukemic stem cells (LSC). These are BCR-ABL1 kinase independent, refractory to apoptosis, and serve as a reservoir to drive relapse or TKI resistance. We demonstrate that Polycomb Repressive Complex 2 is misregulated in chronic phase CML LSCs. This is associated with extensive reprogramming of H3K27me3 targets in LSCs, thus sensitizing them to apoptosis upon treatment with an EZH2-specific inhibitor (EZH2i). EZH2i does not impair normal hematopoietic stem cell survival. Strikingly, treatment of primary CML cells with either EZH2i or TKI alone caused significant upregulation of H3K27me3 targets, and combined treatment further potentiated these effects and resulted in significant loss of LSCs compared to TKI alone, in vitro, and in long-term bone marrow murine xenografts. Our findings point to a promising epigenetic-based therapeutic strategy to more effectively target LSCs in patients with CML receiving TKIs. SIGNIFICANCE: In CML, TKI-persistent LSCs remain an obstacle to cure, and approaches to eradicate them remain a significant unmet clinical need. We demonstrate that EZH2 and H3K27me3 reprogramming is important for LSC survival, but renders LSCs sensitive to the combined effects of EZH2i and TKI. This represents a novel approach to more effectively target LSCs in patients receiving TKI treatment. Cancer Discov; 6(11); 1248-57. ©2016 AACR.See related article by Xie et al., p. 1237This article is highlighted in the In This Issue feature, p. 1197.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cellular Reprogramming/genetics , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Epigenesis, Genetic/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...