Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Sci Adv ; 10(12): eadl4018, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38517966

ABSTRACT

In a phenotypical screen of 56 acute myeloid leukemia (AML) patient samples and using a library of 10,000 compounds, we identified a hit with increased sensitivity toward SF3B1-mutated and adverse risk AMLs. Through structure-activity relationship studies, this hit was optimized into a potent, specific, and nongenotoxic molecule called UM4118. We demonstrated that UM4118 acts as a copper ionophore that initiates a mitochondrial-based noncanonical form of cell death known as cuproptosis. CRISPR-Cas9 loss-of-function screen further revealed that iron-sulfur cluster (ISC) deficiency enhances copper-mediated cell death. Specifically, we found that loss of the mitochondrial ISC transporter ABCB7 is synthetic lethal to UM4118. ABCB7 is misspliced and down-regulated in SF3B1-mutated leukemia, creating a vulnerability to copper ionophores. Accordingly, ABCB7 overexpression partially rescued SF3B1-mutated cells to copper overload. Together, our work provides mechanistic insights that link ISC deficiency to cuproptosis, as exemplified by the high sensitivity of SF3B1-mutated AMLs. We thus propose SF3B1 mutations as a biomarker for future copper ionophore-based therapies.


Subject(s)
Copper , Leukemia, Myeloid, Acute , Humans , Copper/metabolism , RNA Splicing Factors/genetics , Mutation , Leukemia, Myeloid, Acute/genetics , Ionophores/pharmacology , Phosphoproteins/metabolism
2.
Leukemia ; 38(3): 530-537, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38102204

ABSTRACT

Monosomy 5 and deletions of the chromosome 5q (-5/del(5q)) are recurrent events in de novo adult acute myeloid leukemia (AML), reaching up to 40% of cases in secondary AML. These chromosome anomalies are associated with TP53 mutations and with very poor prognosis. Using the large Leucegene genomic and transcriptomic dataset composed of 48 -5/del(5q) patient specimens and 367 control AML, we identified DELE1 - located in the common deleted region - as the most consistently downregulated gene in these leukemias. DELE1 encodes a mitochondrial protein recently characterized as the relay of mitochondrial stress to the cytosol through a newly defined OMA1-DELE1-HRI pathway which ultimately leads to the activation of ATF4, the master transcription factor of the integrated stress response. Here, we showed that the partial loss of DELE1 expression observed in -5/del(5q) patients was sufficient to significantly reduce the sensitivity to mitochondrial stress in AML cells. Overall, our results suggest that DELE1 haploinsufficiency could represent a new driver mechanism in -5/del(5q) AML.


Subject(s)
Haploinsufficiency , Leukemia, Myeloid, Acute , Mitochondrial Proteins , Monosomy , Adult , Humans , Apoptosis/genetics , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Leukemia, Myeloid, Acute/genetics , Mitochondrial Proteins/genetics
3.
Blood Adv ; 6(16): 4793-4806, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35797243

ABSTRACT

High-mobility group AT-hook 2 (HMGA2) is a nonhistone chromatin-binding protein that is normally expressed in stem cells of various tissues and aberrantly detected in several tumor types. We recently observed that one-fourth of human acute myeloid leukemia (AML) specimens express HMGA2, which associates with a very poor prognosis. We present results indicating that HMGA2+ AMLs share a distinct transcriptional signature representing an immature phenotype. Using single-cell analyses, we showed that HMGA2 is expressed in CD34+ subsets of stem cells and early progenitors, whether normal or derived from AML specimens. Of interest, we found that one of the strongest gene expression signatures associated with HMGA2 in AML is the upregulation of G2/M checkpoint genes. Whole-genome CRISPR/Cas9 screening in HMGA2 overexpressing cells further revealed a synthetic lethal interaction with several G2/M checkpoint genes. Accordingly, small molecules that target G2/M proteins were preferentially active in vitro and in vivo on HMGA2+ AML specimens. Together, our findings suggest that HMGA2 is a key functional determinant in AML and is associated with stem cell features, G2/M status, and related drug sensitivity.


Subject(s)
Leukemia, Myeloid, Acute , Antigens, CD34 , Cell Cycle Checkpoints , Humans , Leukemia, Myeloid, Acute/pathology , Up-Regulation
4.
Blood Adv ; 6(2): 509-514, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34731885

ABSTRACT

Cholesterol homeostasis has been proposed as one mechanism contributing to chemoresistance in AML and hence, inclusion of statins in therapeutic regimens as part of clinical trials in AML has shown encouraging results. Chemical screening of primary human AML specimens by our group led to the identification of lipophilic statins as potent inhibitors of AMLs from a wide range of cytogenetic groups. Genetic screening to identify modulators of the statin response uncovered the role of protein geranylgeranylation and of RAB proteins, coordinating various aspect of vesicular trafficking, in mediating the effects of statins on AML cell viability. We further show that statins can inhibit vesicle-mediated transport in primary human specimens, and that statins sensitive samples show expression signatures reminiscent of enhanced vesicular trafficking. Overall, this study sheds light into the mechanism of action of statins in AML and identifies a novel vulnerability for cytogenetically diverse AML.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Leukemia, Myeloid, Acute , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics
5.
Blood ; 138(25): 2642-2654, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34499717

ABSTRACT

Hematopoietic stem cells (HSCs) sustain blood cell homeostasis throughout life and can regenerate all blood lineages after transplantation. Despite this clear functional definition, highly enriched isolation of human HSCs can currently only be achieved through combinatorial assessment of multiple surface antigens. Although several transgenic HSC reporter mouse strains have been described, no analogous approach to prospectively isolate human HSCs has been reported. To identify genes with the most selective expression in human HSCs, we profiled population and single-cell transcriptomes of unexpanded and ex vivo cultured cord blood-derived hematopoietic stem and progenitor cells as well as peripheral blood, adult bone marrow, and fetal liver. On the basis of these analyses, we propose the master transcription factor HLF (hepatic leukemia factor) as one of the most specific HSC marker genes. To directly track its expression in human hematopoietic cells, we developed a genomic HLF reporter strategy, capable of selectively labeling the most immature blood cells on the basis of a single engineered parameter. Most importantly, HLF-expressing cells comprise all stem cell activity in culture and in vivo during serial transplantation. Taken together, these results experimentally establish HLF as a defining gene of the human HSC state and outline a new approach to continuously mark these cells with high fidelity.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Gene Expression , Hematopoietic Stem Cells/metabolism , Transcriptome , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Single-Cell Analysis
6.
Blood ; 135(21): 1882-1886, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32315381

ABSTRACT

RUNX1 is mutated in ∼10% of adult acute myeloid leukemia (AML). Although most RUNX1 mutations in this disease are believed to be acquired, they can also be germline. Indeed, germline RUNX1 mutations result in the well-described autosomal-dominant familial platelet disorder with predisposition to hematologic malignancies (RUNX1-FPD, FPD/AML, FPDMM); ∼44% of affected individuals progress to AML or myelodysplastic syndromes. Using the Leucegene RUNX1 AML patient group, we sought to investigate the proportion of germline vs acquired RUNX1 mutations in this cohort. Our results showed that 30% of RUNX1 mutations in our AML cohort are germline. Molecular profiling revealed higher frequencies of NRAS mutations and other mutations known to activate various signaling pathways in these patients with RUNX1 germline-mutated AML. Moreover, 2 patients (mother and son) had co-occurrence of RUNX1 and CEBPA germline mutations, with variable AML disease onset at 59 and 27 years, respectively. Together, these data suggest a higher than anticipated frequency of germline RUNX1 mutations in the Leucegene cohort and further highlight the importance of testing for RUNX1 mutations in instances in which allogeneic stem cell transplantation using a related donor is envisioned.


Subject(s)
Biomarkers, Tumor/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Core Binding Factor Alpha 2 Subunit/genetics , GATA2 Transcription Factor/genetics , Germ-Line Mutation , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Aged , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis
7.
Leukemia ; 34(1): 63-74, 2020 01.
Article in English | MEDLINE | ID: mdl-31300747

ABSTRACT

Acute myeloid leukemias (AML) with mutations in the NPM1 gene (NPM1c+) represent a large AML subgroup with varying response to conventional treatment, highlighting the need to develop targeted therapeutic strategies for this disease. We screened a library of clinical drugs on a cohort of primary human AML specimens and identified the BCL2 inhibitor ABT-199 as a selective agent against NPM1c+ AML. Mutational analysis of ABT-199-sensitive and -resistant specimens identified mutations in NPM1, RAD21, and IDH1/IDH2 as predictors of ABT-199 sensitivity. Comparative transcriptome analysis further uncovered BCL2A1 as a potential mediator of ABT-199 resistance in AML. In line with our observation that RAD21 mutation confers sensitivity to ABT-199, we provide functional evidence that reducing RAD21 levels can sensitize AML cells to BCL2 inhibition. Moreover, we demonstrate that ABT-199 is able to produce selective anti-AML activity in vivo toward AML with mutations associated with compound sensitivity in PDX models. Overall, this study delineates the contribution of several genetic events to the response to ABT-199 and provides a rationale for the development of targeted therapies for NPM1c+ AML.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/genetics , Minor Histocompatibility Antigens/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Mutation , Nuclear Proteins/genetics , Nucleophosmin , Tumor Cells, Cultured
8.
Cancer Cell ; 36(1): 84-99.e8, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31287994

ABSTRACT

To identify therapeutic targets in acute myeloid leukemia (AML), we chemically interrogated 200 sequenced primary specimens. Mubritinib, a known ERBB2 inhibitor, elicited strong anti-leukemic effects in vitro and in vivo. In the context of AML, mubritinib functions through ubiquinone-dependent inhibition of electron transport chain (ETC) complex I activity. Resistance to mubritinib characterized normal CD34+ hematopoietic cells and chemotherapy-sensitive AMLs, which displayed transcriptomic hallmarks of hypoxia. Conversely, sensitivity correlated with mitochondrial function-related gene expression levels and characterized a large subset of chemotherapy-resistant AMLs with oxidative phosphorylation (OXPHOS) hyperactivity. Altogether, our work thus identifies an ETC complex I inhibitor and reveals the genetic landscape of OXPHOS dependency in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Oxazoles/pharmacology , Oxidative Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , Animals , Biomarkers , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hematopoiesis/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Mice , Models, Biological , Receptor, ErbB-2/antagonists & inhibitors
9.
Blood Adv ; 3(4): 552-563, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30782614

ABSTRACT

Patients diagnosed with acute myeloid leukemia with complex karyotype (CK AML) have an adverse prognosis using current therapies, especially when accompanied by TP53 alterations. We hereby report the RNA-sequencing analysis of the 68 CK AML samples included in the Leucegene 415 patient cohort. We confirm the frequent occurrence of TP53 alterations in this subgroup and further characterize the allele expression profile and transcript alterations of this gene. We also document that the RAS pathway (N/KRAS, NF1, PTPN11, BRAF) is frequently altered in this disease. Targeted chemical interrogation of genetically characterized primary CK AML samples identifies polo-like kinase 1 (PLK1) inhibitors as the most selective agents for this disease subgroup. TP53 status did not alter sensitivity to PLK1 inhibitors. Interestingly, CK AML specimens display a G2/M transcriptomic signature that includes higher expression levels of PLK1 and correlates with PLK1 inhibition sensitivity. Together, our results highlight vulnerability in CK AML. In line with these in vitro data, volasertib shows a strong anti-AML activity in xenotransplantation mouse models of human adverse AML. Considering that PLK1 inhibitors are currently being investigated clinically in AML and myelodysplastic syndromes, our results provide a new rationale for PLK1-directed therapy in patients with adverse cytogenetic AML.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Karyotype , M Phase Cell Cycle Checkpoints/drug effects , Male , Mice , Middle Aged , Tumor Suppressor Protein p53/genetics , Young Adult , Polo-Like Kinase 1
10.
Blood ; 130(20): 2204-2214, 2017 11 16.
Article in English | MEDLINE | ID: mdl-28855157

ABSTRACT

Neomorphic missense mutations affecting crucial lysine residues in histone H3 genes significantly contribute to a variety of solid cancers. Despite the high prevalence of H3K27M mutations in pediatric glioblastoma and their well-established impact on global histone H3 lysine 27 di- and trimethylation (H3K27me2/3), the relevance of these mutations has not been studied in acute myeloid leukemia (AML). Here, we report the first identification of H3K27M and H3K27I mutations in patients with AML. We find that these lesions are major determinants of reduced H3K27me2/3 in these patients and that they are associated with common aberrations in the RUNX1 gene. We demonstrate that H3K27I/M mutations are strong disease accelerators in a RUNX1-RUNX1T1 AML mouse model, suggesting that H3K27me2/3 has an important and selective leukemia-suppressive activity in this genetic context.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Histones/genetics , Leukemia, Myeloid, Acute/genetics , Mutation, Missense , Transformation, Genetic , Adolescent , Aged, 80 and over , Animals , DNA Methylation , Female , Humans , Lysine/metabolism , Male , Mice , Middle Aged , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Sequence Analysis, DNA
11.
Blood ; 127(24): 3054-61, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27034432

ABSTRACT

In this study, we analyzed RNA-sequencing data of 14 samples characterized by biallelic CEBPA (CEBPA(bi)) mutations included in the Leucegene collection of 415 primary acute myeloid leukemia (AML) specimens, and describe for the first time high frequency recurrent mutations in the granulocyte colony-stimulating factor receptor gene CSF3R, which signals through JAK-STAT proteins. Chemical interrogation of these primary human specimens revealed a uniform and specific sensitivity to all JAK inhibitors tested irrespective of their CSF3R mutation status, indicating a general sensitization of JAK-STAT signaling in this leukemia subset. Altogether, these results identified the co-occurrence of mutations in CSF3R and CEBPA in a well-defined AML subset, which uniformly responds to JAK inhibitors and paves the way to personalized clinical trials for this disease.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Janus Kinases/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/therapeutic use , Receptors, Colony-Stimulating Factor/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , DNA Mutational Analysis/methods , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Gene Frequency , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation , Precision Medicine , Transcriptome , Tumor Cells, Cultured , Young Adult
13.
Blood ; 127(16): 2018-27, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26834243

ABSTRACT

Acute myeloid leukemia (AML) is a genetically heterogeneous hematologic malignancy, which is initiated and driven by a rare fraction of leukemia stem cells (LSCs). Despite the difficulties of identifying a common LSC phenotype, there is increasing evidence that high expression of stem cell gene signatures is associated with poor clinical outcome. Identification of functionally distinct subpopulations in this disease is therefore crucial to dissecting the molecular machinery underlying LSC self-renewal. Here, we combined next-generation sequencing technology with in vivo assessment of LSC frequencies and identified the adhesion G protein-coupled receptor 56 (GPR56) as a novel and stable marker for human LSCs for the majority of AML samples. High GPR56 expression was significantly associated with high-risk genetic subgroups and poor outcome. Analysis of GPR56 in combination with CD34 expression revealed engraftment potential of GPR56(+)cells in both the CD34(-)and CD34(+)fractions, thus defining a novel LSC compartment independent of the CD34(+)CD38(-)LSC phenotype.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Proliferation , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Adult , Animals , Cell Separation , Cells, Cultured , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Mice , Mice, Inbred NOD , Mice, Transgenic , Neoplastic Stem Cells/physiology , Receptors, G-Protein-Coupled/physiology , Survival Analysis
14.
J Hepatol ; 51(1): 39-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19446909

ABSTRACT

BACKGROUND/AIMS: VCH-759 is a non-nucleoside inhibitor of HCV RNA-dependent polymerase with sub-micromolar IC(50) values versus genotype 1a/1b replicons. METHODS: The antiviral activity, pharmacokinetics and tolerability of VCH-759 administered as monotherapy for 10 days with a 14 day follow-up period were evaluated in 31 treatment-nai ve genotype 1 participants. Three cohorts received: 400mg thrice (t.i.d.), 800 mg twice (b.i.d.), 800 mg t.i.d or placebo. RESULTS: VCH-759 was well tolerated with the most frequent adverse event being gastrointestinal upset in both the active and placebo groups attributable, in part, to the dosing vehicle. VCH-759 was rapidly absorbed and trough plasma levels were at or above the IC(90) (non protein-adjusted) for all dosing regimens. The mean maximal decrease in HCV RNA log(10) (IU/mL) was 1.97, 2.30 and 2.46 for 400mg t.i.d., 800 mg b.i.d. and 800 mg t.i.d. doses. Viral polymerase genotypic sequencing revealed emergence of HCV variants in a majority of participants that coincided with on-treatment viral rebound. CONCLUSIONS: VCH-759 was well tolerated and achieved a> or =2 log(10) decline in HCV RNA with 800 mg b.i.d. and t.i.d doses. In a subset of participants, viral rebound was observed and associated with resistant variants. This data supports further evaluation of VCH-759 in combination with interferon-ribavirin treatment.


Subject(s)
Antiviral Agents/pharmacology , Carboxylic Acids/therapeutic use , Hepatitis C/drug therapy , Thiophenes/therapeutic use , Viral Nonstructural Proteins/antagonists & inhibitors , Adult , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Carboxylic Acids/adverse effects , Carboxylic Acids/pharmacokinetics , Double-Blind Method , Female , Hepacivirus/classification , Hepacivirus/genetics , Hepatitis C/virology , Humans , Male , Middle Aged , Phenotype , Thiophenes/adverse effects , Thiophenes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...