Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell Rep Med ; 5(1): 101359, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38232702

ABSTRACT

Acute myeloid leukemia is a poor-prognosis cancer commonly stratified by genetic aberrations, but these mutations are often heterogeneous and fail to consistently predict therapeutic response. Here, we combine transcriptomic, proteomic, and phosphoproteomic datasets with ex vivo drug sensitivity data to help understand the underlying pathophysiology of AML beyond mutations. We measure the proteome and phosphoproteome of 210 patients and combine them with genomic and transcriptomic measurements to identify four proteogenomic subtypes that complement existing genetic subtypes. We build a predictor to classify samples into subtypes and map them to a "landscape" that identifies specific drug response patterns. We then build a drug response prediction model to identify drugs that target distinct subtypes and validate our findings on cell lines representing various stages of quizartinib resistance. Our results show how multiomics data together with drug sensitivity data can inform therapy stratification and drug combinations in AML.


Subject(s)
Leukemia, Myeloid, Acute , Proteogenomics , Humans , Proteomics/methods , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Genomics/methods , Mutation
3.
Cancer Cell ; 39(7): 999-1014.e8, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34171263

ABSTRACT

Our study details the stepwise evolution of gilteritinib resistance in FLT3-mutated acute myeloid leukemia (AML). Early resistance is mediated by the bone marrow microenvironment, which protects residual leukemia cells. Over time, leukemia cells evolve intrinsic mechanisms of resistance, or late resistance. We mechanistically define both early and late resistance by integrating whole-exome sequencing, CRISPR-Cas9, metabolomics, proteomics, and pharmacologic approaches. Early resistant cells undergo metabolic reprogramming, grow more slowly, and are dependent upon Aurora kinase B (AURKB). Late resistant cells are characterized by expansion of pre-existing NRAS mutant subclones and continued metabolic reprogramming. Our model closely mirrors the timing and mutations of AML patients treated with gilteritinib. Pharmacological inhibition of AURKB resensitizes both early resistant cell cultures and primary leukemia cells from gilteritinib-treated AML patients. These findings support a combinatorial strategy to target early resistant AML cells with AURKB inhibitors and gilteritinib before the expansion of pre-existing resistance mutations occurs.


Subject(s)
Aniline Compounds/pharmacology , Aurora Kinase B/metabolism , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Pyrazines/pharmacology , Tumor Microenvironment , Aurora Kinase B/genetics , Biomarkers, Tumor/genetics , Exome , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Metabolome , Protein Kinase Inhibitors/pharmacology , Proteome , Tumor Cells, Cultured
4.
Oncotarget ; 11(29): 2807-2818, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32754299

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous malignancy with the most common genomic alterations in NPM1, DNMT3A, and FLT3. Midostaurin was the first FLT3 inhibitor FDA approved for AML and is standard of care for FLT3 mutant patients undergoing induction chemotherapy [1, 2]. As there is a spectrum of response, we hypothesized that biological factors beyond FLT3 could play a role in drug sensitivity and that select FLT3-ITD negative samples may also demonstrate sensitivity. Thus, we aimed to identify features that would predict response to midostaurin in FLT3 mutant and wild-type samples. We performed an ex vivo drug sensitivity screen on primary and relapsed AML samples with corresponding targeted sequencing and RNA sequencing. We observed a correlation between FLT3-ITD mutations and midostaurin sensitivity as expected and observed KRAS and TP53 mutations correlating with midostaurin resistance in FLT3-ITD negative samples. Further, we identified genes differentially expressed in sensitive vs. resistant samples independent of FLT3-ITD status. Within FLT3-ITD mutant samples, over-expression of RGL4, oncogene and regulator of the Ras-Raf-MEK-ERK cascade, distinguished resistant from sensitive samples. Overall, this study highlights the complexity underlying midostaurin response. And, our results suggest that therapies that target both FLT3 and MAPK/ERK signaling may help circumvent some cases of resistance.

5.
Blood ; 135(24): 2159-2170, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32315394

ABSTRACT

Much of what is known about the neurotrophic receptor tyrosine kinase (NTRK) genes in cancer was revealed through identification and characterization of activating Trk fusions across many tumor types. A resurgence of interest in these receptors has emerged owing to the realization that they are promising therapeutic targets. The remarkable efficacy of pan-Trk inhibitors larotrectinib and entrectinib in clinical trials led to their accelerated, tissue-agnostic US Food and Drug Administration (FDA) approval for adult and pediatric patients with Trk-driven solid tumors. Despite our enhanced understanding of Trk biology in solid tumors, the importance of Trk signaling in hematological malignancies is underexplored and warrants further investigation. Herein, we describe mutations in NTRK2 and NTRK3 identified via deep sequencing of 185 patients with hematological malignancies. Ten patients contained a point mutation in NTRK2 or NTRK3; among these, we identified 9 unique point mutations. Of these 9 mutations, 4 were oncogenic (NTRK2A203T, NTRK2R458G, NTRK3E176D, and NTRK3L449F), determined via cytokine-independent cellular assays. Our data demonstrate that these mutations have transformative potential to promote downstream survival signaling and leukemogenesis. Specifically, the 3 mutations located within extracellular (ie, NTRK2A203T and NTRK3E176D) and transmembrane (ie, NTRK3L449F) domains increased receptor dimerization and cell-surface abundance. The fourth mutation, NTRK2R458G, residing in the juxtamembrane domain, activates TrkB via noncanonical mechanisms that may involve altered interactions between the mutant receptor and lipids in the surrounding environment. Importantly, these 4 activating mutations can be clinically targeted using entrectinib. Our findings contribute to ongoing efforts to define the mutational landscape driving hematological malignancies and underscore the utility of FDA-approved Trk inhibitors for patients with aggressive Trk-driven leukemias.


Subject(s)
Hematologic Neoplasms/genetics , Membrane Glycoproteins/genetics , Point Mutation , Receptor, trkB/genetics , Receptor, trkC/genetics , Animals , Base Sequence , Benzamides/therapeutic use , Cell Line , Drug Resistance, Neoplasm/genetics , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Humans , Indazoles/therapeutic use , Lipid Metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oncogenes , Protein Kinase Inhibitors/therapeutic use , Protein Multimerization/genetics , RNA, Small Interfering/genetics , Receptor, trkB/chemistry , Receptor, trkB/metabolism , Receptor, trkC/chemistry , Receptor, trkC/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Nat Commun ; 10(1): 5455, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31784538

ABSTRACT

Acute Myeloid Leukemia (AML) develops due to the acquisition of mutations from multiple functional classes. Here, we demonstrate that activating mutations in the granulocyte colony stimulating factor receptor (CSF3R), cooperate with loss of function mutations in the transcription factor CEBPA to promote acute leukemia development. The interaction between these distinct classes of mutations occurs at the level of myeloid lineage enhancers where mutant CEBPA prevents activation of a subset of differentiation associated enhancers. To confirm this enhancer-dependent mechanism, we demonstrate that CEBPA mutations must occur as the initial event in AML initiation. This improved mechanistic understanding will facilitate therapeutic development targeting the intersection of oncogene cooperativity.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Receptors, Colony-Stimulating Factor/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Humans , K562 Cells , Loss of Function Mutation , Mice , Mutation
7.
Blood ; 133(6): 588-599, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30425048

ABSTRACT

To identify new therapeutic targets in acute myeloid leukemia (AML), we performed small-molecule and small-interfering RNA (siRNA) screens of primary AML patient samples. In 23% of samples, we found sensitivity to inhibition of colony-stimulating factor 1 (CSF1) receptor (CSF1R), a receptor tyrosine kinase responsible for survival, proliferation, and differentiation of myeloid-lineage cells. Sensitivity to CSF1R inhibitor GW-2580 was found preferentially in de novo and favorable-risk patients, and resistance to GW-2580 was associated with reduced overall survival. Using flow cytometry, we discovered that CSF1R is not expressed on the majority of leukemic blasts but instead on a subpopulation of supportive cells. Comparison of CSF1R-expressing cells in AML vs healthy donors by mass cytometry revealed expression of unique cell-surface markers. The quantity of CSF1R-expressing cells correlated with GW-2580 sensitivity. Exposure of primary AML patient samples to a panel of recombinant cytokines revealed that CSF1R inhibitor sensitivity correlated with a growth response to CSF1R ligand, CSF1, and other cytokines, including hepatocyte growth factor (HGF). The addition of CSF1 increased the secretion of HGF and other cytokines in conditioned media from AML patient samples, whereas adding GW-2580 reduced their secretion. In untreated cells, HGF levels correlated significantly with GW-2580 sensitivity. Finally, recombinant HGF and HS-5-conditioned media rescued cell viability after GW-2580 treatment in AML patient samples. Our results suggest that CSF1R-expressing cells support the bulk leukemia population through the secretion of HGF and other cytokines. This study identifies CSF1R as a novel therapeutic target of AML and provides a mechanism of paracrine cytokine/growth factor signaling in this disease.


Subject(s)
Anisoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Paracrine Communication/drug effects , Pyrimidines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tumor Microenvironment/drug effects , Antineoplastic Agents/pharmacology , Cell Differentiation , Cell Survival , Culture Media, Conditioned/pharmacology , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Prognosis , Survival Rate , Tumor Cells, Cultured
8.
J Biol Chem ; 293(19): 7387-7396, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29572350

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF or CSF3) and its receptor CSF3R regulate granulopoiesis, neutrophil function, and hematopoietic stem cell mobilization. Recent studies have uncovered an oncogenic role of mutations in the CSF3R gene in many hematologic malignancies. To find additional CSF3R mutations that give rise to cell transformation, we performed a cellular transformation assay in which murine interleukin 3 (IL-3)-dependent Ba/F3 cells were transduced with WT CSF3R plasmid and screened for spontaneous growth in the absence of IL-3. Any outgrowth clones were sequenced to identify CSF3R mutations with transformation capacity. We identified several novel mutations and determined that they transform cells via four distinct mechanisms: 1) cysteine- and disulfide bond-mediated dimerization (S581C); 2) polar, noncharged amino acid substitution at the transmembrane helix dimer interface at residue Thr-640; 3) increased internalization by a Glu-524 substitution that mimics a low G-CSF dose; and 4) hydrophobic amino acid substitutions in the membrane-proximal residues Thr-612, Thr-615, and Thr-618. Furthermore, the change in signaling activation was related to an altered CSF3R localization. We also found that CSF3R-induced STAT3 and ERK activations require CSF3R internalization, whereas STAT5 activation occurred at the cell surface. Cumulatively, we have expanded the regions of the CSF3R extracellular and transmembrane domains in which missense mutations exhibit leukemogenic capacity and have further elucidated the mechanistic underpinnings that underlie altered CSF3R expression, dimerization, and signaling activation.


Subject(s)
Gain of Function Mutation , Receptors, Colony-Stimulating Factor/genetics , Receptors, Colony-Stimulating Factor/metabolism , Amino Acid Substitution , Animals , Cell Line , Cell Transformation, Neoplastic , Cysteine/metabolism , Dimerization , Disulfides/metabolism , Endocytosis , Humans , Hydrophobic and Hydrophilic Interactions , Leukemia/genetics , Mice , Mutagenesis , Receptors, Colony-Stimulating Factor/chemistry , STAT3 Transcription Factor/metabolism , Signal Transduction , Subcellular Fractions/metabolism , Threonine/chemistry , Threonine/metabolism
10.
Cancer Res ; 77(16): 4258-4267, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28652245

ABSTRACT

Exclusive of membrane-proximal mutations seen commonly in chronic neutrophilic leukemia (e.g., T618I), functionally defective mutations in the extracellular domain of the G-CSF receptor (CSF3R) have been reported only in severe congenital and idiopathic neutropenia patients. Here, we describe the first activating mutation in the fibronectin-like type III domain of the extracellular region of CSF3R (W341C) in a leukemia patient. This mutation transformed cells via cysteine-mediated intermolecular disulfide bonds, leading to receptor dimerization. Interestingly, a CSF3R cytoplasmic truncation mutation (W791X) found on the same allele as the extracellular mutation and the expansion of the compound mutation was associated with increased leukocytosis and disease progression of the patient. Notably, the primary patient sample and cells transformed by W341C and W341C/W791X exhibited sensitivity to JAK inhibitors. We further showed that disruption of original cysteine pairs in the CSF3R extracellular domain resulted in either gain- or loss-of-function changes, part of which was attributable to cysteine-mediated dimer formation. This, therefore, represents the first characterization of unpaired cysteines that mediate both gain- and loss-of-function phenotypes. Overall, our results show the structural and functional importance of conserved extracellular cysteine pairs in CSF3R and suggest the necessity for broader screening of CSF3R extracellular domain in leukemia patients. Cancer Res; 77(16); 4258-67. ©2017 AACR.


Subject(s)
Cysteine/genetics , Leukemia, Myeloid, Acute/genetics , Mutation, Missense , Receptors, Colony-Stimulating Factor/genetics , Aged , Animals , Exome , Female , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Phenotype
11.
Oncotarget ; 8(8): 12596-12606, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28208123

ABSTRACT

The identification and functional validation of potentially oncogenic mutations in leukemia is an essential step toward a future of personalized targeted therapy. To assess the oncogenic capacity of individual mutations, reliable and scalable in vitro experimental approaches are required. Since 1988, researchers have used the IL-3 dependent Ba/F3 transformation assay to validate the oncogenic potential of mutations to drive factor-independent growth. Here we report a previously unrecognized phenomenon whereby Ba/F3 cells, engineered to express weakly transforming mutations, present with additional acquired mutations in the expressed transgene following factor withdrawal. Using four mutations with known transformative capacity in three cytokine receptors (CSF2RB, CSF3R and IL7R), we demonstrate that the mutated receptors are highly susceptible to acquiring additional mutations. These acquired mutations of unknown functional significance are selected by factor withdrawal but appear to exist prior to the removal of growth factor. This anomaly has the potential to confound efforts to both validate and characterize oncogenic mutations in leukemia, particularly when it is not standard practice to sequence validate cDNAs from transformed Ba/F3 lines. We present specific recommendations to detect and mitigate this phenomenon in future research using Ba/F3 transformation assays, along with methods to make the Ba/F3 assay more quantitative.


Subject(s)
Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Genetic Techniques , Leukemia/genetics , Transgenes/genetics , Animals , Mice , Mutation
12.
Pediatr Blood Cancer ; 61(4): 740-2, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24115584

ABSTRACT

Fanconi anemia (FA) patients suffer from progressive bone marrow failure and often develop cancers. Previous studies showed that antioxidants tempol and resveratrol (RV) delayed tumor onset and reduced hematologic defects in FA murine models, respectively. Here we tested whether antioxidants N-acetylcysteine (NAC) or RV could delay cancer in tumor prone Fancd2(-/-) /Trp53(+/-) mice. Unlike tempol, neither compound had any significant chemopreventive effect in this model. We conclude that not all anti-oxidants are chemopreventive in FA. In addition, when given to Fancd2(-/-) mice, NAC helped maintain Fancd2(-/-) KSL cells in quiescence while tempol did not. The mechanisms behind the different actions of these antioxidants await further investigation.


Subject(s)
Acetylcysteine/therapeutic use , Antioxidants/therapeutic use , Fanconi Anemia Complementation Group D2 Protein/physiology , Fanconi Anemia/prevention & control , Free Radical Scavengers/therapeutic use , Stilbenes/therapeutic use , Tumor Suppressor Protein p53/physiology , Animals , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Flow Cytometry , Mice , Mice, Knockout , Resveratrol
13.
Stem Cell Res ; 11(2): 687-92, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23721813

ABSTRACT

Fanconi anemia patients suffer from progressive bone marrow failure. An overactive p53 response to DNA damage contributes to the progressive elimination of Fanconi anemia hematopoietic stem and progenitor cells (HSPC), and hence presents a potential target for therapeutic intervention. To investigate whether the cell cycle regulatory protein p21 is the primary mediator of the p53-dependent stem cell loss, p21/Fancd2 double-knockout mice were generated. Surprisingly double mutant mice displayed even more severe loss of HSPCs than Fancd2(-/-) single mutants. p21 deletion did not rescue the abnormal cell cycle profile and had no impact on the long-term repopulating potential of Fancd2(-/-) bone marrow cells. Collectively, our data indicate that p21 has an indispensable role in maintaining a normal HSPC pool and suggest that other p53-targeted factors, not p21, mediate the progressive elimination of HSPC in Fanconi anemia.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Hematopoietic Stem Cells/cytology , Stem Cells/cytology , Animals , Cell Size , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/genetics , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Stem Cells/metabolism
14.
Blood ; 116(24): 5140-8, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20826722

ABSTRACT

Progressive bone marrow failure is a major cause of morbidity and mortality in human Fanconi Anemia patients. In an effort to develop a Fanconi Anemia murine model to study bone marrow failure, we found that Fancd2(-/-) mice have readily measurable hematopoietic defects. Fancd2 deficiency was associated with a significant decline in the size of the c-Kit(+)Sca-1(+)Lineage(-) (KSL) pool and reduced stem cell repopulation and spleen colony-forming capacity. Fancd2(-/-) KSL cells showed an abnormal cell cycle status and loss of quiescence. In addition, the supportive function of the marrow microenvironment was compromised in Fancd2(-/-) mice. Treatment with Sirt1-mimetic and the antioxidant drug, resveratrol, maintained Fancd2(-/-) KSL cells in quiescence, improved the marrow microenvironment, partially corrected the abnormal cell cycle status, and significantly improved the spleen colony-forming capacity of Fancd2(-/-) bone marrow cells. We conclude that Fancd2(-/-) mice have readily quantifiable hematopoietic defects, and that this model is well suited for pharmacologic screening studies.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia/drug therapy , Hematopoietic System/drug effects , Stilbenes/pharmacology , Animals , Antioxidants , Bone Marrow/drug effects , Cell Cycle , Cell Lineage , Colony-Forming Units Assay , Fanconi Anemia Complementation Group D2 Protein/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Resveratrol , Spleen/cytology , Stilbenes/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...