Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Blood Adv ; 6(17): 5049-5060, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35797240

ABSTRACT

Despite the clinical benefit associated with gilteritinib in relapsed/refractory acute myeloid leukemia (AML), most patients eventually develop resistance through unknown mechanisms. To delineate the mechanistic basis of resistance to gilteritinib, we performed targeted sequencing and scRNASeq on primary FLT3-ITD-mutated AML samples. Co-occurring mutations in RAS pathway genes were the most common genetic abnormalities, and unresponsiveness to gilteritinib was associated with increased expression of bone marrow-derived hematopoietic cytokines and chemokines. In particular, we found elevated expression of the TEK-family kinase, BMX, in gilteritinib-unresponsive patients pre- and post-treatment. BMX contributed to gilteritinib resistance in FLT3-mutant cell lines in a hypoxia-dependent manner by promoting pSTAT5 signaling, and these phenotypes could be reversed with pharmacological inhibition and genetic knockout. We also observed that inhibition of BMX in primary FLT3-mutated AML samples decreased chemokine secretion and enhanced the activity of gilteritinib. Collectively, these findings indicate a crucial role for microenvironment-mediated factors modulated by BMX in the escape from targeted therapy and have implications for the development of novel therapeutic interventions to restore sensitivity to gilteritinib.


Subject(s)
Aniline Compounds , Leukemia, Myeloid, Acute , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Protein-Tyrosine Kinases/genetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Tumor Microenvironment , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/therapeutic use
3.
Clin Cancer Res ; 28(12): 2536-2546, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35344039

ABSTRACT

PURPOSE: To evaluate the safety, activity, and emergence of FLT3-kinase domain (KD) mutations with combination therapy of crenolanib and sorafenib in acute myeloid leukemia (AML) with FLT3-internal tandem duplication (ITD). PATIENTS AND METHODS: After in vitro and xenograft efficacy studies using AML cell lines that have FLT3-ITD with or without FLT3-KD mutation, a pilot study was performed with crenolanib (67 mg/m2/dose, three times per day on days 1-28) and two dose levels of sorafenib (150 and 200 mg/m2/day on days 8-28) in 9 pediatric patients with refractory/relapsed FLT3-ITD-positive AML. Pharmacokinetic, pharmacodynamic, and FLT3-KD mutation analysis were done in both preclinical and clinical studies. RESULTS: The combination of crenolanib and sorafenib in preclinical models showed synergy without affecting pharmacokinetics of each agent, inhibited p-STAT5 and p-ERK for up to 8 hours, and led to significantly better leukemia response (P < 0.005) and survival (P < 0.05) compared with single agents. Fewer FLT3-KD mutations emerged with dose-intensive crenolanib (twice daily) and low-intensity sorafenib (three times/week) compared with daily crenolanib or sorafenib (P < 0.05). The crenolanib and sorafenib combination was tolerable without dose-limiting toxicities, and three complete remissions (one with incomplete count recovery) and one partial remission were observed in 8 evaluable patients. Median crenolanib apparent clearance showed a nonsignificant decrease during treatment (45.0, 40.5, and 20.3 L/hour/m2 on days 1, 7, and 14, respectively) without drug-drug interaction. Only 1 patient developed a FLT3-KD mutation (FLT3 F691L). CONCLUSIONS: The combination of crenolanib and sorafenib was tolerable with antileukemic activities and rare emergence of FLT3-TKD mutations, which warrants further investigation.


Subject(s)
Antineoplastic Agents , Benzimidazoles , Leukemia, Myeloid, Acute , Piperidines , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Child , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Phenylurea Compounds , Pilot Projects , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Sorafenib/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
4.
Cancers (Basel) ; 15(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36612026

ABSTRACT

Acute myeloid leukemia (AML) with mutations in the tumor suppressor gene TP53 confers a dismal prognosis with 3-year overall survival of <5%. While inhibition of kinases involved in cell cycle regulation induces synthetic lethality in a variety of TP53 mutant cancers, this strategy has not been evaluated in mutant TP53 AML. Previously, we demonstrated that TP-0903 is a novel multikinase inhibitor with low nM activity against AURKA/B, Chk1/2, and other cell cycle regulators. Here, we evaluated the preclinical activity of TP-0903 in TP53 mutant AML cell lines, including a single-cell clone of MV4-11 containing a TP53 mutation (R248W), Kasumi-1 (R248Q), and HL-60 (TP 53 null). TP-0903 inhibited cell viability (IC50, 12−32 nM) and induced apoptosis at 50 nM. By immunoblot, 50 nM TP-0903 upregulated pChk1/2 and pH2AX, suggesting induction of DNA damage. The combination of TP-0903 and decitabine was additive in vitro, and in vivo significantly prolonged median survival compared to single-agent treatments in mice xenografted with HL-60 (vehicle, 46 days; decitabine, 55 days; TP-0903, 63 days; combination, 75 days) or MV4-11 (R248W) (51 days; 62 days; 81 days; 89 days) (p < 0.001). Together, these results provide scientific premise for the clinical evaluation of TP-0903 in combination with decitabine in TP53 mutant AML.

5.
Blood Adv ; 5(23): 5041-5046, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34614509

ABSTRACT

Drug resistance and relapse are common challenges in acute myeloid leukemia (AML), particularly in an aggressive subset bearing internal tandem duplications (ITDs) of the FLT3 receptor (FLT3-ITD+). The tyrosine kinase inhibitor gilteritinib is approved for the treatment of relapse/refractory AML with FLT3 mutations, yet resistance to gilteritinib remains a clinical concern, and the underlying mechanisms remain incompletely understood. Using transcriptomic analyses and functional validation studies, we identified the calcium-binding proteins S100A8 and S100A9 (S100A8/A9) as contributors to gilteritinib resistance in FLT3-ITD+ AML. Exposure of FLT3-ITD+ AML cells to gilteritinib increased S100A8/A9 expression in vivo and in vitro and decreased free calcium levels, and genetic manipulation of S100A9 was associated with altered sensitivity to gilteritinib. Using a transcription factor screen, we identified the transcriptional corepressor BCL6, as a regulator of S100A9 expression and found that gilteritinib decreased BCL6 binding to the S100A9 promoter, thereby increasing S100A9 expression. Furthermore, pharmacological inhibition of BCL6 accelerated the growth rate of gilteritinib-resistant FLT3-ITD+ AML cells, suggesting that S100A9 is a functional target of BCL6. These findings shed light on mechanisms of resistance to gilteritinib through regulation of a target that can be therapeutically exploited to enhance the antileukemic effects of gilteritinib.


Subject(s)
Leukemia, Myeloid, Acute , Aniline Compounds , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-bcl-6 , Pyrazines , Up-Regulation
6.
Mol Cancer Ther ; 20(11): 2207-2217, 2021 11.
Article in English | MEDLINE | ID: mdl-34518298

ABSTRACT

Acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITD) mutation is an aggressive hematologic malignancy associated with frequent relapse and poor overall survival. The tyrosine kinase inhibitor gilteritinib is approved for the treatment of relapse/refractory AML with FLT3 mutations, yet its mechanism of action is not completely understood. Here, we sought to identify additional therapeutic targets that can be exploited to enhance gilteritinib's antileukemic effect. Based on unbiased transcriptomic analyses, we identified the glutamine transporter SNAT1 (SLC38A1) as a novel target of gilteritinib that leads to impaired glutamine uptake and utilization within leukemic cells. Using metabolomics and metabolic flux analyses, we found that gilteritinib decreased glutamine metabolism through the TCA cycle and cellular levels of the oncometabolite 2-hydroxyglutarate. In addition, gilteritinib treatment was associated with decreased ATP production and glutathione synthesis and increased reactive oxygen species, resulting in cellular senescence. Finally, we found that the glutaminase inhibitor CB-839 enhanced antileukemic effect of gilteritinib in ex vivo studies using human primary FLT3-ITD-positive AML cells harboring mutations in the enzyme isocitrate dehydrogenase, which catalyzes the oxidative decarboxylation of isocitrate, producing α-ketoglutarate. Collectively, this work has identified a previously unrecognized, gilteritinib-sensitive metabolic pathway downstream of SLC38A1 that causes decreased glutaminolysis and disruption of redox homeostasis. These findings provide a rationale for the development and therapeutic exploration of targeted combinatorial treatment strategies for this subset of relapse/refractory AML.


Subject(s)
Aniline Compounds/therapeutic use , Glutamine/drug effects , Leukemia, Myeloid, Acute/drug therapy , Pyrazines/therapeutic use , fms-Like Tyrosine Kinase 3/metabolism , Aniline Compounds/pharmacology , Animals , Female , Humans , Mice , Pyrazines/pharmacology
7.
Clin Transl Sci ; 14(1): 137-142, 2021 01.
Article in English | MEDLINE | ID: mdl-32905646

ABSTRACT

Reduced expression of the uptake transporter, OCTN1 (SLC22A4), has been reported as a strong predictor of poor event-free and overall survival in multiple cohorts of patients with acute myeloid leukemia (AML) receiving the cytidine nucleoside analog, cytarabine (Ara-C). To further understand the mechanistic basis of interindividual variability in the functional expression of OCTN1 in AML, we hypothesized a mechanistic connection to DNA methylation-based epigenetic repression of SLC22A4. We found increased basal SLC22A4 methylation was associated with decreased Ara-C uptake in AML cell lines. Pre-treatment with hypomethylating agents, 5-azacytidine, or decitabine, restored SLC22A4 mRNA expression, increased cellular uptake of Ara-C, and was associated with increased cellular sensitivity to Ara-C compared with vehicle-treated cells. Additionally, lower SLC22A4 methylation status was associated with distinct clinical advantages in both adult and pediatric patients with AML. These findings suggest a regulatory mechanism is involved in the interindividual variability in response to Ara-C, and provides a basis for the integration of hypomethylating agents into Ara-C-based treatment regimens.


Subject(s)
Cytarabine/pharmacology , Drug Resistance, Neoplasm/genetics , Epigenetic Repression/genetics , Leukemia, Myeloid, Acute/drug therapy , Organic Cation Transport Proteins/genetics , Symporters/genetics , Adolescent , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/pharmacology , Azacitidine/therapeutic use , Cell Line, Tumor , Child , Clinical Trials, Phase III as Topic , Cytarabine/therapeutic use , DNA Methylation/drug effects , DNA Methylation/genetics , Datasets as Topic , Decitabine/pharmacology , Decitabine/therapeutic use , Epigenetic Repression/drug effects , Female , Gene Expression Regulation, Leukemic/drug effects , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Organic Cation Transport Proteins/metabolism , Progression-Free Survival , Randomized Controlled Trials as Topic , Symporters/metabolism , Young Adult
8.
JCI Insight ; 5(23)2020 12 03.
Article in English | MEDLINE | ID: mdl-33268594

ABSTRACT

Effective treatment for AML is challenging due to the presence of clonal heterogeneity and the evolution of polyclonal drug resistance. Here, we report that TP-0903 has potent activity against protein kinases related to STAT, AKT, and ERK signaling, as well as cell cycle regulators in biochemical and cellular assays. In vitro and in vivo, TP-0903 was active in multiple models of drug-resistant FLT3 mutant AML, including those involving the F691L gatekeeper mutation and bone marrow microenvironment-mediated factors. Furthermore, TP-0903 demonstrated preclinical activity in AML models with FLT3-ITD and common co-occurring mutations in IDH2 and NRAS genes. We also showed that TP-0903 had ex vivo activity in primary AML cells with recurrent mutations including MLL-PTD, ASXL1, SRSF2, and WT1, which are associated with poor prognosis or promote clinical resistance to AML-directed therapies. Our preclinical studies demonstrate that TP-0903 is a multikinase inhibitor with potent activity against multiple drug-resistant models of AML that will have an immediate clinical impact in a heterogeneous disease like AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Gene Duplication/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mice , Mice, Nude , Mutation/drug effects , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrimidines/metabolism , Sulfonamides/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
9.
Invest New Drugs ; 38(2): 340-349, 2020 04.
Article in English | MEDLINE | ID: mdl-31102119

ABSTRACT

Activating FLT3 internal tandem duplication (FLT3-ITD) mutations in acute myeloid leukemia (AML) associate with inferior outcomes. We determined that pacritinib, a JAK2/FLT3 inhibitor, has in vitro activity against FLT3-ITD and tyrosine kinase domain (TKD) mutations. Therefore, we conducted a phase I study of pacritinib in combination with chemotherapy in AML patients with FLT3 mutations to determine the pharmacokinetics and preliminary toxicity and clinical activity. Pacritinib was administered at a dose of 100 mg or 200 mg twice daily following a 3 + 3 dose-escalation in combination with cytarabine and daunorubicin (cohort A) or with decitabine induction (cohort B). A total of thirteen patients were enrolled (five in cohort A; eight in cohort B). Dose limiting toxicities include hemolytic anemia and grade 3 QTc prolongation in two patients who received 100 mg. Complete remission was achieved in two patients in cohort A, one of whom had a minor D835Y clone at baseline. One patient in cohort B achieved morphologic leukemia free state. Seven patients (two in cohort A; five in cohort B) had stable disease. In conclusion, pacritinib, an inhibitor of FLT3-ITD and resistant-conferring TKD mutations, was well tolerated and demonstrated preliminary anti-leukemic activity in combination with chemotherapy in patients with FLT3 mutations.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged-Ring Compounds/therapeutic use , Janus Kinase 2/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged-Ring Compounds/adverse effects , Bridged-Ring Compounds/pharmacokinetics , Bridged-Ring Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cytarabine/adverse effects , Cytarabine/therapeutic use , Daunorubicin/adverse effects , Daunorubicin/therapeutic use , Decitabine/adverse effects , Decitabine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Mutation , Pilot Projects , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
10.
Clin Transl Sci ; 12(6): 641-647, 2019 11.
Article in English | MEDLINE | ID: mdl-31350825

ABSTRACT

Fms-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations, common in pediatric acute myeloid leukemia (AML), associate with early relapse and poor prognosis. Past studies have suggested additional cooperative mutations are required for leukemogenesis in FLT3-ITD+ AML. Using RNA sequencing and a next-generation targeted gene panel, we broadly characterize the co-occurring genomic alterations in pediatric cytogenetically normal (CN) FLT3-ITD+ AML to gain a deeper understanding of the clonal patterns and heterogeneity at diagnosis and relapse. We show that chimeric transcripts were present in 21 of 34 (62%) of de novo samples, 2 (6%) of these samples included a rare reoccurring fusion partner BCL11B. At diagnosis, the median number of mutations other than FLT3 per patient was 1 (range 0-3), which involved 8 gene pathways; WT1 and NPM1 mutations were frequently observed (35% and 24%, respectively). Fusion transcripts and high variant allele frequency (VAF) mutants, which included WT1, NPM1, SMARCA2, RAD21, and TYK2, were retained from diagnosis to relapse. We did observe reduction in VAF of simple or single mutation clones, but VAFs were preserved or expanded in more complex clones with multiple mutations. Our data provide the first insight into the genomic complexity of pediatric CN FLT3-ITD+ AML and could help stratify future targeted treatment strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genetic Heterogeneity , Leukemia, Myeloid, Acute/genetics , Neoplasm Recurrence, Local/genetics , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Child , Child, Preschool , Cytogenetic Analysis , Disease-Free Survival , Female , Gene Duplication , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Nucleophosmin , Precision Medicine , Prognosis , Randomized Controlled Trials as Topic , Remission Induction/methods , Sequence Analysis, RNA , Tandem Repeat Sequences/genetics , Young Adult
11.
Nat Commun ; 10(1): 2189, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097698

ABSTRACT

Improvement in survival has been achieved for children and adolescents with AML but is largely attributed to enhanced supportive care as opposed to the development of better treatment regimens. High risk subtypes continue to have poor outcomes with event free survival rates <40% despite the use of high intensity chemotherapy in combination with hematopoietic stem cell transplant. Here we combine high-throughput screening, intracellular accumulation assays, and in vivo efficacy studies to identify therapeutic strategies for pediatric AML. We report therapeutics not currently used to treat AML, gemcitabine and cabazitaxel, have broad anti-leukemic activity across subtypes and are more effective relative to the AML standard of care, cytarabine, both in vitro and in vivo. JAK inhibitors are selective for acute megakaryoblastic leukemia and significantly prolong survival in multiple preclinical models. Our approach provides advances in the development of treatment strategies for pediatric AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Janus Kinase Inhibitors/pharmacology , Leukemia, Experimental/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Adult , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Bone Marrow/radiation effects , Bone Marrow Transplantation , Cell Line, Tumor , Child , Child, Preschool , Cytarabine/pharmacology , Cytarabine/therapeutic use , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , High-Throughput Screening Assays/methods , Humans , Infant , Janus Kinase Inhibitors/therapeutic use , Leukemia, Experimental/etiology , Leukemia, Experimental/mortality , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Taxoids/pharmacology , Taxoids/therapeutic use , Whole-Body Irradiation/adverse effects , Xenograft Model Antitumor Assays , Young Adult , Gemcitabine
12.
Nat Commun ; 10(1): 244, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651561

ABSTRACT

FLT3 mutations are prevalent in AML patients and confer poor prognosis. Crenolanib, a potent type I pan-FLT3 inhibitor, is effective against both internal tandem duplications and resistance-conferring tyrosine kinase domain mutations. While crenolanib monotherapy has demonstrated clinical benefit in heavily pretreated relapsed/refractory AML patients, responses are transient and relapse eventually occurs. Here, to investigate the mechanisms of crenolanib resistance, we perform whole exome sequencing of AML patient samples before and after crenolanib treatment. Unlike other FLT3 inhibitors, crenolanib does not induce FLT3 secondary mutations, and mutations of the FLT3 gatekeeper residue are infrequent. Instead, mutations of NRAS and IDH2 arise, mostly as FLT3-independent subclones, while TET2 and IDH1 predominantly co-occur with FLT3-mutant clones and are enriched in crenolanib poor-responders. The remaining patients exhibit post-crenolanib expansion of mutations associated with epigenetic regulators, transcription factors, and cohesion factors, suggesting diverse genetic/epigenetic mechanisms of crenolanib resistance. Drug combinations in experimental models restore crenolanib sensitivity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , Epigenesis, Genetic/drug effects , Female , GTP Phosphohydrolases/genetics , HEK293 Cells , Humans , Inhibitory Concentration 50 , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Membrane Proteins/genetics , Mice , Middle Aged , Mutation/drug effects , Mutation/genetics , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Tandem Repeat Sequences/genetics , Treatment Outcome , Exome Sequencing
13.
Bio Protoc ; 8(15)2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30899769

ABSTRACT

Hypoxia is a condition in which there is a decrease in oxygen supply to the cellular environment. Changes to cellular oxygen levels can lead to transcriptional changes of oxygen-regulated genes. Reporter assays are used to study gene expression alteration and modifications in response to environmental changes. Dual-reporter assays allow the simultaneous measurement of two different genes within a single cell, thus improving experimental accuracy. Within this protocol, we describe the utilization of the LightSwitch Dual Assay System to measure BMX expression in response to hypoxic conditions.

14.
J Clin Invest ; 128(1): 369-380, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29227282

ABSTRACT

Oncogenic addiction to the Fms-like tyrosine kinase 3 (FLT3) is a hallmark of acute myeloid leukemia (AML) that harbors the FLT3-internal tandem duplication (FLT3-ITD) mutation. While FLT3 inhibitors like sorafenib show initial therapeutic efficacy, resistance rapidly develops through mechanisms that are incompletely understood. Here, we used RNA-Seq-based analysis of patient leukemic cells and found that upregulation of the Tec family kinase BMX occurs during sorafenib resistance. This upregulation was recapitulated in an in vivo murine FLT3-ITD-positive (FLT3-ITD+) model of sorafenib resistance. Mechanistically, the antiangiogenic effects of sorafenib led to increased bone marrow hypoxia, which contributed to HIF-dependent BMX upregulation. In in vitro experiments, hypoxia-dependent BMX upregulation was observed in both AML and non-AML cell lines. Functional studies in human FLT3-ITD+ cell lines showed that BMX is part of a compensatory signaling mechanism that promotes AML cell survival during FLT3 inhibition. Taken together, our results demonstrate that hypoxia-dependent upregulation of BMX contributes to therapeutic resistance through a compensatory prosurvival signaling mechanism. These results also reveal the role of off-target drug effects on tumor microenvironment and development of acquired drug resistance. We propose that the bone marrow niche can be altered by anticancer therapeutics, resulting in drug resistance through cell-nonautonomous microenvironment-dependent effects.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Protein-Tyrosine Kinases/biosynthesis , Tumor Microenvironment , Up-Regulation , Cell Hypoxia , Child , Child, Preschool , Female , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Protein-Tyrosine Kinases/genetics , Signal Transduction , Sorafenib/pharmacology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
16.
Arch Pathol Lab Med ; 138(11): 1474-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25357108

ABSTRACT

CONTEXT: Invasive fungal infections are an important cause of morbidity and mortality among immunocompromised patients. OBJECTIVE: To design and evaluate a multiplexed assay aimed at quantitative detection and differentiation of the 5 molds that are most commonly responsible for pulmonary infections. DESIGN: Using labeled primer polymerase chain reaction chemistry, an assay was designed to target the 5.8S and 28S ribosomal RNA genes of Aspergillus spp, Fusarium spp, Scedosporium spp, and members of the order Mucorales ( Rhizopus oryzae , Rhizopus microsporus, Cunninghamella bertholletiae, Mucor circinelloides, Lichtheimia corymbifera, and Rhizomucor pusillus). This assay was split into 2 multiplexed reactions and was evaluated using both samples seeded with purified nucleic acid from 42 well-characterized clinical fungal isolates and 105 archived samples (47 blood [45%], 42 bronchoalveolar lavage fluid [40%], and 16 tissue [15%]) collected from rabbit models of invasive pulmonary fungal infections. RESULTS: Assay detection sensitivity was less than 25 copies of the target sequence per reaction for Aspergillus spp, 5 copies for Fusarium spp and Scedosporium spp, and 10 copies for the Mucorales. The assay showed quantitative linearity from 5 × 10(1) to 5 × 10(5) copies of target sequence per reaction. Sensitivities and specificities for bronchoalveolar lavage fluid, tissue, and blood samples were 0.86 and 0.99, 0.60 and 1.00, and 0.46 and 1.00, respectively. CONCLUSIONS: Labeled primer polymerase chain reaction permits rapid, quantitative detection and differentiation of common agents of invasive fungal infection. The assay described herein shows promise for clinical implementation that may have a significant effect on the rapid diagnosis and treatment of patients' severe infections caused by these pulmonary fungal pathogens.


Subject(s)
Lung Diseases, Fungal/diagnosis , Lung Diseases, Fungal/microbiology , Multiplex Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/methods , Animals , Aspergillus/genetics , Disease Models, Animal , Female , Fusarium/genetics , Genes, Fungal , Humans , Molecular Diagnostic Techniques/methods , Molecular Diagnostic Techniques/statistics & numerical data , Mucorales/genetics , Multiplex Polymerase Chain Reaction/statistics & numerical data , Mycological Typing Techniques/methods , Mycological Typing Techniques/statistics & numerical data , RNA, Fungal/analysis , RNA, Fungal/genetics , RNA, Ribosomal, 28S/analysis , RNA, Ribosomal, 28S/genetics , RNA, Ribosomal, 5.8S/analysis , RNA, Ribosomal, 5.8S/genetics , Rabbits , Real-Time Polymerase Chain Reaction/statistics & numerical data , Scedosporium/genetics , Sensitivity and Specificity
17.
J Clin Virol ; 58(1): 84-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23751960

ABSTRACT

BACKGROUND: Several analyte specific reagents (ASRs) are available for the detection and differentiation of HSV-1, HSV-2, and VZV in clinical specimens. However, there is limited data on the test performance of these reagents used in multiplexed PCR assays. OBJECTIVE: This study compared the performance of two multiplexed ASR sets for detection of HSV-1, HSV-2, and VZV in dermal specimens. STUDY DESIGN: Two commercially available ASRs were combined to produce multiplexed PCR assays for simultaneous detection of HSV-1, HSV-2, and VZV. Seeded samples were used to determine the limit of detection (LOD) for each assay. Patient samples (n=156) were tested in duplicate and results for each method compared to the reference standard of culture. Both extracted and unextracted specimens were used in the study. RESULTS: Both multiplexed PCR assays showed similar test performance, with minimal LOD differences observed. The LOD was 10(3) copies/mL for HSV-1 and HSV-2 using the Focus assay compared to 5×10(3) copies/mL and 2×10(4) copies/mL, respectively for the EraGen assay. Both assays showed equal performance for VZV with a LOD of 5×10(3) copies/mL. Analytical specificity testing showed no cross reactivity with other selected DNA viruses. Both assays showed similar performance when clinical samples were tested using both extracted and unextracted specimens. CONCLUSION: Commercially available ASRs can be successfully multiplexed for the PCR detection of HSV-1, HSV-2, and VZV using dermal specimens. Either direct testing or nucleic acid extracted specimens can be used with similar performance in these assays.


Subject(s)
Chickenpox/diagnosis , Herpesviridae Infections/diagnosis , Molecular Diagnostic Techniques/methods , Mucous Membrane/virology , Multiplex Polymerase Chain Reaction/methods , Skin/virology , Chickenpox/virology , Herpesviridae Infections/virology , Herpesvirus 1, Human/isolation & purification , Herpesvirus 2, Human/isolation & purification , Humans , Reagent Kits, Diagnostic , Sensitivity and Specificity , Varicellovirus/isolation & purification , Virology/methods
18.
J Clin Microbiol ; 51(3): 959-66, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23303501

ABSTRACT

Invasive fungal infections are a significant cause of morbidity and mortality among immunocompromised patients. Early and accurate identification of these pathogens is central to direct therapy and to improve overall outcome. PCR coupled with electrospray ionization mass spectrometry (PCR/ESI-MS) was evaluated as a novel means for identification of fungal pathogens. Using a database grounded by 60 ATCC reference strains, a total of 394 clinical fungal isolates (264 molds and 130 yeasts) were analyzed by PCR/ESI-MS; results were compared to phenotypic identification, and discrepant results were sequence confirmed. PCR/ESI-MS identified 81.4% of molds to either the genus or species level, with concordance rates of 89.7% and 87.4%, respectively, to phenotypic identification. Likewise, PCR/ESI-MS was able to identify 98.4% of yeasts to either the genus or species level, agreeing with 100% of phenotypic results at both the genus and species level. PCR/ESI-MS performed best with Aspergillus and Candida isolates, generating species-level identification in 94.4% and 99.2% of isolates, respectively. PCR/ESI-MS is a promising new technology for broad-range detection and identification of medically important fungal pathogens that cause invasive mycoses.


Subject(s)
Fungi/isolation & purification , Microbiological Techniques/methods , Molecular Diagnostic Techniques/methods , Mycology/methods , Polymerase Chain Reaction/methods , Spectrometry, Mass, Electrospray Ionization/methods , Fungi/classification , Fungi/genetics , Humans , Mycoses/diagnosis , Mycoses/microbiology
19.
Med Mycol ; 50(7): 775-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22435876

ABSTRACT

Invasive fungal infections are the cause of serious morbidity and high mortality in immunocompromised patients. Early laboratory diagnostic options remain limited; however, rapid detection and accurate identification may improve outcome. Herein, multiplexed PCR followed by liquid-phase array was evaluated for detection and identification of common respiratory fungal pathogens, including Aspergillus fumigatus, Rhizopus microsporus, Scedosporium apiospermum and Fusarium solani. The limit of detection ranged 0.1-1 ng of DNA, depending on the fungus being tested. Primer cross-reactivity was seen for some fungi: Aspergillus flavus primers detected Aspergillus oryzae; Scedosporium apiospermum primers detected Paecilomyces lilacinus, and Aspergillus terreus primers detected S. apiospermum. PCR followed by liquid-phase array is potentially useful for the identification of clinically relevant fungal pathogens.


Subject(s)
Fungi/isolation & purification , Microarray Analysis/methods , Molecular Diagnostic Techniques/methods , Multiplex Polymerase Chain Reaction/methods , Mycology/methods , Mycoses/diagnosis , Respiratory Tract Diseases/diagnosis , Cross Reactions , DNA Primers/genetics , Fungi/classification , Humans , Mycoses/microbiology , Respiratory Tract Diseases/microbiology , Sensitivity and Specificity
20.
Mol Microbiol ; 80(5): 1296-312, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21435039

ABSTRACT

Although it is known that Campylobacter jejuni invade the cells that line the human intestinal tract, the bacterial proteins that enable this pathogen to survive within Campylobacter-containing vacuoles (CCV) have not been identified. Here, we describe the identification and characterization of a protein that we termed CiaI for Campylobacter invasion antigen involved in intracellular survival. We show that CiaI harbours an amino-terminal type III secretion sequence and is secreted from C. jejuni through the flagellar type III secretion system. In addition, the ciaI mutant was impaired in intracellular survival when compared with a wild-type strain, as judged by the gentamicin-protection assay. Fluorescence microscopy examination of epithelial cells infected with the C. jejuni ciaI mutant revealed that the CCV were more frequently co-localized with Cathepsin D (a lysosomal marker) than the CCV in cells infected with a C. jejuni wild-type strain. Ectopic expression of CiaI-GFP in epithelial cells yielded a punctate phenotype not observed with the other C. jejuni genes, and this phenotype was abolished by mutation of a dileucine motif located in the carboxy-terminus of the protein. Based on the data, we conclude that CiaI contributes to the ability of C. jejuni to survive within epithelial cells.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter Infections/microbiology , Campylobacter jejuni/growth & development , Campylobacter jejuni/metabolism , Epithelial Cells/microbiology , Microbial Viability , Bacterial Proteins/genetics , Campylobacter jejuni/genetics , Gene Expression Regulation, Bacterial , Humans , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...