Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 16(12): 2668-2676, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28978721

ABSTRACT

An increasing number of cancers are known to harbor mutations, translocations, or amplifications in the fibroblast growth factor receptor (FGFR) family of kinases. The FGFR inhibitors evaluated in clinical trials to date have shown promise at treating these cancers. Here, we describe PRN1371, an irreversible covalent inhibitor of FGFR1-4 targeting a cysteine within the kinase active site. PRN1371 demonstrated strong FGFR potency and excellent kinome-wide selectivity in a number of biochemical and cellular assays, including in various cancer cell lines exhibiting FGFR alterations. Furthermore, PRN1371 maintained FGFR inhibition in vivo, not only when circulating drug levels were high but also after the drug had been cleared from circulation, indicating the possibility of sustained FGFR inhibition in the clinic without the need for continuous drug exposure. Durable tumor regression was also obtained in multiple tumor xenografts and patient-derived tumor xenograft models and was sustained even using an intermittent dosing strategy that provided drug holidays. PRN1371 is currently under clinical investigation for treatment of patients with solid tumors. Mol Cancer Ther; 16(12); 2668-76. ©2017 AACR.


Subject(s)
Pyridones/therapeutic use , Pyrimidines/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Pyridones/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
2.
J Med Chem ; 60(15): 6516-6527, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28665128

ABSTRACT

Aberrant signaling of the FGF/FGFR pathway occurs frequently in cancers and is an oncogenic driver in many solid tumors. Clinical validation of FGFR as a therapeutic target has been demonstrated in bladder, liver, lung, breast, and gastric cancers. Our goal was to develop an irreversible covalent inhibitor of FGFR1-4 for use in oncology indications. An irreversible covalent binding mechanism imparts many desirable pharmacological benefits including high potency, selectivity, and prolonged target inhibition. Herein we report the structure-based design, medicinal chemistry optimization, and unique ADME assays of our irreversible covalent drug discovery program which culminated in the discovery of compound 34 (PRN1371), a highly selective and potent FGFR1-4 inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pyridones/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dogs , Drug Design , Drug Stability , Female , Humans , Intestinal Absorption , Macaca fascicularis , Male , Pyridones/administration & dosage , Pyridones/chemical synthesis , Pyridones/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Solubility , Structure-Activity Relationship
3.
JCO Precis Oncol ; 1: 1-19, 2017 Nov.
Article in English | MEDLINE | ID: mdl-35172518

ABSTRACT

PURPOSE: The androgen receptor (AR) is increasingly recognized as a potential biomarker for identifying a subset of patients with possible hormonally driven triple-negative breast cancer (TNBC). However, its performance as a companion diagnostic remains elusive. Thus, we evaluated AR expression by immunohistochemistry in patients with advanced TNBC before treatment with the AR inhibitor enzalutamide. METHODS: We optimized and validated immunohistochemistry assays in breast and prostate cancer cell lines and tissues using two commercial AR monoclonal antibodies (SP107 and AR441). AR expression was then examined in patients with advanced TNBC enrolled in a phase II study of enzalutamide (ClinicalTrials.gov identifier: NCT01889238) on archived or fresh tissue before treatment. Association with clinical response was assessed by sensitivity, specificity, positive predictive value (PPV), drop-out rate, and survival. RESULTS: AR expression was detected in 80% and 63% of breast cancer tissue using SP107 and AR441, respectively. SP107 was selected for additional analyses because of its higher sensitivity and robustness. Total AR nuclear staining demonstrated the best accuracy in predicting clinical response (area under receiver operating characteristic curve, 0.72; P = .0001). At a threshold of 10%, 74.6% of patients were AR positive, leading to 30% PPV, 90% sensitivity, and 30% specificity. These patients showed a significantly higher median progression-free survival (hazard ratio, 0.56; 95% CI, 0.36 to 0.88; P = .011) and overall survival (hazard ratio, 0.54; 95% CI, 0.32 to 0.91; P = .019) compared with those with AR-negative (< 10%) TNBC. CONCLUSION: At a threshold of ≥ 10% nuclear expression, the AR was associated with TNBC response to enzalutamide. However, the modest PPV may restrict its clinical application, and additional diagnostic tools may be helpful for improved patient selection.

4.
Mol Cancer Res ; 14(11): 1054-1067, 2016 11.
Article in English | MEDLINE | ID: mdl-27565181

ABSTRACT

Androgen receptor (AR) is expressed in 90% of estrogen receptor alpha-positive (ER+) breast tumors, but its role in tumor growth and progression remains controversial. Use of two anti-androgens that inhibit AR nuclear localization, enzalutamide and MJC13, revealed that AR is required for maximum ER genomic binding. Here, a novel global examination of AR chromatin binding found that estradiol induced AR binding at unique sites compared with dihydrotestosterone (DHT). Estradiol-induced AR-binding sites were enriched for estrogen response elements and had significant overlap with ER-binding sites. Furthermore, AR inhibition reduced baseline and estradiol-mediated proliferation in multiple ER+/AR+ breast cancer cell lines, and synergized with tamoxifen and fulvestrant. In vivo, enzalutamide significantly reduced viability of tamoxifen-resistant MCF7 xenograft tumors and an ER+/AR+ patient-derived model. Enzalutamide also reduced metastatic burden following cardiac injection. Finally, in a comparison of ER+/AR+ primary tumors versus patient-matched local recurrences or distant metastases, AR expression was often maintained even when ER was reduced or absent. These data provide preclinical evidence that anti-androgens that inhibit AR nuclear localization affect both AR and ER, and are effective in combination with current breast cancer therapies. In addition, single-agent efficacy may be possible in tumors resistant to traditional endocrine therapy, as clinical specimens of recurrent disease demonstrate AR expression in tumors with absent or refractory ER. IMPLICATIONS: This study suggests that AR plays a previously unrecognized role in supporting E2-mediated ER activity in ER+/AR+ breast cancer cells, and that enzalutamide may be an effective therapeutic in ER+/AR+ breast cancers. Mol Cancer Res; 14(11); 1054-67. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , Chromatin/metabolism , Drug Resistance, Neoplasm/drug effects , Phenylthiohydantoin/analogs & derivatives , Receptors, Androgen/metabolism , Receptors, Estrogen/genetics , Tamoxifen/administration & dosage , Anilides/pharmacology , Benzamides , Binding Sites , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclohexanes/pharmacology , Disease Progression , Estradiol , Female , Humans , MCF-7 Cells , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/pharmacology , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology
5.
Nat Chem Biol ; 11(7): 525-31, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26006010

ABSTRACT

Drugs with prolonged on-target residence times often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here we made progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Using an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, we identified potent and selective BTK inhibitors that demonstrated biochemical residence times spanning from minutes to 7 d. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK for more than 18 h after clearance from the circulation. The inverted cyanoacrylamide strategy was further used to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating the generalizability of the approach. Targeting of noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates 'residence time by design', the ability to modulate and improve the duration of target engagement in vivo.


Subject(s)
Acrylamides/pharmacokinetics , B-Lymphocytes/drug effects , Cyanoacrylates/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Acrylamides/chemical synthesis , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Cell Line, Tumor , Crystallography, X-Ray , Cyanoacrylates/chemical synthesis , Dasatinib , Female , Gene Expression , Humans , Ligands , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sf9 Cells , Spodoptera , Structure-Activity Relationship , Substrate Specificity , Thiazoles/pharmacokinetics , Time Factors
6.
Proc Natl Acad Sci U S A ; 110(15): 6079-84, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530240

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) promotes mobilization of CD11b(+)Gr1(+) myeloid cells and has been implicated in resistance to anti-VEGF therapy in mouse models. High G-CSF production has been associated with a poor prognosis in cancer patients. Here we show that activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. Several growth factors induced G-CSF expression by a MEK-dependent mechanism. Inhibition of G-CSF release with a MEK inhibitor markedly reduced G-CSF production in vitro and synergized with anti-VEGF antibodies to reduce CD11b(+)Ly6G(+) neutrophil mobilization and tumor growth and led to increased survival in animal models of cancer, including a genetically engineered mouse model of pancreatic adenocarcinoma. Analysis of biopsies from pancreatic cancer patients revealed increased phospho-MEK, G-CSF, and Ets expression and enhanced neutrophil recruitment compared with normal pancreata. These results provide insights into G-CSF regulation and on the mechanism of action of MEK inhibitors and point to unique anticancer strategies.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , MAP Kinase Signaling System , Neutrophils/cytology , Proto-Oncogene Protein c-ets-2/metabolism , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Binding Sites , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Mice, Transgenic , Neoplasms/metabolism , Neovascularization, Pathologic , Neutrophil Infiltration , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 108(28): 11590-5, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21709213

ABSTRACT

PlGF, one of the ligands for VEGFR-1, has been implicated in tumor angiogenesis. However, more recent studies indicate that genetic or pharmacological inhibition of PlGF signaling does not result in reduction of microvascular density in a variety of tumor models. Here we screened 12 human tumor cell lines and identified 3 that are growth inhibited by anti-PlGF antibodies in vivo. We found that efficacy of anti-PlGF treatment strongly correlates with VEGFR-1 expression in tumor cells, but not with antiangiogenesis. In addition, PlGF induced VEGFR-1 signaling and biological responses in tumor cell lines sensitive to anti-PlGF, but not in refractory tumor cell lines or in endothelial cells. Also, genetic ablation of VEGFR-1 signaling in the host did not affect the efficacy of PlGF blockade. Collectively, these findings suggest that the role of PlGF in tumorigenesis largely consists of promoting autocrine/paracrine growth of tumor cells expressing a functional VEGFR-1 rather than stimulation of angiogenesis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , Pregnancy Proteins/antagonists & inhibitors , Pregnancy Proteins/immunology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Cell Line, Tumor , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Neoplasms/blood supply , Neoplasms/etiology , Neovascularization, Pathologic , Placenta Growth Factor , Pregnancy Proteins/pharmacology , RNA, Small Interfering/genetics , Signal Transduction , Stromal Cells/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics
8.
J Exp Med ; 207(12): 2581-94, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21059853

ABSTRACT

Gain of chromosome 8 is the most common chromosomal gain in human acute myeloid leukemia (AML). It has been hypothesized that gain of the MYC protooncogene is of central importance in trisomy 8, but the experimental data to support this are limited and controversial. In a mouse model of promyelocytic leukemia in which the MRP8 promoter drives expression of the PML-RARA fusion gene in myeloid cells, a Myc allele is gained in approximately two-thirds of cases as a result of trisomy for mouse chromosome 15. We used this model to test the idea that MYC underlies acquisition of trisomy in AML. We used a retroviral vector to drive expression of wild-type, hypermorphic, or hypomorphic MYC in bone marrow that expressed the PML-RARA transgene. MYC retroviruses cooperated in myeloid leukemogenesis and suppressed gain of chromosome 15. When the PML-RARA transgene was expressed in a Myc haploinsufficient background, we observed selection for increased copies of the wild-type Myc allele concomitant with leukemic transformation. In addition, we found that human myeloid leukemias with trisomy 8 have increased MYC. These data show that gain of MYC can contribute to the pathogenic effect of the most common trisomy of human AML.


Subject(s)
Chromosomes, Human, Pair 8 , Genes, myc , Leukemia, Promyelocytic, Acute/genetics , Trisomy , Animals , Cells, Cultured , Disease Models, Animal , Humans , Leukemia, Promyelocytic, Acute/etiology , Mice , Oncogene Proteins, Fusion/genetics , Recurrence
9.
Mol Cell Biol ; 30(9): 2264-79, 2010 May.
Article in English | MEDLINE | ID: mdl-20160012

ABSTRACT

The neurofibromatosis type 1 (NF1) gene encodes the GTPase-activating protein (GAP) neurofibromin, which negatively regulates Ras activity. The yeast Saccharomyces cerevisiae has two neurofibromin homologs, Ira1 and Ira2. To understand how these proteins are regulated, we utilized an unbiased proteomics approach to identify Ira2 and neurofibromin binding partners. We demonstrate that the Gpb1/Krh2 protein binds and negatively regulates Ira2 by promoting its ubiquitin-dependent proteolysis. We extended our findings to show that in mammalian cells, the ETEA/UBXD8 protein directly interacts with and negatively regulates neurofibromin. ETEA contains both UBA and UBX domains. Overexpression of ETEA downregulates neurofibromin in human cells. Purified ETEA, but not a mutant of ETEA that lacks the UBX domain, ubiquitinates the neurofibromin GAP-related domain in vitro. Silencing of ETEA expression increases neurofibromin levels and downregulates Ras activity. These findings provide evidence for conserved ubiquitination pathways regulating the RasGAP proteins Ira2 (in yeast) and neurofibromin (in humans).


Subject(s)
Blood Proteins/metabolism , GTPase-Activating Proteins/metabolism , Neurofibromin 1/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Blood Proteins/chemistry , Cell Line , Down-Regulation/drug effects , Gene Silencing/drug effects , Glucose/pharmacology , Humans , Mass Spectrometry , Membrane Proteins , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Stability/drug effects , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Ubiquitination/drug effects
10.
Adv Cancer Res ; 102: 1-17, 2009.
Article in English | MEDLINE | ID: mdl-19595305

ABSTRACT

More than 25 years have passed since activating mutations in Ras genes were identified in DNA from human tumors. In this time, it has been established beyond doubt that these mutations play a direct role in causing cancer, and do so in collaboration with a number of other oncogenes and tumor suppressors. Oncogenic mutant Ras proteins are resistant to downregulation by GAP-mediated hydrolysis of bound GTP, and therefore signal persistently. Efforts to develop therapies that block Ras oncoprotein function directly have failed. The high affinity of Ras proteins for GTP has discouraged attempts to identify GTP-analogs. Ras processing enzymes have been targeted, but unfortunately, K-Ras, the Ras protein that plays the major role in human cancer, has proven refractory to these approaches. Further progress has been made with drugs that block downstream signaling: the approved drug Sorafenib inhibits Raf kinase, and its clinical benefits in liver cancer are greatest in patients in which the mitogen activated protein kinase (MAPK) signaling pathway is hyperactive. Other Raf kinase inhibitors, as well as drugs that block mitogen-activated protein kinase / extracellular signal-regulated kinase kinase (MEK) and various steps in the PI 3' kinase pathway, are under development. Here we will discuss the complexities of Ras signaling and their effects on targeting the Ras pathway in the future.


Subject(s)
Neurofibromatoses/metabolism , Neurofibromatoses/therapy , ras Proteins/physiology , Humans , Signal Transduction
11.
Cancer Cell ; 9(2): 81-94, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16473276

ABSTRACT

RARA becomes an acute promyelocytic leukemia (APL) oncogene by fusion with any of five translocation partners. Unlike RARalpha, the fusion proteins homodimerize, which may be central to oncogenic activation. This model was tested by replacing PML with dimerization domains from p50NFkappaB (p50-RARalpha) or the rapamycin-sensitive dimerizing peptide of FKBP12 (F3-RARalpha). The X-RARalpha fusions recapitulated in vitro activities of PML-RARalpha. For F3-RARalpha, these properties were rapamycin sensitive. Although in vivo the artificial fusions alone are poor initiators of leukemia, p50-RARalpha readily cooperates with an activated mutant CDw131 to induce APL-like disease. These results demonstrate that the dimerization interface of RARalpha fusion partners is a critical element in APL pathogenesis while pointing to other features of PML for enhancing penetrance and progression.


Subject(s)
Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Animals , Bone Marrow/pathology , Carcinogens/metabolism , Cell Line , DNA/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Down-Regulation/genetics , Mice , Mice, Transgenic , Mutation/genetics , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Structure, Quaternary , Receptors, Cytokine/metabolism , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Transcription, Genetic/genetics
12.
Mol Cell Biol ; 23(13): 4573-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12808098

ABSTRACT

We utilized a mouse model of acute promyelocytic leukemia (APL) to investigate how aberrant activation of cytokine signaling pathways interacts with chimeric transcription factors to generate acute myeloid leukemia. Expression in mice of the APL-associated fusion, PML-RARA, initially has only modest effects on myelopoiesis. Whereas treatment of control animals with interleukin-3 (IL-3) resulted in expanded myelopoiesis without a block in differentiation, PML-RARA abrogated differentiation that normally characterizes the response to IL-3. Retroviral transduction of bone marrow with an IL-3-expressing retrovirus revealed that IL-3 and promyelocytic leukemia-retinoic acid receptor alpha (PML-RARalpha) combined to generate a lethal leukemia-like syndrome in <21 days. We also observed that a constitutively activated mutant IL-3 receptor, beta(c)V449E, cooperated with PML-RARalpha in leukemogenesis, whereas a different activated mutant, beta(c)I374N, did not. Analysis of additional mutations introduced into beta(c)V449E showed that, although tyrosine phosphorylation of beta(c) is necessary for cooperation, the Src homology 2 domain-containing transforming protein binding site is dispensable. Our results indicate that chimeric transcription factors can block the differentiative effects of growth factors. This combination can be potently leukemogenic, but the particular manner in which these types of mutations interact determines the ability of such combinations to generate acute myeloid leukemia.


Subject(s)
Cytokines/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Signal Transduction , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Flow Cytometry , Immunophenotyping , Interleukin-3/metabolism , Karyotyping , Leukemia, Myeloid, Acute/etiology , Mice , Mutation , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Phosphorylation , Receptors, Interleukin-3/metabolism , Retroviridae/genetics , Spleen/cytology , Time Factors , Tyrosine/metabolism , src Homology Domains
13.
Blood ; 102(3): 1072-4, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12689927

ABSTRACT

Acute promyelocytic leukemia (APL) is characterized by the PML-RARA fusion gene. To identify genetic changes that cooperate with PML-RARA, we performed spectral karyotyping analysis of myeloid leukemias from transgenic PML-RARA mice and from mice coexpressing PML-RARA and BCL2, IL3, activated IL3R, or activated FLT3. A cooperating mutation that enhanced survival (BCL2) was not sufficient to complete transformation and was associated with multiple numeric abnormalities, whereas cooperating mutations that deregulated growth and enhanced survival were associated with normal karyotypes (IL3) or simple karyotypic changes (IL3R, FLT3). Recurring abnormalities included trisomy 15 (49%), trisomy 8 (46%), and -X/-Y (54%). The most common secondary abnormality in human APL is +8 or partial trisomy of 8q24, syntenic to mouse 15. These murine leukemias have a defined spectrum of changes that recapitulates, in part, the cytogenetic abnormalities found in human APL. Our results demonstrate that different cooperating events may generate leukemia via different pathways.


Subject(s)
Chromosome Aberrations , Leukemia, Promyelocytic, Acute/genetics , Leukemia/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cell Division/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Karyotyping , Leukemia, Promyelocytic, Acute/etiology , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Transgenic
14.
Blood ; 101(8): 3188-97, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12515727

ABSTRACT

The PML-RAR alpha fusion protein is central to the pathogenesis of acute promyelocytic leukemia (APL). Expression of this protein in transgenic mice initiates myeloid leukemias with features of human APL, but only after a long latency (8.5 months in MRP8 PML-RARA mice). Thus, additional changes contribute to leukemic transformation. Activating mutations of the FLT3 receptor tyrosine kinase are common in human acute myeloid leukemias and are frequent in human APL. To assess how activating mutations of FLT3 contribute to APL pathogenesis and impact therapy, we used retroviral transduction to introduce an activated allele of FLT3 into control and MRP8 PML-RARA transgenic bone marrow. Activated FLT3 cooperated with PML-RAR alpha to induce leukemias in 62 to 299 days (median latency, 105 days). In contrast to the leukemias that arose spontaneously in MRP8 PML-RARA mice, the activated FLT3/PML-RAR alpha leukemias were characterized by leukocytosis, similar to human APL with FLT3 mutations. Cytogenetic analysis revealed clonal karyotypic abnormalities, which may contribute to pathogenesis or progression. SU11657, a selective, oral, multitargeted tyrosine kinase inhibitor that targets FLT3, cooperated with all-trans retinoic acid to rapidly cause regression of leukemia. Our results suggest that the acquisition of FLT3 mutations by cells with a pre-existing t(15;17) is a frequent pathway to the development of APL. Our findings also indicate that APL patients with FLT3 mutations may benefit from combination therapy with all-trans retinoic acid plus an FLT3 inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Enzyme Inhibitors/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Organic Chemicals/pharmacology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Tretinoin/pharmacology , Administration, Oral , Animals , Chromosome Aberrations , Clone Cells/ultrastructure , Drug Implants , Drug Screening Assays, Antitumor , Enzyme Inhibitors/administration & dosage , Humans , Leukemia, Promyelocytic, Acute/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Organic Chemicals/administration & dosage , Organic Chemicals/therapeutic use , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins/metabolism , Radiation Chimera , Receptor Protein-Tyrosine Kinases/metabolism , Tretinoin/administration & dosage , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...