Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 12982, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737414

ABSTRACT

Chordoma is a devastating rare cancer that affects one in a million people. With a mean-survival of just 6 years and no approved medicines, the primary treatments are surgery and radiation. In order to speed new medicines to chordoma patients, a drug repurposing strategy represents an attractive approach. Drugs that have already advanced through human clinical safety trials have the potential to be approved more quickly than de novo discovered medicines on new targets. We have taken two strategies to enable this: (1) generated and validated machine learning models of chordoma inhibition and screened compounds of interest in vitro. (2) Tested combinations of approved kinase inhibitors already being individually evaluated for chordoma. Several published studies of compounds screened against chordoma cell lines were used to generate Bayesian Machine learning models which were then used to score compounds selected from the NIH NCATS industry-provided assets. Out of these compounds, the mTOR inhibitor AZD2014, was the most potent against chordoma cell lines (IC50 0.35 µM U-CH1 and 0.61 µM U-CH2). Several studies have shown the importance of the mTOR signaling pathway in chordoma and suggest it as a promising avenue for targeted therapy. Additionally, two currently FDA approved drugs, afatinib and palbociclib (EGFR and CDK4/6 inhibitors, respectively) demonstrated synergy in vitro (CI50 = 0.43) while AZD2014 and afatanib also showed synergy (CI50 = 0.41) against a chordoma cell in vitro. These findings may be of interest clinically, and this in vitro- and in silico approach could also be applied to other rare cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chordoma/drug therapy , Drug Repositioning , Machine Learning , Antineoplastic Combined Chemotherapy Protocols/chemistry , Benzamides/agonists , Benzamides/pharmacology , Cell Line, Tumor , Chordoma/metabolism , Chordoma/pathology , Humans , Morpholines/agonists , Morpholines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Piperazines/agonists , Piperazines/pharmacology , Pyridines/agonists , Pyridines/pharmacology , Pyrimidines/agonists , Pyrimidines/pharmacology
2.
Braz. J. Pharm. Sci. (Online) ; 54(4): e00153, 2018. tab, graf
Article in English | LILACS | ID: biblio-1001583

ABSTRACT

Pyrimidine derivative 3 was afforded through the reaction of compound (1) with 5-ureidohydantion (2). Product 3 underwent a cyclization to produce fused pyrimidine derivative 7, although the latter product 7 was synthesized through one step via the reaction of compound (1) with 5-ureidohydantion (2) using another catalyst. Compound 3 was oriented to react with cyclic ketones 8a,b in the presence of elemental sulfur, salicylaldehyde (10), aryldiazonium chlorides 12a,b and ω-bromo-4-methoxy- acetophenone (14), which afforded, fused thiophene derivatives 9a,b, coumarin derivative 11, arylhdrazono derivatives 13a,b and 4-methoxyphenyl butenyl derivative 15, respectively. The latter product 15 was reacted with either potassium cyanide (16a) or potassium thiocyanide (16b) to form cyano and thiocyano derivatives 17a,b, respectively. Compound 17a underwent further cyclization to afford pyridopyrimidine derivative 19. Compound 15 was reacted with either hydrazine (20a) or phenylhydrazine (20b) to produce hydrazo derivatives 21a,b and these products were cyclize to produce pyrrole derivatives 23a,b. Finally, 5-ureidohydantion (2) was reacted with compounds 24a,b,c to afford pyrimidine derivatives 25a,b,c. The structures of the synthesized compounds were confirmed using IR, 1H NMR, 13C NMR and mass spectrometry techniques. Compounds 11 and 19 have promising as analgesic and antipyretic activities


Subject(s)
Pyridines/analysis , Pyrimidines/agonists , Pyrroles , Thiophenes/analysis , Coumarins/analysis , Antipyretics , Analgesics/classification
3.
Haematologica ; 102(9): 1587-1593, 2017 09.
Article in English | MEDLINE | ID: mdl-28619845

ABSTRACT

Fluorizoline is a new synthetic molecule that induces apoptosis by selectively targeting prohibitins. In the study herein, the pro-apoptotic effect of fluorizoline was assessed in 34 primary samples from patients with chronic lymphocytic leukemia. Fluorizoline induced apoptosis in chronic lymphocytic leukemia cells at concentrations in the low micromolar range. All primary samples were sensitive to fluorizoline irrespective of patients' clinical or genetic features, whereas normal T lymphocytes were less sensitive. Fluorizoline increased the protein levels of the pro-apoptotic B-cell lymphoma 2 family member NOXA in chronic lymphocytic leukemia cells. Furthermore, fluorizoline synergized with ibrutinib, 5-aminoimidazole-4-carboxamide riboside or venetoclax to induce apoptosis. These results suggest that targeting prohibitins could be a new therapeutic strategy for chronic lymphocytic leukemia.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hydrocarbons, Fluorinated/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Repressor Proteins/metabolism , Ribonucleosides/pharmacology , Sulfonamides/pharmacology , Thiazolidines/pharmacology , Up-Regulation/drug effects , Adenine/analogs & derivatives , Aminoimidazole Carboxamide/agonists , Aminoimidazole Carboxamide/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/agonists , Drug Synergism , Female , Humans , Hydrocarbons, Fluorinated/agonists , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Piperidines , Prohibitins , Pyrazoles/agonists , Pyrimidines/agonists , Ribonucleosides/agonists , Sulfonamides/agonists , Thiazolidines/agonists , Tumor Cells, Cultured
4.
Neuroscience ; 305: 279-92, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26254243

ABSTRACT

Amyloid beta (Aß) accumulation plays an important role in the pathogenesis of Alzheimer's disease (AD) by changing the neuronal excitability. However, the cellular mechanisms by which accumulation of Aß affects intrinsic neuronal properties are not well understood. The effect of bilateral intra-frontal cortex Aß (1-42) peptide injection on the intrinsic excitability of hippocampal CA1 pyramidal neurons with particular focus on the contribution of hyperpolarization-activated (Ih) channel currents was examined using whole-cell patch-clamp recording. Passive avoidance memory impairment and morphological changes in rats receiving intra-frontal Aß treatment were observed, which was associated with significant changes both in passive and active intrinsic electrical membrane properties of CA1 pyramidal neurons. Electrophysiological recording showed a significant decrease in neuronal excitability associated with an augmentation in the first spike after-hyperpolarization (AHP) amplitude. In addition, the depolarizing sag voltage was altered in neurons recorded from Aß-treated group. In voltage-clamp condition, a hyperpolarizing activated inward current sensitive to ZD7288 and capsaicin was significantly increased in neurons from Aß-treated rats. The Ih current density was increased and the activation curve was shifted toward less negative potential in the Aß-treated group as compared to control group. The enhancing effect of Aß treatment on Ih current was confirmed by showing upregulation of the mRNA of HCN1 channel in the CA1 pyramidal layer of hippocampi. These findings suggest the contribution of Ih and possibly TRPV1 channel currents to the changes induced by Aß treatment in the intrinsic membrane properties, which, in turn, may provide therapeutic targets for treatment of AD.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , CA1 Region, Hippocampal/pathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neurons/physiology , Alzheimer Disease/complications , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Disease Models, Animal , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/agonists , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , In Vitro Techniques , Male , Memory Disorders/etiology , Nerve Net/drug effects , Nerve Net/physiology , Neurons/drug effects , Patch-Clamp Techniques , Pyrimidines/agonists , Pyrimidines/pharmacology , Rats , Rats, Wistar , Sensory System Agents/pharmacology , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors
6.
J Clin Invest ; 124(12): 5490-502, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401474

ABSTRACT

Estrogen receptor-positive (ER(+)) breast cancers adapt to hormone deprivation and become resistant to antiestrogen therapy. Here, we performed deep sequencing on ER(+) tumors that remained highly proliferative after treatment with the aromatase inhibitor letrozole and identified a D189Y mutation in the inhibitory SH2 domain of the SRC family kinase (SFK) LYN. Evaluation of 463 breast tumors in The Cancer Genome Atlas revealed four LYN mutations, two of which affected the SH2 domain. In addition, LYN was upregulated in multiple ER(+) breast cancer lines resistant to long-term estrogen deprivation (LTED). An RNAi-based kinome screen revealed that LYN is required for growth of ER(+) LTED breast cancer cells. Kinase assays and immunoblot analyses of SRC substrates in transfected cells indicated that LYN(D189Y) has higher catalytic activity than WT protein. Further, LYN(D189Y) exhibited reduced phosphorylation at the inhibitory Y507 site compared with LYN(WT). Other SH2 domain LYN mutants, E159K and K209N, also exhibited higher catalytic activity and reduced inhibitory site phosphorylation. LYN(D189Y) overexpression abrogated growth inhibition by fulvestrant and/or the PI3K inhibitor BKM120 in 3 ER(+) breast cancer cell lines. The SFK inhibitor dasatinib enhanced the antitumor effect of BKM120 and fulvestrant against estrogen-deprived ER(+) xenografts but not LYN(D189Y)-expressing xenografts. These results suggest that LYN mutations mediate escape from antiestrogens in a subset of ER(+) breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Estrogen Receptor Modulators/pharmacology , Mutation, Missense , Receptors, Estrogen/metabolism , src-Family Kinases/metabolism , Amino Acid Substitution , Aminopyridines/agonists , Aminopyridines/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Dasatinib , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Morpholines/agonists , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/agonists , Pyrimidines/pharmacology , Receptors, Estrogen/genetics , Thiazoles/agonists , Thiazoles/pharmacology , Xenograft Model Antitumor Assays , src Homology Domains , src-Family Kinases/genetics
7.
J Dairy Sci ; 97(1): 345-9, 2014.
Article in English | MEDLINE | ID: mdl-24210485

ABSTRACT

Recent studies in rodents demonstrated that peroxisome proliferator-activated receptor α (PPARα), a central regulator of energy homeostasis, is an important transcriptional regulator of the gene encoding the carnitine transporter novel organic cation transporter 2 (OCTN2). Less is known with regard to the regulation of OCTN2 by PPARα and its role for carnitine transport in cattle, even though PPARα activation physiologically occurs in the liver of high-producing cows during early lactation. To explore the role of PPARα for OCTN2 expression and carnitine transport in cattle, we studied the effect of the PPARα activator WY-14,643 on the expression of OCTN2 in the presence and absence of PPARα antagonists and on OCTN2-mediated carnitine transport in the Madin-Darby bovine kidney (MDBK) cell line. The results show that WY-14,643 increases mRNA and protein levels of OCTN2, whereas co-treatment of MDBK cells with WY-14,643 and the PPARα antagonist GW6471 blocks the WY-14,643-induced increase in mRNA and protein levels of OCTN2 in bovine cells. In addition, treatment of MDBK cells with WY-14,643 stimulates specifically Na(+)-dependent carnitine uptake in MDBK cells, which is likely the consequence of the increased carnitine transport capacity of cells due to the elevated expression of OCTN2. In conclusion, our results indicate that OCTN2 expression and carnitine transport in cattle, as in rodents, are regulated by PPARα.


Subject(s)
Carnitine/pharmacokinetics , Kidney/cytology , Kidney/drug effects , Organic Cation Transport Proteins/metabolism , PPAR alpha/agonists , Pyrimidines/pharmacology , Animals , Cattle , Cell Line , Female , Gene Expression Regulation , Homeostasis , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Organic Cation Transport Proteins/genetics , Pyrimidines/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vitamin B Complex/pharmacokinetics
8.
Acta Neuropathol ; 125(6): 795-813, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23604588

ABSTRACT

In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and prion diseases, deposits of aggregated disease-specific proteins are found. Oligomeric aggregates are presumed to be the key neurotoxic agent. Here we describe the novel oligomer modulator anle138b [3-(1,3-benzodioxol-5-yl)-5-(3-bromophenyl)-1H-pyrazole], an aggregation inhibitor we developed based on a systematic high-throughput screening campaign combined with medicinal chemistry optimization. In vitro, anle138b blocked the formation of pathological aggregates of prion protein (PrP(Sc)) and of α-synuclein (α-syn), which is deposited in PD and other synucleinopathies such as dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Notably, anle138b strongly inhibited all prion strains tested including BSE-derived and human prions. Anle138b showed structure-dependent binding to pathological aggregates and strongly inhibited formation of pathological oligomers in vitro and in vivo both for prion protein and α-synuclein. Both in mouse models of prion disease and in three different PD mouse models, anle138b strongly inhibited oligomer accumulation, neuronal degeneration, and disease progression in vivo. Anle138b had no detectable toxicity at therapeutic doses and an excellent oral bioavailability and blood-brain-barrier penetration. Our findings indicate that oligomer modulators provide a new approach for disease-modifying therapy in these diseases, for which only symptomatic treatment is available so far. Moreover, our findings suggest that pathological oligomers in neurodegenerative diseases share structural features, although the main protein component is disease-specific, indicating that compounds such as anle138b that modulate oligomer formation by targeting structure-dependent epitopes can have a broad spectrum of activity in the treatment of different protein aggregation diseases.


Subject(s)
Brain/drug effects , Parkinson Disease/therapy , Prion Diseases/therapy , Prions/drug effects , Pyrazoles/agonists , Pyrimidines/agonists , Animals , Brain/metabolism , Brain/pathology , Cell Culture Techniques , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Parkinson Disease/etiology , Parkinson Disease/metabolism , Prion Diseases/etiology , Prion Diseases/metabolism , Prions/metabolism , Rotenone/pharmacology , alpha-Synuclein/pharmacology
9.
PLoS One ; 8(3): e57523, 2013.
Article in English | MEDLINE | ID: mdl-23520471

ABSTRACT

Sarcomas are rare and heterogeneous mesenchymal tumors affecting both pediatric and adult populations with more than 70 recognized histologies. Doxorubicin and ifosfamide have been the main course of therapy for treatment of sarcomas; however, the response rate to these therapies is about 10-20% in metastatic setting. Toxicity with the drug combination is high, response rates remain low, and improvement in overall survival, especially in the metastatic disease, remains negligible and new agents are needed. Wee1 is a critical component of the G2/M cell cycle checkpoint control and mediates cell cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by MK1775 has been reported to enhance the cytotoxic effect of DNA damaging agents in different types of carcinomas. In this study we investigated the therapeutic efficacy of MK1775 in various sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo both alone and in combination with gemcitabine, which is frequently used in the treatment of sarcomas. Our data demonstrate that MK1775 treatment as a single agent at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. Additionally, MK1775 significantly enhances the cytotoxic effect of gemcitabine in sarcoma cells lines with different p53 mutational status. In patient-derived bone and soft tissue sarcoma samples we showed that MK1775 alone and in combination with gemcitabine causes significant apoptotic cell death. Magnetic resonance imaging (MRI) and histopathologic studies showed that MK1775 induces significant cell death and terminal differentiation in a patient-derived xenograft mouse model of osteosarcoma in vivo. Our results together with the high safety profile of MK1775 strongly suggest that this drug can be used as a potential therapeutic agent in the treatment of both adult as well as pediatric sarcoma patients.


Subject(s)
Antimetabolites, Antineoplastic , Cell Cycle Proteins/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Femoral Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Osteosarcoma/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles , Pyrimidines , Adolescent , Adult , Animals , Antimetabolites, Antineoplastic/agonists , Antimetabolites, Antineoplastic/pharmacology , Cell Death , Cell Differentiation , Cell Line, Tumor , Child , Child, Preschool , Deoxycytidine/agonists , Deoxycytidine/pharmacology , Drug Synergism , Female , Femoral Neoplasms/pathology , Humans , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Osteosarcoma/pathology , Pyrazoles/agonists , Pyrazoles/pharmacology , Pyrimidines/agonists , Pyrimidines/pharmacology , Pyrimidinones , Transplantation, Heterologous , Xenograft Model Antitumor Assays , Gemcitabine
10.
Br J Haematol ; 153(2): 199-211, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21352196

ABSTRACT

As antigenic stimulation of the B cell antigen receptor (BCR) is key to chronic lymphocytic leukaemia (CLL) pathogenesis, targeting dysregulated kinases involved in BCR signalling is an attractive therapeutic approach. We studied the effects of the Src/c-Abl tyrosine kinase inhibitor dasatinib on BCR signal transduction in CLL cells. Treatment of CLL cells with 100 nmol/l dasatinib induced apoptosis by an average reduction in viability of 33·7% at 48 h, with dasatinib sensitivity correlating with inhibition of Syk(Y348) phosphorylation. Dasatinib inhibited calcium flux, phosphatidylinositol-3-kinase and mitogen-activated protein kinase activation following BCR crosslinking, and blocked the Mcl-1-dependent increase in CLL cell survival on prolonged BCR stimulation. However, the pro-apoptotic effect of dasatinib was abrogated by stromal cell contact alone or in the presence of CD154 and interleukin (IL)-4 (CD154L/IL-4 system). Whilst dasatinib retained the ability to sensitize CLL cells in stromal co-culture to both fludarabine and chlorambucil, the addition of CD154 and IL-4 rendered cells resistant to these drug combinations. We demonstrate that the HSP90 inhibitor 17-DMAG exhibited synergy with dasatinib in vitro, and moreover, induced apoptosis of CLL cells in the CD154L/IL-4 system. Our data provide evidence that dasatinib would be most clinically effective in combination with agents able to target antigen-independent microenvironmental signals.


Subject(s)
B-Lymphocytes/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , Thiazoles/pharmacology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , B-Lymphocytes/pathology , Benzoquinones/agonists , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , CD40 Ligand/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dasatinib , Dose-Response Relationship, Drug , Drug Synergism , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Interleukin-4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lactams, Macrocyclic/agonists , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/agonists , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines/agonists , Pyrimidines/therapeutic use , Stromal Cells/metabolism , Stromal Cells/pathology , Syk Kinase , Thiazoles/agonists , Thiazoles/therapeutic use , Time Factors
11.
Mol Imaging Biol ; 11(6): 386-98, 2009.
Article in English | MEDLINE | ID: mdl-19330384

ABSTRACT

BACKGROUND: Activation of microglia cells plays an important role in neurological diseases. Positron emission tomography (PET) with [(11)C]-(R)-PK11195 has already been used to visualize activated microglia cells in neurological diseases. However, [(11)C]-(R)-PK11195 may not possess the required sensitivity to visualize mild neuroinflammation. In this study, we evaluated the PET tracers [(11)C]-DPA-713 and [(18)F]-DPA-714 as agents for imaging of activated microglia in a rat model of herpes encephalitis. MATERIALS AND METHODS: Rats were intranasally inoculated with HSV-1. On day 6 or 7 after inoculation, small animal PET studies were performed to compare [(11)C]-(R)-PK11195, [(11)C]-DPA-713, and [(18)F]-DPA-714. RESULTS: Uptake of [(11)C]-DPA-713 in infected brain areas was comparable to that of [(11)C]-(R)-PK11195, but [(11)C]-DPA-713 showed lower non-specific binding. Non-specific uptake of [(18)F]-DPA-714 was lower than that of [(11)C]-(R)-PK11195. In the infected brain, total [(18)F]-DPA-714 uptake was lower than that of [(11)C]-(R)-PK11195, with comparable specific uptake. CONCLUSIONS: [(11)C]-DPA-713 may be more suitable for visualizing mild inflammation than [(11)C]-(R)-PK11195. In addition, the fact that [(18)F]-DPA-714 is an agonist PET tracer opens new possibilities to evaluate different aspects of neuroinflammation. Therefore, both tracers warrant further investigation in animal models and in a clinical setting.


Subject(s)
Acetamides/metabolism , Carrier Proteins/metabolism , Encephalitis, Herpes Simplex/diagnostic imaging , Isoquinolines/metabolism , Pyrazoles/agonists , Pyrazoles/metabolism , Pyrimidines/agonists , Pyrimidines/metabolism , Receptors, GABA-A/metabolism , Acetamides/chemistry , Animals , Animals, Outbred Strains , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/metabolism , Disease Models, Animal , Encephalitis, Herpes Simplex/metabolism , Fluorine Radioisotopes/metabolism , Immunohistochemistry , Isoquinolines/chemistry , Male , Molecular Structure , Positron-Emission Tomography/methods , Pyrazoles/chemistry , Pyrimidines/chemistry , Radioligand Assay , Radiopharmaceuticals/metabolism , Rats , Rats, Wistar
12.
Toxicol In Vitro ; 21(8): 1678-85, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17604596

ABSTRACT

The effect of docosahexaenoic acid (DHA) on the killing efficacy of imatinib on HL-60 cells expressing the Bcr-Abl protein was investigated. Imatinib is an Abl tyrosine kinase inhibitor used in the treatment of patients with chronic myeloid leukemia. The pre-treatment with DHA for 24 h raised the effect of imatinib at 100 microM concentration only. On the other hand, after 72 h pre-treatment, all concentrations of DHA tested (25, 50 and 100 microM) enhanced the toxic effect of imatinib. These results indicate that long-term pre-treatment with DHA makes Bcr-Abl HL-60 cells more susceptible to the toxic effect of imatinib.


Subject(s)
Antineoplastic Agents/toxicity , Docosahexaenoic Acids/pharmacology , Genes, abl/genetics , Piperazines/agonists , Piperazines/toxicity , Pyrimidines/agonists , Pyrimidines/toxicity , Benzamides , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation , HL-60 Cells , Humans , Imatinib Mesylate
14.
Blood ; 109(9): 4016-9, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17213283

ABSTRACT

Chronic myeloid leukemia (CML) stem and progenitor cells overexpress BcrAbl and are insensitive to imatinib mesylate (IM). We therefore investigated whether these cells were efficiently targeted by nilotinib. In K562, the inhibitory concentration (IC50) of nilotinib was 30 nM versus 600 nM for IM, consistent with its reported 20-fold-higher potency. However, in primary CD34(+) CML cells, nilotinib and IM were equipotent for inhibition of BcrAbl activity, producing equivalent but incomplete reduction in CrkL phosphorylation at 5 microM. CML CD34(+) cells were still able to expand over 72 hours with 5 microM of either drug, although there was a concentration-dependent restriction of amplification. As for IM, the most primitive cells (CFSE(max)) persisted and accumulated over 72 hours with nilotinib and remained caspase-3 negative. Furthermore, nilotinib with IM led to further accumulation of this population, suggesting at least additive antiproliferative effects. These results confirmed that, like IM, the predominant effect of nilotinib is antiproliferative rather than proapoptotic.


Subject(s)
Antigens, CD34 , Apoptosis/drug effects , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Neoplastic Stem Cells/enzymology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Tumor Stem Cell Assay , Benzamides , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/agonists , Protein Kinase Inhibitors/agonists , Pyrimidines/agonists , Time Factors
15.
Blood ; 109(9): 4006-15, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17218385

ABSTRACT

Interactions between MEK1/2 inhibitors and the dual Abl/Src kinase inhibitor dasatinib (BMS-354825) were examined in chronic myeloid leukemia (CML) cell lines and primary specimens. Cotreatment of K562 or LAMA cells with subtoxic or marginally toxic concentrations of PD184352 (or U0126) and dasatinib synergistically potentiated mitochondrial damage, caspase activation, and apoptosis. Similar interactions were observed in CD34(+) cells from one CML patient-derived but not in a normal human CD34(+) bone marrow cell specimen. These interactions were associated with multiple perturbations in survival signaling pathways, including inactivation of Bcr/Abl, STAT5, and ERK1/2; down-regulation of Bcl-x(L) and Mcl-1; and dephosphorylation/activation of Bim. They were also associated with BAX/BAK conformational change, mitochondrial dysfunction, and caspase activation. Bim knockdown by shRNA suppressed BAX and BAK conformational change and protected cells from dasatinib/PD184352 lethality. Conversely, K562 cells ectopically expressing Mcl-1 or Bcl-x(L) were significantly less susceptible to dasatinib/PD184352 toxicity. Notably, the dasatinib/PD184352 regimen was active against leukemic cells exhibiting various forms of imatinib mesylate resistance, including Bcr/Abl overexpression, Lyn activation, and several Bcr/Abl kinase domain mutations (eg, E255K, M351T), but not T315I. Together, these findings suggest that strategies combining dasatanib with MEK1/2 inhibitors warrant further investigation in Bcr/Abl(+) malignancies, particularly in the setting of imatinib mesylate-resistant disease.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Benzamides/agonists , Benzamides/pharmacology , Butadienes/agonists , Butadienes/pharmacology , Dasatinib , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Drug Synergism , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Kinase 2/pharmacology , Nitriles/agonists , Nitriles/pharmacology , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/agonists , Pyrimidines/agonists , Pyrimidines/therapeutic use , Thiazoles/agonists , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
16.
Clin Cancer Res ; 8(9): 2976-84, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231544

ABSTRACT

PURPOSE: The goal of this study was to characterize interactions between the Bcr/Abl kinase inhibitor STI571 and the cyclin-dependent kinase inhibitor flavopiridol in Bcr/Abl(+) human leukemia cells. EXPERIMENTAL DESIGN: K562 leukemia cells were exposed to STI571 +/- flavopiridol for 24 or 48 h, after which mitochondrial damage, caspase activation, expression/activation of signaling and cell cycle regulatory proteins, and apoptosis were assessed. RESULTS: In K562 cells, coadministration of marginally toxic concentrations of STI571 (200 nM) and flavopiridol (150 nM) for 48 h resulted in a marked increase in mitochondrial damage (e.g., cytochrome c release), activation of caspase-3, caspase-8, and Bid, and apoptosis. Similar interactions were observed in Bcr/Abl(+) LAMA-84 cells but not in leukemic cells that fail to express Bcr/Abl (e.g., HL-60, U937, Jurkat). STI571/flavopiridol-mediated apoptosis was associated with the caspase-independent down-regulation of Bcl-x(L) and Mcl-1, activation of extracellular signal-regulated kinase and c-Jun NH(2)-terminal kinase, and the caspase-dependent release of Smac/DIABLO and loss of deltapsi(m). Coadministration of flavopiridol and STI571 did not result in changes in levels of expression of Bcl-2, phopho-Stat5, phospho-p34(cdc2), or Bcr/Abl. Finally, STI571/flavopiridol effectively induced apoptosis in STI571-resistant K562 cells displaying amplification of the Bcr/Abl protein. CONCLUSIONS: Together, these findings indicate that the cyclin-dependent kinase inhibitor flavopiridol induces multiple perturbations in signaling pathways in STI571-treated Bcr/Abl(+) human leukemia cells that culminate in mitochondrial injury, caspase activation, and apoptosis. They also suggest that simultaneous disruption of survival signaling and cell cycle regulatory pathways may represent an effective strategy in Bcr/Abl(+) malignancies.


Subject(s)
Biomarkers, Tumor/analysis , Flavonoids/pharmacology , Fusion Proteins, bcr-abl/analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mitochondria/drug effects , Piperazines/agonists , Piperidines/pharmacology , Pyrimidines/agonists , Apoptosis/drug effects , Benzamides , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , HL-60 Cells/drug effects , Humans , Imatinib Mesylate , Jurkat Cells/drug effects , K562 Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Phosphorylation , Piperazines/pharmacology , Protein Processing, Post-Translational/drug effects , Pyrimidines/pharmacology , Signal Transduction/drug effects , U937 Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...