Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Chemother Pharmacol ; 81(2): 291-304, 2018 02.
Article in English | MEDLINE | ID: mdl-29204687

ABSTRACT

PURPOSE: Zoptarelin doxorubicin is a fusion molecule of the chemotherapeutic doxorubicin and a luteinizing hormone-releasing hormone receptor (LHRHR) agonist, designed for drug targeting to LHRHR positive tumors. The aim of this study was to establish a physiologically based pharmacokinetic (PBPK) parent-metabolite model of zoptarelin doxorubicin and to apply it for drug-drug interaction (DDI) potential analysis. METHODS: The PBPK model was built in a two-step procedure. First, a model for doxorubicin was developed, using clinical data of a doxorubicin study arm. Second, a parent-metabolite model for zoptarelin doxorubicin was built, using clinical data of three different zoptarelin doxorubicin studies with a dosing range of 10-267 mg/m2, integrating the established doxorubicin model. DDI parameters determined in vitro were implemented to predict the impact of zoptarelin doxorubicin on possible victim drugs. RESULTS: In vitro, zoptarelin doxorubicin inhibits the drug transporters organic anion-transporting polypeptide 1B3 (OATP1B3) and organic cation transporter 2 (OCT2). The model was applied to evaluate the in vivo inhibition of these transporters in a generic manner, predicting worst-case scenario decreases of 0.5% for OATP1B3 and of 2.5% for OCT2 transport rates. Specific DDI simulations using PBPK models of simvastatin (OATP1B3 substrate) and metformin (OCT2 substrate) predict no significant changes of the plasma concentrations of these two victim drugs during co-administration. CONCLUSIONS: The first whole-body PBPK model of zoptarelin doxorubicin and its active metabolite doxorubicin has been successfully established. Zoptarelin doxorubicin shows no potential for DDIs via OATP1B3 and OCT2.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Doxorubicin/analogs & derivatives , Gonadotropin-Releasing Hormone/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Biotransformation , Computer Simulation , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Drug Interactions , Female , Gonadotropin-Releasing Hormone/adverse effects , Gonadotropin-Releasing Hormone/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Male , Metformin/pharmacokinetics , Middle Aged , Models, Biological , Octamer Transcription Factor-2 , Simvastatin/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism
2.
J Hematol Oncol ; 10(1): 9, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28061880

ABSTRACT

BACKGROUND: There is increasing evidence of a constitutive activation of Akt in pancreatic ductal adenocarcinoma (PDAC), associated with poor prognosis and chemoresistance. Therefore, we evaluated the expression of phospho-Akt in PDAC tissues and cells, and investigated molecular mechanisms influencing the therapeutic potential of Akt inhibition in combination with gemcitabine. METHODS: Phospho-Akt expression was evaluated by immunohistochemistry in tissue microarrays (TMAs) with specimens tissue from radically-resected patients (n = 100). Data were analyzed by Fisher and log-rank test. In vitro studies were performed in 14 PDAC cells, including seven primary cultures, characterized for their Akt1 mRNA and phospho-Akt/Akt levels by quantitative-RT-PCR and immunocytochemistry. Growth inhibitory effects of Akt inhibitors and gemcitabine were evaluated by SRB assay, whereas modulation of Akt and phospho-Akt was investigated by Western blotting and ELISA. Cell cycle perturbation, apoptosis-induction, and anti-migratory behaviors were studied by flow cytometry, AnnexinV, membrane potential, and migration assay, while pharmacological interaction with gemcitabine was determined with combination index (CI) method. RESULTS: Immunohistochemistry of TMAs revealed a correlation between phospho-Akt expression and worse outcome, particularly in patients with the highest phospho-Akt levels, who had significantly shorter overall and progression-free-survival. Similar expression levels were detected in LPC028 primary cells, while LPC006 were characterized by low phospho-Akt. Remarkably, Akt inhibitors reduced cancer cell growth in monolayers and spheroids and synergistically enhanced the antiproliferative activity of gemcitabine in LPC028, while this combination was antagonistic in LPC006 cells. The synergistic effect was paralleled by a reduced expression of ribonucleotide reductase, potentially facilitating gemcitabine cytotoxicity. Inhibition of Akt decreased cell migration and invasion, which was additionally reduced by the combination with gemcitabine. This combination significantly increased apoptosis, associated with induction of caspase-3/6/8/9, PARP and BAD, and inhibition of Bcl-2 and NF-kB in LPC028, but not in LPC006 cells. However, targeting the key glucose transporter Glut1 resulted in similar apoptosis induction in LPC006 cells. CONCLUSIONS: These data support the analysis of phospho-Akt expression as both a prognostic and a predictive biomarker, for the rational development of new combination therapies targeting the Akt pathway in PDAC. Finally, inhibition of Glut1 might overcome resistance to these therapies and warrants further studies.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/analysis , Aged , Apoptosis/drug effects , Biopsy , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle/drug effects , Cell Movement/drug effects , Deoxycytidine/therapeutic use , Drug Synergism , Female , Glucose Transporter Type 1/drug effects , Humans , Male , Middle Aged , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , Phosphoproteins/analysis , Phosphoproteins/antagonists & inhibitors , Prognosis , RNA, Messenger/analysis , Spheroids, Cellular , Tumor Cells, Cultured , Gemcitabine
3.
Bioorg Med Chem Lett ; 26(10): 2408-2412, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27072910

ABSTRACT

Introducing a second chiral center on our previously described 1,2,4-triazole, allowed us to increase diversity and elongate the 'C-terminal part' of the molecule. Therefore, we were able to explore mimics of the substance P analogs described as inverse agonists. Some compounds presented affinities in the nanomolar range and potent biological activities, while one exhibited a partial inverse agonist behavior similar to a Substance P analog.


Subject(s)
Receptors, Ghrelin/metabolism , Triazoles/chemistry , Fluorescence Resonance Energy Transfer , Indoles/chemistry , Indoles/pharmacology , Inhibitory Concentration 50 , Ligands , Receptors, Ghrelin/agonists , Structure-Activity Relationship , Substance P/chemistry , Tryptophan/analogs & derivatives , Tryptophan/chemistry , Tryptophan/pharmacology
4.
Bioorg Med Chem Lett ; 25(1): 20-4, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25435152

ABSTRACT

Ghrelin receptor ligands based on a trisubstituted 1,2,4-triazole scaffold were recently synthesized and evaluated for their in vitro affinity for the GHS-R1a receptor and their biological activity. In this study, replacement of the α-aminoisobutyryl (Aib) moiety (a common feature present in numerous growth hormone secretagogues described in the literature) by aromatic and heteroaromatic groups was explored. We found potent antagonists incorporating the picolinic moiety in place of the Aib moiety. In an attempt to increase affinity and activity of our lead compound 2, we explored the modulation of the pyridine ring. Herein we report the design and the structure-activity relationships study of these new ghrelin receptor ligands.


Subject(s)
Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/metabolism , Triazoles/chemical synthesis , Triazoles/metabolism , Animals , Cell Line , Humans , Mice , Protein Binding/physiology , Structure-Activity Relationship , Triazoles/pharmacology
5.
Bioorg Med Chem Lett ; 24(16): 3748-52, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25022204

ABSTRACT

A novel series of (7-aryl-1,5-naphthyridin-2-yl)ureas was discovered as dual ERK2 and Aurora B kinases inhibitors. Several analogues were active at micromolar and submicromolar range against ERK2 and Aurora B, associated with very promising antiproliferative activity toward various cancer cell lines. Synthesis, structure activity relationship and docking study are reported. In vitro ADME properties and safety data are also discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase B/antagonists & inhibitors , Drug Discovery , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Urea/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aurora Kinase B/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
6.
Anticancer Agents Med Chem ; 14(4): 629-35, 2014 May.
Article in English | MEDLINE | ID: mdl-24628236

ABSTRACT

Perifosine treatment exhibits a complex molecular response including the inhibition of Akt or the induction of apoptosis via clustering of death receptors in lipid rafts. However, the molecular response can vary between different tumor entities and the contribution of each target pathway to the activity of Perifosine might be distinct depending on the tumor entity or the agent combined with Perifosine. In this review we discuss the current view on the mechanism of action of perifosine in cancer and the contribution of the molecular targets of Perifosine to its activity.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Phosphorylcholine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Lipid Metabolism/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Death Domain/metabolism , Signal Transduction
7.
ChemMedChem ; 9(1): 217-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24273104

ABSTRACT

As part of our research projects to identify new chemical entities of biological interest, we developed a synthetic approach and the biological evaluation of (7-aryl-1,5-naphthyridin-4-yl)ureas as a novel class of Aurora kinase inhibitors for the treatment of malignant diseases based on pathological cell proliferation. 1,5-Naphthyridine derivatives showed excellent inhibitory activities toward Aurora kinases A and B, and the most active compound, 1-cyclopropyl-3-[7-(1-methyl-1H-pyrazol-4-yl)-1,5-naphthyridin-4-yl]urea (49), displayed IC50 values of 13 and 107 nM against Aurora kinases A and B, respectively. In addition, the selectivity toward a panel of seven cancer-related protein kinases was highlighted. In vitro ADME properties were also determined in order to rationalize the difficulties in correlating antiproliferative activity with Aurora kinase inhibition. Finally, the good safety profile of these compounds imparts promising potential for their further development as anticancer agents.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Protein Kinase Inhibitors/analogs & derivatives , Urea/analogs & derivatives , Animals , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , HCT116 Cells , Half-Life , Humans , Mice , Microsomes, Liver/metabolism , Naphthyridines/chemistry , Protein Binding , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Urea/pharmacokinetics , Urea/pharmacology
8.
Bioorg Med Chem Lett ; 23(19): 5264-6, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23988351

ABSTRACT

A new series of substituted tri-/tetraazabenzo[3,2-a]fluorene-5,6-diones and their corresponding oxime derivatives have been synthesized and spectroscopically characterized. The antiproliferative activities of all compounds were evaluated on at least three different cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Aza Compounds/chemical synthesis , Fluorenes/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aza Compounds/chemistry , Benzene Derivatives/chemical synthesis , Benzene Derivatives/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Molecular Structure
9.
Bioorg Med Chem Lett ; 23(6): 1846-52, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23395656

ABSTRACT

A series of indazolo[4,3-gh]isoquinolinones derivatives have been synthesized to decrease cardiotoxic side effects in comparison to Mitoxantrone. The antiproliferative effects of different side chains were investigated and tested on at least four different cell lines of cervix, ovarian, CNS, NSCLC (non-small-cell lung cancer) and colon carcinoma. In addition to antiproliferative activities, influence on cell cycle and intercalation behavior have been tested.


Subject(s)
Antineoplastic Agents/chemical synthesis , Indazoles/chemistry , Quinolones/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , DNA/metabolism , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Mitoxantrone/chemistry , Mitoxantrone/toxicity , Quinolones/chemical synthesis , Quinolones/toxicity , S Phase Cell Cycle Checkpoints/drug effects , Structure-Activity Relationship
10.
Invest New Drugs ; 30(4): 1396-403, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21750922

ABSTRACT

The novel AKT inhibitor perifosine possesses myelopoiesis-stimulating effects in rodents. We studied the in vitro effects of the novel agents perifosine, bortezomib and lenalidomide in addition to adriamycin against normal human hematopoietic progenitor cells (HPC) using different clonogenic and non-clonogenic assays. All agents inhibited colony-forming unit (CFU) formation, perifosine inhibiting mainly CFU-granulocyte/macrophage formation and the other agents burst-forming unit-erythroid formation. Perifosine combined with lenalidomide or adriamycin tended to act antagonistically in suppressing CFU formation. Despite their inhibition of CFU formation, perifosine, bortezomib and lenalidomide induced only slight or moderate cytotoxicity in CD34(+) selected HPC, as assessed using different assays such as flow cytometry-based detection of activated caspases and immunohistochemistry studies (e.g., Ki-67 staining). In contrast to its myelopoiesis-stimulating effects in rodents, perifosine--like bortezomib and lenalidomide--suppresses the clonogenic potential of HPC from healthy donors in vitro and thus probably plays no role in preventing neutropenia or in shorting its duration after intensive chemotherapy. However, all these novel agents typically induce only slight or moderate suppression of the clonogenic potential or loss of viability of normal HPC at clinically achievable plasma concentrations, assuming that hematoxicity is manageable and functional HPC can be collected after treatment with these compounds.


Subject(s)
Boronic Acids/pharmacology , Health , Hematopoietic Stem Cells/drug effects , Phosphorylcholine/analogs & derivatives , Pyrazines/pharmacology , Thalidomide/analogs & derivatives , Tissue Donors , Annexin A5/metabolism , Antigens, CD34/metabolism , Bortezomib , Caspases/metabolism , Colony-Forming Units Assay , Doxorubicin/pharmacology , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Humans , Immunohistochemistry , Lenalidomide , Phosphorylcholine/pharmacology , Staining and Labeling , Thalidomide/pharmacology , Trypan Blue/metabolism
11.
Invest New Drugs ; 30(2): 480-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21080211

ABSTRACT

The novel AKT inhibitor perifosine, a synthetic alkylphospholipid, is currently being investigated in clinical trials for the treatment of different hematological and oncological malignancies. The in vitro cytotoxicity of perifosine, bortezomib and lenalidomide against 6 cell lines derived from hematological malignancies was investigated using trypan blue staining, flow cytometry-based detection of activated caspases, Annexin V assays, immunohistochemistry studies (KI-67 and caspase-3 staining) and the immature-myeloid-information (IMI) technique. Perifosine and bortezomib induced concentration- and time-dependent cytotoxicity in all cell lines tested. Perifosine together with bortezomib largely exerted additive or synergistic effects with combination indices ranging from 1.13 to 0.22 for combined efficacies of 25% to 75% after 24-hour incubation. Lenalidomide-triggered cytotoxicity was low in all cell lines tested with any assay (less than 10% compared to the negative control). Finally, perifosine, but not bortezomib or lenalidomide, significantly increased the number of cells detected in the IMI channel. Perifosine and bortezomib- but not lenalidomide- trigger substantial cytotoxicity by caspase activation and mainly act additively or synergistically. The IMI technique might be a useful tool for studying cytotoxicity of agents like perifosine that interact mainly with the cellular membrane.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/pharmacology , Lymphoma/pathology , Multiple Myeloma/pathology , Phosphorylcholine/analogs & derivatives , Pyrazines/pharmacology , Thalidomide/analogs & derivatives , Apoptosis/drug effects , Bortezomib , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Flow Cytometry , HL-60 Cells , Humans , Immunohistochemistry , Inhibitory Concentration 50 , K562 Cells , Ki-67 Antigen/metabolism , Lenalidomide , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Lymphoma/metabolism , Multiple Myeloma/metabolism , Phosphorylcholine/pharmacology , Thalidomide/pharmacology , Time Factors
12.
J Med Chem ; 54(12): 4247-63, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21563750

ABSTRACT

A total of 53 N-benzoylated phenoxazines and phenothiazines, including their S-oxidized analogues, were synthesized and evaluated for antiproliferative activity, interaction with tubulin, and cell cycle effects. Potent inhibitors of multiple cancer cell lines emerged with the 10-(4-methoxybenzoyl)-10H-phenoxazine-3-carbonitrile (33b, IC(50) values in the range of 2-15 nM) and the isovanillic analogue 33c. Seventeen compounds strongly inhibited tubulin polymerization with activities higher than or comparable to those of the reference compounds such as colchicine. Concentration-dependent flow cytometric studies revealed that inhibition of K562 cell growth was associated with an arrest in the G2/M phases of the cell cycle, indicative of mitotic blockade. Structure-activity relationship studies showed that best potencies were obtained with agents bearing a methoxy group placed para at the terminal phenyl ring and a 3-cyano group in the phenoxazine. A series of analogues highlight not only the phenoxazine but also the phenothiazine structural scaffold as valuable pharmacophores for potent tubulin polymerization inhibitors, worthy of further investigation.


Subject(s)
Antineoplastic Agents/chemical synthesis , Oxazines/chemical synthesis , Phenothiazines/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biopolymers , Cell Cycle/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Organ Specificity , Oxazines/chemistry , Oxazines/pharmacology , Phenothiazines/chemistry , Phenothiazines/pharmacology , Structure-Activity Relationship , Tubulin/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
13.
Bioorg Med Chem Lett ; 21(10): 3117-21, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21458262

ABSTRACT

A series of 6-azanaphthoquinone pyrrolo-annelated derivatives carrying different basic side chains have been synthesized. The antiproliferative activities of all compounds were evaluated on at least four different cell lines with Mitoxantrone as reference compound. Cytotoxic effects and DNA intercalation behavior were investigated.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Naphthoquinones/chemistry , Pyrroles/chemistry , Structure-Activity Relationship
15.
Cancer Res ; 69(16): 6473-81, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19638591

ABSTRACT

Recently, we could show that gonadotropin-releasing hormone (GnRH)-II antagonists induce apoptosis in human endometrial, ovarian, and breast cancer cells in vitro and in vivo. In the present study, we have ascertained receptor binding and effects of GnRH-II antagonists on mitogenic signal transduction and on activation of proapoptotic protein Bax. The GnRH-II antagonists tested showed EC50 values for GnRH-I receptor binding in the range of 1 to 2 nmol/L. The GnRH-II agonist [D-Lys6]GnRH-II showed an EC50 value for GnRH-I receptor binding of approximately 1,000 nmol/L. Agonistic activity on GnRH-I receptor function with an EC50 of 13 nmol/L has been determined for [D-Lys6]GnRH-II. Antagonistic activities with EC50 values in the range of 1 nmol/L were determined for the GnRH-II antagonists. Treatment of human endometrial, ovarian, and breast cancer cells with GnRH-II antagonists resulted in time-dependent activation of stress-induced mitogen-activated protein kinases p38 and c-Jun NH2-terminal kinase. In addition, treatment with GnRH-II antagonists induced time-dependent activation of proapoptotic protein Bax. GnRH-II antagonists are not involved in activation of protein kinase B/Akt or extracellular signal-regulated kinase 1/2. The GnRH-II antagonists tested had similar binding affinities to the GnRH-I receptor comparable with that of GnRH-I antagonist Cetrorelix. Referring to the cyclic AMP response element reporter gene activation assay, the GnRH-II agonist [D-Lys6]GnRH-II has to be classified as an agonist at the GnRH-I receptor, whereas the GnRH-II antagonists tested are clear antagonists at the GnRH-I receptor. GnRH-II antagonists induce apoptotic cell death in human endometrial, ovarian, and breast cancer cells via activation of stress-induced mitogen-activated protein kinases p38 and c-Jun NH2-terminal kinase followed by activation of proapoptotic protein Bax.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Endometrial Neoplasms/pathology , Gonadotropin-Releasing Hormone/analogs & derivatives , JNK Mitogen-Activated Protein Kinases/metabolism , Ovarian Neoplasms/pathology , bcl-2-Associated X Protein/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Protein Binding , Receptors, LHRH/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/physiology
16.
Bioorg Med Chem Lett ; 17(22): 6091-5, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17904839

ABSTRACT

Two series of azanaphthoquinone annelated pyrrolo oximes have been synthesized. The antiproliferative activities of 10 compounds were evaluated on at least four different cell lines. One series of pyrrolo derivatives showed high cytotoxic activity. The effects on cell cycle and caspase activity were investigated. Compounds 9a and 9b showed an accumulation of cells in G2/M phase. Substantial and dose-dependent caspase activity was found after treatment of cells with 9a and 9b. This indicates an apoptosis inducing property of these compounds.


Subject(s)
Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Mitoxantrone/chemical synthesis , Naphthoquinones/chemical synthesis , Oximes/chemical synthesis , Pyrroles/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Caspases/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Mitoxantrone/analogs & derivatives , Mitoxantrone/pharmacology , Molecular Structure , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Oximes/chemistry , Oximes/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology
17.
EMBO J ; 25(17): 4020-32, 2006 Sep 06.
Article in English | MEDLINE | ID: mdl-16917500

ABSTRACT

Autoinhibition is a recurring mode of protein kinase regulation and can be based on diverse molecular mechanisms. Here, we show by crystal structure analysis, nuclear magnetic resonance (NMR)-based nucleotide affinity studies and rational mutagenesis that nonphosphorylated mitogen-activated protein (MAP) kinases interacting kinase (Mnk) 1 is autoinhibited by conversion of the activation segment into an autoinhibitory module. In a Mnk1 crystal structure, the activation segment is repositioned via a Mnk-specific sequence insertion at the N-terminal lobe with the following consequences: (i) the peptide substrate binding site is deconstructed, (ii) the interlobal cleft is narrowed, (iii) an essential Lys-Glu pair is disrupted and (iv) the magnesium-binding loop is locked into an ATP-competitive conformation. Consistently, deletion of the Mnk-specific insertion or removal of a conserved phenylalanine side chain, which induces a blockade of the ATP pocket, increase the ATP affinity of Mnk1. Structural rearrangements required for the activation of Mnks are apparent from the cocrystal structure of a Mnk2 D228G -staurosporine complex and can be modeled on the basis of crystal packing interactions. Our data suggest a novel regulatory mechanism specific for the Mnk subfamily.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Protein Serine-Threonine Kinases/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Magnesium/chemistry , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Phenylalanine/chemistry , Phosphorylation , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Staurosporine/chemistry
18.
Structure ; 13(10): 1559-68, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16216586

ABSTRACT

Human mitogen-activated protein kinases (MAPK)-interacting kinases 1 and 2 (Mnk1 and Mnk2) target the translational machinery by phosphorylation of the eukaryotic initiation factor 4E (eIF4E). Here, we present the 2.1 A crystal structure of a nonphosphorylated Mnk2 fragment that encompasses the kinase domain. The results show Mnk-specific features such as a zinc binding motif and an atypical open conformation of the activation segment. In addition, the ATP binding pocket contains an Asp-Phe-Asp (DFD) in place of the canonical magnesium binding Asp-Phe-Gly (DFG) motif. The phenylalanine of this motif sticks into the ATP binding pocket and blocks ATP binding as observed with inhibitor bound and, thus, inactive p38 kinase. Replacement of the DFD by the canonical DFG motif affects the conformation of Mnk2, but not ATP binding and kinase activity. The results suggest that the ATP binding pocket and the activation segment of Mnk2 require conformational switches to provide kinase activity.


Subject(s)
Crystallography, X-Ray , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Zinc/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Alternative Splicing , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Catalysis , Chromatography, Gel , Humans , Intracellular Signaling Peptides and Proteins , Light , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Phenylalanine/chemistry , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Scattering, Radiation , Sequence Homology, Amino Acid , Spectrum Analysis, Raman , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...