Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Steroid Biochem Mol Biol ; 192: 105115, 2019 09.
Article in English | MEDLINE | ID: mdl-29438723

ABSTRACT

We report the discovery of a novel nonsteroidal dual-action compound, ODM-204, that holds promise for treating patients with castration-resistant prostate cancer (CRPC), an advanced form of prostate cancer characterised by high androgen receptor (AR) expression and persistent activation of the AR signaling axis by residual tissue androgens. For ODM-204, has a dual mechanism of action. The compound is anticipated to efficiently dampen androgenic stimuli in the body by inhibiting CYP17A1, the prerequisite enzyme for the formation of dihydrotestosterone (DHT) and testosterone (T), and by blocking AR with high affinity and specificity. In our study, ODM-204 inhibited the proliferation of androgen-dependent VCaP and LNCaP cells in vitro and reduced significantly tumour growth in a murine VCaP xenograft model in vivo. Intriguingly, after a single oral dose of 10-30 mg/kg, ODM-204 dose-dependently inhibited adrenal and testicular steroid production in sexually mature male cynomolgus monkeys. Similar results were obtained in human chorionic gonadotropin-treated male rats. In rats, leuprolide acetate-mediated (LHRH agonist) suppression of the circulating testosterone levels and decrease in weights of androgen-sensitive organs was significantly and dose-dependently potentiated by the co-administration of ODM-204. ODM-204 was well tolerated in both rodents and primates. Based on our data, ODM-204 could provide an effective therapeutic option for men with CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/chemistry , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Haplorhini , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 18(1): 28-38, 2019 01.
Article in English | MEDLINE | ID: mdl-30301864

ABSTRACT

Alterations in the gene encoding for the FGFR and upregulation of the VEGFR are found often in cancer, which correlate with disease progression and unfavorable survival. In addition, FGFR and VEGFR signaling synergistically promote tumor angiogenesis, and activation of FGFR signaling has been described as functional compensatory angiogenic signal following development of resistance to VEGFR inhibition. Several selective small-molecule FGFR kinase inhibitors are currently in clinical development. ODM-203 is a novel, selective, and equipotent inhibitor of the FGFR and VEGFR families. In this report we show that ODM-203 inhibits FGFR and VEGFR family kinases selectively and with equal potency in the low nanomolar range (IC50 6-35 nmol/L) in biochemical assays. In cellular assays, ODM-203 inhibits VEGFR-induced tube formation (IC50 33 nmol/L) with similar potency as it inhibits proliferation in FGFR-dependent cell lines (IC50 50-150 nmol/L). In vivo, ODM-203 shows strong antitumor activity in both FGFR-dependent xenograft models and in an angiogenic xenograft model at similar well-tolerated doses. In addition, ODM-203 inhibits metastatic tumor growth in a highly angiogenesis-dependent kidney capsule syngenic model. Interestingly, potent antitumor activity in the subcutaneous syngenic model correlated well with immune modulation in the tumor microenvironment as indicated by marked decrease in the expression of immune check points PD-1 and PD-L1 on CD8 T cells and NK cells, and increased activation of CD8 T cells. In summary, ODM-203 shows equipotent activity for both FGFR and VEGFR kinase families and antitumor activity in both FGFR and angigogenesis models.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/metabolism , Protein Kinase Inhibitors/administration & dosage , T-Lymphocytes/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Kidney Neoplasms/metabolism , Killer Cells, Natural/metabolism , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 25(22): 5309-14, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26421993

ABSTRACT

Hepsin, a type II transmembrane serine protease, is upregulated in prostate cancer and known to be involved in the progression of metastasis. Here we report a structure-guided approach, which resulted in the discovery of 2-aryl/pyridin-2-yl-1H-indole derivatives as potent and selective inhibitors of hepsin. Potent and selective inhibition of hepsin by compound 8 is likely due to interactions of the amidine group at the S1 site with the cyclohexyl ring from the 2-aryl group projecting towards the S1' site and the tert-hydroxyl group interacting with His57 side-chain as revealed by X-ray crystallography. Compounds 8 and 10, showed Ki of 0.1 µM for hepsin, and exhibited inhibition of invasion and migration of hepsin-overexpressing cell line. Compounds described here could serve as useful tool reagents to investigate the role of hepsin as a potential therapeutic target in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclohexanes/pharmacology , Indoles/pharmacology , Pyridines/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Movement/drug effects , Cyclohexanes/chemical synthesis , Humans , Indoles/chemical synthesis , Mice , Microsomes, Liver/metabolism , Molecular Docking Simulation , Neoplasm Invasiveness , Pyridines/chemical synthesis , Serine Proteinase Inhibitors/chemical synthesis
4.
Sci Rep ; 5: 12007, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26137992

ABSTRACT

Activation of androgen receptor (AR) is crucial for prostate cancer growth. Remarkably, also castration-resistant prostate cancer (CRPC) is dependent on functional AR, and several mechanisms have been proposed to explain the addiction. Known causes of CRPC include gene amplification and overexpression as well as point mutations of AR. We report here the pharmacological profile of ODM-201, a novel AR inhibitor that showed significant antitumor activity and a favorable safety profile in phase 1/2 studies in men with CRPC. ODM-201 is a full and high-affinity AR antagonist that, similar to second-generation antiandrogens enzalutamide and ARN-509, inhibits testosterone-induced nuclear translocation of AR. Importantly, ODM-201 also blocks the activity of the tested mutant ARs arising in response to antiandrogen therapies, including the F876L mutation that confers resistance to enzalutamide and ARN-509. In addition, ODM-201 reduces the growth of AR-overexpressing VCaP prostate cancer cells both in vitro and in a castration-resistant VCaP xenograft model. In contrast to other antiandrogens, ODM-201 shows negligible brain penetrance and does not increase serum testosterone levels in mice. In conclusion, ODM-201 is a potent AR inhibitor that overcomes resistance to AR-targeted therapies by antagonizing both overexpressed and mutated ARs. ODM-201 is currently in a phase 3 trial in CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Prostatic Neoplasms/drug therapy , Pyrazoles/pharmacology , Androgens/physiology , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Resistance, Neoplasm , HEK293 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/pathology , Pyrazoles/pharmacokinetics , Receptors, Androgen/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Bioorg Med Chem ; 22(12): 3187-203, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24794746

ABSTRACT

Matriptase is a serine protease implicated in cancer invasion and metastasis. Expression of matriptase is frequently dysregulated in human cancers and matriptase has been reported to activate latent growth factors such as hepatocyte growth factor/scatter factor, and proteases such as urokinase plasminogen activator suggesting that matriptase inhibitors could have therapeutic potential in treatment of cancer. Here we report a structure-based approach which led to the discovery of selective and potent matriptase inhibitors with benzene as central core having 1,3,5 tri-substitution pattern. X-ray crystallography of one of the potent analogs in complex with matriptase revealed strong hydrogen bonding and salt-bridge interactions in the S1 pocket, as well as strong CH-π contacts between the P2/P4 cyclohexyl and Trp215 side-chain. An additional interaction of the pendant amine at cyclohexyl with Gln175 side-chain results in substantial improvement in matriptase inhibition and selectivity against other related serine proteases. Compounds 15 and 26 showed tumor growth inhibition in a subcutaneous DU-145 prostate cancer mouse model. These compounds could be useful as tools to further explore the biology of matriptase as a drug target.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Benzene/pharmacology , Cell Proliferation/drug effects , Cyclohexanes/pharmacology , Drug Discovery , Prostatic Neoplasms/pathology , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Benzamides/chemistry , Benzene/chemistry , Binding Sites , Crystallography, X-Ray , Cyclohexanes/chemical synthesis , Humans , Male , Mice , Mice, SCID , Models, Molecular , Molecular Sequence Data , Prostatic Neoplasms/drug therapy , Sequence Homology, Amino Acid , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
6.
ACS Med Chem Lett ; 4(12): 1152-7, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24900621

ABSTRACT

Matriptase belongs to trypsin-like serine proteases involved in matrix remodeling/degradation, growth regulation, survival, motility, and cell morphogenesis. Herein, we report a structure-based approach, which led to the discovery of sulfonamide and amide derivatives of pyridyl bis(oxy)benzamidine as potent and selective matriptase inhibitors. Co-crystal structures of selected compounds in complex with matriptase supported compound designing. Additionally, WaterMap analyses indicated the possibility of occupying a distinct pocket within the catalytic domain, exploration of which resulted in >100-fold improvement in potency. Co-crystal structure of 10 with matriptase revealed critical interactions leading to potent target inhibition and selectivity against other serine proteases.

7.
Adv Cancer Res ; 111: 97-161, 2011.
Article in English | MEDLINE | ID: mdl-21704831

ABSTRACT

Epithelial architecture is formed in tissues and organs when groups of epithelial cells are organized into polarized structures. The epithelial function and integrity as well as signaling across the epithelial layer is orchestrated by apical junctional complexes (AJCs), which are landmarks for PAR/CRUMBS and lateral SCRIB polarity modules and by dynamic interactions of the cells with underlying basement membrane (BM). These highly organized epithelial architectures are demolished in cancer. In all advanced epithelial cancers, malignant cells have lost polarity and connections to the basement membrane and they have become proliferative, motile, and invasive. Clearly, loss of epithelial integrity associates with tumor progression but does it contribute to tumor development? Evidence from studies in Drosophila and recently also in vertebrate models have suggested that even the oncogene-driven enforced cell proliferation can be conditional, dependant on the influence of cell-cell or cell-microenvironment contacts. Therefore, loss of epithelial integrity may not only be an obligate consequence of unscheduled proliferation of malignant cells but instead, malignant epithelial cells may need to acquire capacity to break free from the constraints of integrity to freely and autonomously proliferate. We discuss how epithelial polarity complexes form and regulate epithelial integrity, highlighting the roles of enzymes Rho GTPases, aPKCs, PI3K, and type II transmembrane serine proteases (TTSPs). We also discuss relevance of these pathways to cancer in light of genetic alterations found in human cancers and review molecular pathways and potential pharmacological strategies to revert or selectively eradicate disorganized tumor epithelium.


Subject(s)
Cell Polarity , Epithelial Cells/pathology , Genes/genetics , Neoplasms/genetics , Neoplasms/pathology , Animals , Humans
8.
J Neurochem ; 86(1): 165-72, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12807436

ABSTRACT

Overexpression of human alpha-synuclein in model systems, including cultured neurons, drosophila and mice, leads to biochemical and pathological changes that mimic synucleopathies including Parkinson's disease. We have overexpressed both wild-type (WT) and mutant alanine53-->threonine (A53T) human alpha-synuclein by transgenic injection into Caenorhabditis elegans. Motor deficits were observed when either WT or A53T alpha-synuclein was overexpressed with a pan-neuronal or motor neuron promoter. Neuronal and dendritic loss were accelerated in all three sets of C. elegans dopaminergic neurons when human alpha-synuclein was overexpressed under the control of a dopaminergic neuron or pan-neuronal promoter, but not with a motor neuron promoter. There were no significant differences in neuronal loss between overexpressed WT and A53T forms or between worms of different ages (4 days, 10 days or 2 weeks). These results demonstrate neuronal and behavioral perturbations elicited by human alpha-synuclein in C. elegans that are dependent upon expression in specific neuron subtypes. This transgenic model in C. elegans, an invertebrate organism with excellent experimental resources for further genetic manipulation, may help facilitate dissection of pathophysiologic mechanisms of various synucleopathies.


Subject(s)
Caenorhabditis elegans , Dopamine/metabolism , Nerve Tissue Proteins/biosynthesis , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Animals , Animals, Genetically Modified , Behavior, Animal , Biological Assay/statistics & numerical data , Cell Count , Disease Models, Animal , Genes, Reporter , Humans , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurons/pathology , Synucleins , alpha-Synuclein
9.
Mol Biol Cell ; 13(6): 2106-19, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12058073

ABSTRACT

Nuclear receptors, including the androgen receptor (AR), regulate target cell transcription through interaction with auxiliary proteins to modify chromatin structure. We describe herein a novel AR-interacting protein, termed ARIP4, that has structural features typical of the SNF2-like protein family. With regard to the Snf2 domain, the closest homolog of ARIP4 is the ATRX protein. ARIP4 is a nuclear protein and comprises 1466 amino acids. It interacts with AR in vitro and in cultured yeast and mammalian cells. ARIP4 can be labeled with 8-azido-[gamma-32P]ATP and exhibits DNA-dependent ATPase activity. Like several ATP-dependent chromatin remodeling proteins, ARIP4 generates superhelical torsion within linear DNA fragments in an ATP-dependent manner. With a stably integrated target promoter, ARIP4 elicits a modest enhancement of AR-dependent transactivation. In transient cotransfection assays, ARIP4 modulates AR function in a promoter-dependent manner; it enhances receptor activity on minimal promoters, but does not activate more complex promoters. ARIP4 mutants devoid of ATPase activity fail to alter DNA topology and behave as trans-dominant negative regulators of AR function in transient assays.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Nuclear Proteins , Receptors, Androgen/metabolism , Saccharomyces cerevisiae/physiology , Transcription Factors/metabolism , Transcription, Genetic , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , COS Cells , Chlorocebus aethiops , DNA, Complementary , Genes, Reporter , Insecta , Molecular Sequence Data , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins , Sequence Alignment , Serine Endopeptidases/genetics , Transfection , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...