Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Clin Cancer Res ; 38(1): 440, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31666104

ABSTRACT

In the original publication of this article [1], there are mistakes in Fig. 4d. The corrected Fig. 4 should be.

2.
J Exp Clin Cancer Res ; 38(1): 342, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31387608

ABSTRACT

BACKGROUND: Androgen receptor (AR)-targeted treatments improve the survival of castration-resistant prostate cancer (CRPC) patients; however, secondary resistance to these agents ultimately occurs in virtually all patients. Therefore, alternative therapeutic targets are urgently needed. Since growing evidence demonstrates that WNT/ß-catenin signaling plays an important role in CRPC, the antitumor activity and mechanism of action of CWP232291, a small molecule ß-catenin inhibitor, were investigated in prostate cancer. METHODS: We assessed the antitumor activity of CWP232291 in prostate cancer cell lines and primary cells derived from CRPC patients. The effect of CWP232291 on apoptotic cell death, endoplasmic reticulum (ER) stress, cell viability, and WNT/ß-catenin signaling was evaluated by flow cytometry, western blotting, luciferase reporter assay, and fluorescence microscopy. Antitumor efficacy was assessed in two CRPC xenograft mouse models. RESULTS: CWP232291 induced ER stress, resulting in upregulation of the proapoptotic protein CHOP and activation of caspase-3-dependent apoptosis. In addition, CWP232291 suppressed the expression of ß-catenin by affecting WNT-dependent transcriptional activity, and downregulated AR and its splice variants in prostate cancer cells. Antitumor activity was observed in prostate cancer cells in vitro and ex vivo, and antitumor efficacy was observed in vivo. CONCLUSIONS: Beyond providing preclinical evidence of therapeutic efficacy for the novel small molecule ß-catenin inhibitor CWP232291 in CRPC, our results show that inducing ER stress and targeting WNT/ß-catenin signaling may be a novel strategy against CRPC.

3.
J Med Chem ; 61(7): 2949-2961, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29579390

ABSTRACT

The histamine H4 receptor (H4R), a member of the G-protein coupled receptor family, has been considered as a potential therapeutic target for treating atopic dermatitis (AD). A large number of H4R antagonists have been disclosed, but no efficient agents controlling both pruritus and inflammation in AD have been developed yet. Here, we have discovered a novel class of orally available H4R antagonists showing strong anti-itching and anti-inflammation activity as well as excellent selectivity against off-targets. A pharmacophore-based virtual screening system constructed in-house successfully identified initial hit compound 9, and the subsequent homology model-guided optimization efficiently led us to discover pyrido[2,3- e]tetrazolo[1,5- a]pyrazine analogue 48 as a novel chemotype of a potent and highly selective H4R antagonist. Importantly, orally administered compound 48 exhibits remarkable efficacy on antipruritus and anti-inflammation with a favorable pharmacokinetic (PK) profile in several mouse models of AD. Thus, these data strongly suggest that our compound 48 is a promising clinical candidate for treatment of AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Histamine Antagonists/chemical synthesis , Histamine Antagonists/therapeutic use , Receptors, Histamine H4/antagonists & inhibitors , Animals , Biological Availability , Computer Simulation , Drug Discovery , Drug Evaluation, Preclinical , Female , Histamine Antagonists/pharmacokinetics , Inflammation/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Pruritus/drug therapy , Receptors, Histamine H4/metabolism , Structure-Activity Relationship
4.
J Pharmacol Exp Ther ; 357(1): 157-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26907620

ABSTRACT

Urate-lowering therapy is indispensable for the treatment of gout, but available drugs do not control serum urate levels tightly enough. Although the uricosurics benzbromarone and probenecid inhibit a urate reabsorption transporter known as renal urate transporter 1 (URAT1) and thus lower serum urate levels, they also inhibit other transporters responsible for secretion of urate into urine, which suggests that inhibiting URAT1 selectively would lower serum urate more effectively. We identified a novel potent and selective URAT1 inhibitor, UR-1102, and compared its efficacy with benzbromarone in vitro and in vivo. In human embryonic kidney (HEK)293 cells overexpressing URAT1, organic anion transporter 1 (OAT1), and OAT3, benzbromarone inhibited all transporters similarly, whereas UR-1102 inhibited URAT1 comparably to benzbromarone but inhibited OAT1 and OAT3 quite modestly. UR-1102 at 3-30 mg/kg or benzbromarone at 3-100 mg/kg was administered orally once a day for 3 consecutive days to tufted capuchin monkeys, whose low uricase activity causes a high plasma urate level. When compared with the same dosage of benzbromarone, UR-1102 showed a better pharmacokinetic profile, increased the fractional excretion of urinary uric acid, and reduced plasma uric acid more effectively. Moreover, the maximum efficacy of UR-1102 was twice that of benzbromarone, suggesting that selective inhibition of URAT1 is effective. Additionally UR-1102 showed lower in vitro potential for mechanisms causing the hepatotoxicity induced by benzbromarone. These results indicate that UR-1102 achieves strong uricosuric effects by selectively inhibiting URAT1 over OAT1 and OAT3 in monkeys, and could be a novel therapeutic option for patients with gout or hyperuricemia.


Subject(s)
Benzbromarone/pharmacology , Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Oxazines/pharmacology , Pyridines/pharmacology , Uricosuric Agents/pharmacology , Animals , Cebus , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Organic Anion Transport Protein 1/biosynthesis , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/biosynthesis , Organic Anion Transporters, Sodium-Independent/genetics , Organic Cation Transport Proteins/genetics , Protein Binding , Uric Acid/blood , Uricosuric Agents/adverse effects
5.
Cancer Res ; 75(8): 1691-702, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25660951

ABSTRACT

Breast cancer stem cells (BCSC) are resistant to conventional chemotherapy and radiotherapy, which may destroy tumor masses but not all BCSC that can mediate relapses. In the present study, we showed that the level of Wnt/ß-catenin signaling in BCSC is relatively higher than in bulk tumor cells, contributing to a relatively higher level of therapeutic resistance. We designed a highly potent small-molecule inhibitor, CWP232228, which antagonizes binding of ß-catenin to T-cell factor (TCF) in the nucleus. Notably, although CWP232228 inhibited the growth of both BCSC and bulk tumor cells by inhibiting ß-catenin-mediated transcription, BCSC exhibited greater growth inhibition than bulk tumor cells. We also documented evidence of greater insulin-like growth factor-I (IGF-I) expression by BCSC than by bulk tumor cells and that CWP232228 attenuated IGF-I-mediated BCSC functions. These results suggested that the inhibitory effect of CWP232228 on BCSC growth might be achieved through the disruption of IGF-I activity. Taken together, our findings indicate that CWP232228 offers a candidate therapeutic agent for breast cancer that preferentially targets BCSC as well as bulk tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Azabicyclo Compounds/pharmacology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Neoplastic Stem Cells/drug effects , Organophosphates/pharmacology , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Small Molecule Libraries , Substrate Specificity , Tumor Cells, Cultured , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...