Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 143: 1535-1542, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29126726

ABSTRACT

ABCB1-mediated multidrug resistance (MDR) is a principal obstacle for successful cancer chemotherapy. A series of pyrimidine-based hybrid molecules containing 1,2,3-triazole moiety were evaluated for their reversal activities against MDR. The majority of target compounds displayed moderate to great reversal potency. Among these compounds, compound 25 displayed the most potent reversal activity, about 7-fold more potent than Verapamil (VRP). Further mechanism studies revealed that compound 25 could obviously reverse paclitaxel (PTX) resistance in SW620/AD300 cells by increasing accumulation and extending maintenance of PTX. Our findings indicate that the 1,2,3-triazole-pyrimidine-based derivatives may serve as an interesting lead for the development of new potent and efficacious ABCB1-dependent MDR modulators.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Pyrimidines/pharmacology , Triazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Pyrimidines/chemistry , Structure-Activity Relationship , Triazoles/chemistry , Tumor Cells, Cultured
2.
Pharmacol Res ; 122: 66-77, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28571892

ABSTRACT

Histone lysine specific demethylase 1 (LSD1) plays an important role in epigenetic modifications, and aberrant expression of LSD1 predicts tumor progression and poor prognosis in human esophageal cancers. In this study, a series of LSD1 inhibitors were synthesized and proved to be highly potent against human esophageal squamous cell carcinoma (ESCC). Our data showed that these LSD1 inhibitors selectively suppressed the viability of esophageal cancer cell line (EC-109) bearing overexpressed LSD1. Among these, compound LPE-1 (LSD1 IC50=0.336±0.003µM) significantly suppressed proliferation, induced apoptosis, arrested cell cycle of EC109 cells at G2/M phase, and caused changes of the associated protein markers correspondingly. We also found that compound LPE-1 potently inhibited the migration and invasion of EC-109 cells. Docking studies showed that the cyano group formed hydrogen bonds with Val811 and Thr810. Additionally, the thiophene moiety formed arene-H interaction with Trp761 residue. In vivo studies showed that compound LPE-1 inhibited tumor growth of xenograft models bearing EC-109 without obvious toxicity. Collectively, our findings indicate that LSD1 may be a potential therapeutic target in ESCC, and compound LPE-1 could serve as a lead compound for further development for anti-ESCC drug discovery.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Esophageal Neoplasms/drug therapy , Histone Demethylases/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophagus/drug effects , Esophagus/metabolism , Esophagus/pathology , Female , Histone Demethylases/metabolism , Humans , Mice, Inbred BALB C , Molecular Docking Simulation , Pyrimidines/therapeutic use
3.
Bioorg Med Chem Lett ; 25(5): 1124-8, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25655718

ABSTRACT

A series of novel 1,2,3-triazole-pyrimidine-urea hybrids were designed, synthesized and evaluated for anticancer activity against four selected cancer cell lines (MGC-803, EC-109, MCF-7 and B16-F10). Majority of the synthesized compounds exhibited moderate to potent activity against all the cancer cell lines assayed. Particularly, compounds 26, 30 and 38 exhibited excellent growth inhibition against B16-F10 with IC50 values of 32nM, 35nM and 42nM, respectively. Flow cytometry analysis demonstrated that compound 26 induced the cellular apoptosis in a concentration-dependent manner.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Pyrimidines/chemistry , Triazoles/chemistry , Urea/analogs & derivatives , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacology , Urea/chemical synthesis , Urea/pharmacology
4.
J Med Chem ; 58(4): 1705-16, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25610955

ABSTRACT

Histone lysine specific demethylase 1 (LSD1) was reported to be overexpressed in several human cancers and recognized as a promising anticancer drug target. In the current study, we designed and synthesized a novel series of pyrimidine-thiourea hybrids and evaluated their potential LSD1 inhibitory effect. One of the compounds, 6b, containing a terminal alkyne appendage, was shown to be the most potent and selective LSD1 inhibitor in vitro and exhibited strong cytotoxicity against LSD1 overexpressed gastric cancer cells. Compound 6b also showed marked inhibition of cell migration and invasion as well as significant in vivo tumor suppressing and antimetastasis role, without significant side effects by oral administration. Our findings indicate that the pyrimidine-thiourea-based LSD1 inactivator may serve as a leading compound targeting LSD1 overexpressed cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Pyrimidines/pharmacology , Thiourea/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Structure , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Structure-Activity Relationship , Thiourea/administration & dosage , Thiourea/chemistry
5.
Eur J Med Chem ; 86: 368-80, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25180925

ABSTRACT

A series of novel 1,2,3-triazole-pyrimidine hybrids were designed, synthesized and evaluated for their anticancer activity against four selected cancer cell lines (MGC-803, EC-109, MCF-7 and B16-F10). Most of the synthesized compounds exhibited moderate to good activity against all the cancer cell lines selected. Compound 17 showed the most excellent anticancer activity with single-digit micromolar IC50 values ranging from 1.42 to 6.52 µM. Further mechanism studies revealed that compound 17 could obviously inhibit the proliferation of EC-109 cancer cells by inducing apoptosis and arresting the cell cycle at G2/M phase.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Pyrimidines/pharmacology , Triazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Molecular Structure , Pyrimidines/chemistry , Structure-Activity Relationship , Triazoles/chemistry
6.
Steroids ; 78(12-13): 1200-8, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-23911850

ABSTRACT

Using dehydroepiandrosterone as the starting material, we have synthesized a series of steroid analogs possessing a D-ring fused with heterocycles which are pyridine, imidazo [2,1-b]thiazoles or substituted thiazole imines. All the final structures are first reported and identified by NMR and MS spectroscopys, the yields of these products are moderate to good and the reaction conditions are mild. The cytotoxicity of the synthesized compounds against EC-109(human esophageal carcinoma), EC-9706(human esophageal carcinoma), MGC-803(human gastric carcinoma) were investigated.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/chemical synthesis , Imines/chemical synthesis , Thiazoles/chemical synthesis , Cyclization , Drug Design , Halogenation
SELECTION OF CITATIONS
SEARCH DETAIL
...