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.
Front Chem ; 8: 166, 2020.
Article in English | MEDLINE | ID: mdl-32328475

ABSTRACT

Naphthalimides, such as amonafide and mitonafide in clinical trials, have been developed as antitumor agents for orthotopic tumor. However, the serious side effects in cancer patients limit their applications. Herein, a new class of polyamine-based naphthalimide conjugates 5a-5c, 7a-7b, and 11a-11b with and without the alkylation of the distant nitrogen in the polyamine chain were synthesized and the mechanism was determined. Compared with amonafide, dinitro-naphthalimide conjugate 5c with a 4,3-cyclopropyl motif preferentially accumulates in cancer cells and minimizes side effects in vitro and in vivo. More importantly, 5c at the dosage of as low as 3 mg/kg (57.97%) displays better antitumor effects than the positive control amonafide (53.27%) at 5 mg/kg in vivo. And a remarkably elevated antitumor activity and a reduced toxicity are also observed for 5c at 5 mg/kg (65.90%). The upregulated p53 and the apoptotic cells (73.50%) indicate that the mechanism of 5c to induce apoptosis may result from its enhanced DNA damage. Further investigation indicates that in addition to target DNA, 5c can modulate the polyamine homeostasis by upregulating polyamine oxidase (PAO) in a different way from that of amonafide. And also by targeting PTs overexpressed in most of cancer cells, 5c downregulates the contents of Put, Spd, and Spm, which are in favor of suppressing fast-growing tumor cells. Our study implies a promising strategy for naphthalimide conjugates to treat hepatic carcinoma with notable activities and reduced toxicities at a low dosage.

2.
J Med Chem ; 62(24): 11324-11334, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31765154

ABSTRACT

Diverse platinum drug candidates have been designed to improve inhibitory potency and overcome resistance for orthotopic tumors. However, the antimetastatic properties have rarely been reported. We herein report that homospermidineplatin (4a), a polyamine-Pt(IV) prodrug, can potently inhibit tumor growth in situ and reverse cisplatin resistance as expected, and more importantly, 4a displays remarkably elevated antimetastatic activity in vivo (65.7%), compared to those of cisplatin (27.0%) and oxaliplatin (19.6%). The underlying molecular mechanism indicates that in addition to targeting nuclear DNA, 4a can modulate polyamine metabolism and function in a manner different from that of cisplatin. By upregulating SSAT and PAO, 4a downregulates the concentrations of Put, Spd, and Spm, which favors the suppression of fast-growing tumor cells. Moreover, the p53/SSAT/ß-catenin and PAO/ROS/GSH/GSH-Px pathways are involved in the inhibition of 4a-induced tumor metastasis. Our study implies a promising strategy for the design of platinum drugs for the treatment of terminal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , DNA/metabolism , Lung Neoplasms/drug therapy , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Polyamines/metabolism , Prodrugs/metabolism , Animals , Apoptosis , Biological Transport , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Oxaliplatin/pharmacology , Prodrugs/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polyamine Oxidase
3.
Oncol Rep ; 41(3): 1971-1979, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30569119

ABSTRACT

Liver cancer is among the most common types of cancer worldwide. The aim of the present study was to investigate whether the phosphatidylinositol­3­phosphate 5­kinase (PIKfyve) inhibitor, YM201636, exerts anti­proliferative effects on liver cancer. The methods used in the present study included MTT assay, flow cytometry, western blot analysis and an allograft mouse model of liver cancer. The results revealed that YM201636 inhibited the proliferation of HepG2 and Huh­7 cells in a dose­dependent manner. HepG2 and Huh­7 cells exhibited strong monodansylcadaverine staining following treatment with YM201636. Accordingly, YM201636 treatment increased the expression of the autophagosome­associated marker protein microtubule­associated 1A/1B light chain 3­II in HepG2 and Huh­7 cells. The autophagy inhibitor 3­methyladenine attenuated the inhibitory effects of YM201636 on liver cancer cell proliferation. Further in vivo analysis revealed that YM201636 (2 mg/kg) inhibited tumor growth without notable systemic toxicity. Mechanistic experiments demonstrated that YM201636 induced­autophagy is dependent upon epidermal growth factor receptor (EGFR) overexpression in HepG2 and Huh­7 cells. Collectively, these results suggested that the PIKfyve inhibitor YM201636 may inhibit tumor growth by promoting EGFR expression. This indicates that PIKfyve may be a potential therapeutic target for the treatment of liver cancer.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Liver Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Adult , Aminopyridines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Xenograft Model Antitumor Assays
4.
Cell Commun Signal ; 16(1): 82, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30445978

ABSTRACT

BACKGROUND: Knockdown of Akt1 promotes Epithelial-to-Mesenchymal Transition in breast cancer cells. However, the mechanisms are not completely understood. METHODS: Western blotting, immunofluorescence, luciferase assay, real time PCR, ELISA and Matrigel invasion assay were used to investigate how Akt1 inhibition promotes breast cancer cell invasion in vitro. Mouse model of lung metastasis was used to measure in vivo efficacy of Akt inhibitor MK2206 and its combination with Gefitinib. RESULTS: Knockdown of Akt1 stimulated ß-catenin nuclear accumulation, resulting in breast cancer cell invasion. ß-catenin nuclear accumulation induced by Akt1 inhibition depended on the prolonged activation of EGFR signaling pathway in breast cancer cells. Mechanistic experiments documented that knockdown of Akt1 inactivates PIKfyve via dephosphorylating of PIKfyve at Ser318 site, resulting in a decreased degradation of EGFR signaling pathway. Inhibition of Akt1 using MK2206 could induce an increase in the expression of EGFR and ß-catenin in breast cancer cells. In addition, MK2206 at a low dosage enhance breast cancer metastasis in a mouse model of lung metastasis, while an inhibitor of EGFR tyrosine kinase Gefitinib could potentially suppress breast cancer metastasis induced by Akt1 inhibition. CONCLUSION: EGFR-mediated ß-catenin nuclear accumulation is critical for Akt1 inhibition-induced breast cancer metastasis.


Subject(s)
Breast Neoplasms/pathology , Cell Nucleus/metabolism , ErbB Receptors/metabolism , Gene Knockdown Techniques , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , beta Catenin/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Cell Nucleus/drug effects , ErbB Receptors/antagonists & inhibitors , Gefitinib/pharmacology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MCF-7 Cells , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/metabolism
5.
Front Chem ; 6: 386, 2018.
Article in English | MEDLINE | ID: mdl-30298127

ABSTRACT

Physiological characteristics of human malignancies are increased glycolysis and overexpression of glucose transporters (GLUTs). 18Flurodeoxyglucose-positron emission tomography (FDG-PET) has successfully developed as clinical modality for the diagnosis and staging of many cancers based on the Warburg effect. To leverage this glucose transporter mediated metabolic disparity between normal and malignant cells, in the current report, protected, and de-protected glucose, mannose, galactose, rhamnose, maltose, and lactose-conjugated platinum(IV) complexes were designed and synthesized. The suggested potential of facilitated intravenous to oral switching of glycosylated platinum(IV) prodrugs with cancer-targeting properties were evaluated for glucose transporter 1 (GLUT1) and organic cation transporter 2 (OCT2)-mediated selective properties in vitro and in vivo. The cytotoxicity of 2d, 5d, and 6d were ~23-fold greater than that of the positive controls cisplatin, oxaliplatin, and satraplatin, respectively. The leading compound 6d, the IC50 of which with the GLUT1 inhibitor 4,6-oethylidene-α-D-glucose (EDG) and phloretin (31.80 and 38.71 µM) are 36- and 44-folds higher, respectively, than the 48 h IC50 (0.89 µM), is superior to the reported 5-8, exhibiting enhanced cancer targeting. The compounds also showed reduced toxicity to normal cells (293T IC50 = 12.06 µM and 3T3 cells IC50 > 100 µM) and exhibited no cross-resistance to cisplatin. Moreover, the encouraging selectivity of 6d for MCF-7 cells in vivo indicated that the pyranoside performs an important function in cancer targeting.

SELECTION OF CITATIONS
SEARCH DETAIL
...