Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Drug Resist Updat ; 72: 101016, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37980859

ABSTRACT

Drug resistance remains a major challenge in cancer treatment, necessitating the development of novel strategies to overcome it. Protein arginine methyltransferases (PRMTs) are enzymes responsible for epigenetic arginine methylation, which regulates various biological and pathological processes, as a result, they are attractive therapeutic targets for overcoming anti-cancer drug resistance. The ongoing development of small molecules targeting PRMTs has resulted in the generation of chemical probes for modulating most PRMTs and facilitated clinical treatment for the most advanced oncology targets, including PRMT1 and PRMT5. In this review, we summarize various mechanisms underlying protein arginine methylation and the roles of specific PRMTs in driving cancer drug resistance. Furthermore, we highlight the potential clinical implications of PRMT inhibitors in decreasing cancer drug resistance. PRMTs promote the formation and maintenance of drug-tolerant cells via several mechanisms, including altered drug efflux transporters, autophagy, DNA damage repair, cancer stem cell-related function, epithelial-mesenchymal transition, and disordered tumor microenvironment. Multiple preclinical and ongoing clinical trials have demonstrated that PRMT inhibitors, particularly PRMT5 inhibitors, can sensitize cancer cells to various anti-cancer drugs, including chemotherapeutic, targeted therapeutic, and immunotherapeutic agents. Combining PRMT inhibitors with existing anti-cancer strategies will be a promising approach for overcoming anti-cancer drug resistance. Furthermore, enhanced knowledge of the complex functions of arginine methylation and PRMTs in drug resistance will guide the future development of PRMT inhibitors and may help identify new clinical indications.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Arginine/metabolism , Arginine/therapeutic use , Tumor Microenvironment , Repressor Proteins/therapeutic use
2.
Mol Cell Biochem ; 479(4): 831-841, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37199893

ABSTRACT

Metastasis is the cause of poor prognosis in ovarian cancer (OC). Enhancer of Zeste homolog 2 (EZH2), a histone-lysine N-methyltransferase enzyme, promotes OC cell migration and invasion by regulating the expression of tissue inhibitor of metalloproteinase-2 (TIMP2) and matrix metalloproteinases-9 (MMP9). Hence, we speculated that EZH2-targeting therapy might suppress OC migration and invasion. In this study, the expression of EZH2, TIMP2, and MMP9 in OC tissues and cell lines was analyzed using The Cancer Genome Atlas (TCGA) database and western blotting, respectively. The effects of SKLB-03220, an EZH2 covalent inhibitor, on OC cell migration and invasion were investigated using wound-healing assays, Transwell assays, and immunohistochemistry. TCGA database analysis confirmed that the EZH2 and MMP9 mRNA expression was significantly higher in OC tissues, whereas TIMP2 expression was significantly lower than that in normal ovarian tissues. Moreover, EZH2 negatively correlated with TIMP2 and positively correlated with MMP9 expression. In addition to the anti-tumor activity of SKLB-03220 in a PA-1 xenograft model, immunohistochemistry results showed that SKLB-03220 markedly increased the expression of TIMP2 and decreased the expression of MMP9. Additionally, wound-healing and Transwell assays showed that SKLB-03220 significantly inhibited the migration and invasion of both A2780 and PA-1 cells in a concentration-dependent manner. SKLB-03220 inhibited H3K27me3 and MMP9 expression and increased TIMP2 expression in PA-1 cells. Taken together, these results indicate that the EZH2 covalent inhibitor SKLB-03220 inhibits metastasis of OC cells by upregulating TIMP2 and downregulating MMP9, and could thus serve as a therapeutic agent for OC.


Subject(s)
Acrylamides , Enhancer of Zeste Homolog 2 Protein , Ovarian Neoplasms , Humans , Female , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Ovarian Neoplasms/genetics , Cell Line, Tumor , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Matrix Metalloproteinase 9/genetics , Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic
3.
Bioorg Chem ; 148: 107427, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38728911

ABSTRACT

Histone acetyltransferase CREB-binding protein (CBP) and its homologous protein p300 are key transcriptional activators that can activate oncogene transcription, which present promising targets for cancer therapy. Here, we designed and synthesized a series of p300/CBP targeted low molecular weight PROTACs by assembling the covalent ligand of RNF126 E3 ubiquitin ligase and the bromodomain ligand of the p300/CBP. The optimal molecule A8 could effectively degrade p300 and CBP through the ubiquitin-proteasome system in time- and concentration-dependent manners, with half-maximal degradation (DC50) concentrations of 208.35/454.35 nM and 82.24/79.45 nM for p300/CBP in MV4-11 and Molm13 cell lines after 72 h of treatment. And the degradation of p300/CBP by A8 is dependent on the ubiquitin-proteasome pathway and its simultaneous interactions with the target proteins and RNF126. A8 exhibits good antiproliferative activity in a series of p300/CBP-dependent cancer cells. It could transcriptionally inhibit the expression of c-Myc, induce cell cycle arrest in the G0/G1 phase and apoptosis in MV4-11 cells. This study thus provided us a new chemotype for the development of drug-like PROTACs targeting p300/CBP, which is expected to be applied in cancer therapy.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Ubiquitin-Protein Ligases , p300-CBP Transcription Factors , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Structure-Activity Relationship , Molecular Structure , Apoptosis/drug effects , Cell Line, Tumor
4.
Chirality ; 34(1): 147-159, 2022 01.
Article in English | MEDLINE | ID: mdl-34749430

ABSTRACT

Chiral resolution of binaphthylamine is often a toilful conundrum in the field of analytical chemistry and biomedicine. The work puts forward a selective, sensitive, and miniaturized analytical method based on molecularly imprinted polymers (MIPs) as adsorbent for miniaturized tip solid-phase extraction (MTSPE) in the separation of binaphthylamine enantiomer. This method combines the advantages of MIPs (high selectivity), MTSPE (low consumption), and high-performance liquid chromatography (HPLC, high sensitivity). A simple synthesis methodology of MIP (P2) was conducted through bulk polymerization with (S)-(-)-1,1'-binaphthyl-2,2'-diamine (S-DABN) as template together with methacrylic acid monomer, and ethylene glycol dimethacrylate as cross-linker in proper porogen, realizing a selective recognition and efficient enrichment for S-DABN. The method exhibited appreciable linearity (0.06-1.00 mg ml-1 ), low quantification limit (0.056 mg ml-1 ), good absolute recoveries (45.70%-69.29%), and high precision (relative standard deviations ≤ 3.54%), along with low consumption (0.50 ml sample solution and 25.0 mg adsorbent). Based on the density functional theory, computational simulation was used to make a preliminary prediction for rational design of MIPs and gave a reasonable elaboration involving the potential mechanism of templates interacting with functional monomers. The adsorption kinetics and thermodynamics were investigated to evaluate the recombination process of substrates. In addition, the selectivity of MIPs for S-DABN was obtained by MIP-MTSPE coupled with HPLC, which supports the feasibility of this convenient design process. The proposed method was employed for selective extraction of S-DABN and exhibited promising potential in the application of chiral analysis.


Subject(s)
Molecular Imprinting , Polymers , Adsorption , Chromatography, High Pressure Liquid , Diamines , Naphthalenes , Solid Phase Extraction , Stereoisomerism
5.
BMC Cancer ; 21(1): 738, 2021 Jun 27.
Article in English | MEDLINE | ID: mdl-34176478

ABSTRACT

BACKGROUND: To investigate the efficacy and safety of interval debulking surgery (IDS) combined with dense hyperthermic intraperitoneal chemotherapy (HIPEC) with cisplatin in Chinese patients with FIGO stage III serous epithelial ovarian cancer (EOC). METHODS: This retrospective single-center study reviewed the demographic and clinical data of 197 patients with primary FIGO stage III serous EOC who were treated with IDS with (n = 121) or without (n = 76, control group) dense HIPEC between January 2012 and April 2017. The co-primary endpoints were progression-free survival (PFS) and overall survival (OS), and the secondary endpoint was the occurrence of adverse events. RESULTS: The median PFS was 24 months in the IDS plus dense HIPEC group, whereas it was 19 months in the IDS alone group (hazard ratio [HR] 0.46, 95% confidence interval [CI]: 0.33-0.65, p = 0.000). The median OS in patients treated with IDS plus dense HIPEC (51 months) was significantly longer than that in patients treated with IDS alone (40 months, HR 0.52, 95% CI: 0.35-0.78, p = 0.001). The demographic and preoperative clinical characteristics of these two groups were comparable (p > 0.05). In the IDS alone group, no adverse events were recorded in 42 (55.3%) of the 76 patients, and 14 (18.4%) patients were reported to have grade III/IV adverse events. In the IDS plus dense HIPEC group, no adverse events were recorded in 55 (45.5%) of the 121 patients, and 23 (19.0%) patients were reported to have grade III/IV adverse events. No postoperative deaths occurred within 30 days in either group and neither did severe fatal complications in the IDS plus dense HIPEC group. CONCLUSIONS: IDS plus dense HIPEC with cisplatin in Chinese patients with FIGO stage III serous EOC is associated with improved survival and is reasonably well tolerated by patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Cisplatin/therapeutic use , Hyperthermic Intraperitoneal Chemotherapy/methods , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Ovarian Epithelial/mortality , Cisplatin/pharmacology , Female , Humans , Neoplasm Staging , Retrospective Studies , Survival Analysis
6.
Bioorg Med Chem Lett ; 30(20): 127479, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32784091

ABSTRACT

Two classes of piperazinone-containing thieno[3,2-d]pyrimidines were designed and synthesized as new PI3Kδ inhibitors in this study. Detailed SAR study with respect to the piperazinone substituents at the 6-position of thieno[3,2-d]pyrimidine core demonstrated that piperazinone-containing thieno[3,2-d]pyrimidines would be more potent and selective for PI3Kδ than their piperazine counterparts, which led to the discovery of several potent PI3Kδ inhibitors with comparable or better antiproliferative activity against a panel of non-Hodgkin lymphoma (NHL) cell lines as compared with idelalisib. Our study will promote the development of new PI3Kδ inhibitors based on piperazinone-containing thieno[3,2-d]pyrimidine scaffold.


Subject(s)
Antineoplastic Agents/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Design , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Class I Phosphatidylinositol 3-Kinases/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/chemistry , Piperazines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
7.
Bioorg Chem ; 105: 104344, 2020 12.
Article in English | MEDLINE | ID: mdl-33091667

ABSTRACT

PI3Kδ has proved to be an effective target for anti-lymphoma drugs. However, the application of current approved PI3Kδ inhibitors has been greatly limited due to their specific immune-mediated toxicity and increased risk of infection, it is necessary to develop more PI3Kδ inhibitors with new scaffold. In this study, SAR study with respect to piperazinone-containing purine derivatives led to the discovery of a potent and selective PI3Kδ inhibitor, 4-(cyclobutanecarbonyl)-1-((2-(2-ethyl-1H-benzo[d]imidazol-1-yl)-9-methyl-6-morpholino-9H-purin-8-yl)methyl)piperazin-2-one (WNY1613). WNY1613 exhibits good antiproliferative activity against a panel of non-Hodgkin's lymphoma (NHL) cell lines by inducing cancer cell apoptosis and inhibiting the phosphorylation of PI3K and MAPK downstream components. In addition, it can also prevent the tumor growth in both SU-DHL-6 and JEKO-1 xenograft models without observable toxicity. WNY1613 thus could be developed as a promising candidate for the treatment of NHL after subsequent extensive pharmacodynamics and pharmacokinetics investigation.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Lymphoma, Non-Hodgkin/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Piperazines/chemistry , Purines/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Enzyme Inhibitors/pharmacology , Heterografts , Humans , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Docking Simulation , Morpholines/chemistry , Neoplasms, Experimental , Phosphorylation , Purines/pharmacology
8.
Mol Pharm ; 16(11): 4582-4593, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31573817

ABSTRACT

Poor uptake of antitumor drugs by tumor cells is a critical challenge for anticancer therapeutics. Moreover, the deficiency of specific tumor selectivity for tumor sites may further limit the therapeutic efficacy and cause side effects in healthy regions of the body. Vincristine (VCR) is an effective antitumor drug; however, because of its severe nerve toxicity, short half-life, and fast metabolism, its clinical application is limited. Herein, novel anti-CD133 monoclonal antibody (CD133mAb)-targeted therapeutic immunomagnetic albumin microbeads (CD133mAb/TMAMbs) are smartly constructed for enhancing antiglioblastoma treatment. Superparamagnetic iron oxide nanoparticles (SPIO NPs) were first fabricated as nanocarrier cores, then encapsulated with human serum albumin (HSA), and loaded antitumor drug VCR. Then CD133mAb, which has specific affinity with the cell membrane CD133, was subsequently conjugated to form CD133mAb-decorated therapeutic immunomagnetic albumin microbeads (CD133mAb/TMAMbs). The influence of CD133mAb/TMAMbs on the viability, cell cycle, apoptosis, cell cytoskeleton, migration, and invasion of CD133-overexpressing U251 cells was explored. The CD133mAb-conjugated magnetic albumin microbeads exhibited a high drug loading capacity, stability and hemocompatibility, and active targeting ability by specific recognition of the CD133 surface antigen by the bioconjugation of CD133mAb. More importantly, the constructed therapeutic CD133mAb/TMAMbs have a specifically effective uptake via the CD133 transmembrane protein that is overexpressed in U251 glioblastoma cells and displayed an effective antitumor proliferation and invasive ability. Therefore, based on these results, the fabricated CD133mAb/TMAMbs demonstrate promising uses in brain cancer-targeted diagnosis and therapy.


Subject(s)
AC133 Antigen/metabolism , Albumins/metabolism , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Vincristine/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Delivery Systems/methods , Glioblastoma/metabolism , Humans , Magnetics/methods , Microspheres , Nanoparticles/chemistry
9.
Mol Divers ; 23(3): 681-696, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30612258

ABSTRACT

The histone lysine methyltransferase EZH2 has been reported to play important roles in cancer aggressiveness, metastasis and poor prognosis. In this study, a series of benzomorpholine derivatives were synthesized and biologically evaluated as EZH2 inhibitors. The target compounds were obtained in good yields from 3-amino-5-bromo-2-hydroxybenzoic acid via cyclization, Suzuki coupling and amidation as the key steps. A preliminary optimization study led to the discovery of several potent novel EZH2 inhibitors (6b, 6c, 6x and 6y). Moreover, 6y inhibited the A549 and NCI-H1975 cell lines (IC50 = 1.1 µM and 1.1 µM, respectively). Further studies indicated that 6y can reduce EZH2 expression in intact cells and cause cell arrest in the G2/M phase.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lung Neoplasms/pathology , Morpholines/chemical synthesis , Morpholines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Morpholines/chemistry , Structure-Activity Relationship
10.
Mol Divers ; 21(1): 125-136, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27858214

ABSTRACT

The bromodomain and extra-terminal proteins (BETs), in particular BRD4, has been reported to play important roles in cancer, inflammation, obesity, cardiovascular disease, and neurological disorders. In this paper, a series of benzomorpholinone derivatives were synthesized and biologically evaluated as BETs inhibitors. Detailed structure-activity relationship studies led to the discovery of several new potent compounds, of which 15h and 15i displayed [Formula: see text] values of 2.8 and 4.5 [Formula: see text] against BRD4 (D1), respectively, and showed good anti-proliferation activities against four hematologic malignancies cell lines at low-micromolar concentrations, including MV4-11, OCI-LY10, Pfeifer, and Su-DHL-6 cells. This chemotype could be further optimized with respect to its potency and drug-like properties in the future.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Hematologic Neoplasms/pathology , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Epigenesis, Genetic , Humans , Protein Domains , Structure-Activity Relationship , Sulfonamides/chemistry , Benzenesulfonamides
11.
Cell Physiol Biochem ; 40(1-2): 297-308, 2016.
Article in English | MEDLINE | ID: mdl-27855372

ABSTRACT

BACKGROUND: Cancer is still a major public health issue worldwide, and new therapeutics with anti-tumor activity are still urgently needed. METHODS: The anti-tumor activity of TH-39, which shows potent anti-proliferative activity against K562 cells with an IC50 of 0.78 µM, was investigated using immunoblot, co-immunoprecipitation, the MTT assay, and flow cytometry. RESULTS: Mechanistically, TH-39 may disrupt the interaction between Hec1 and Nek2 in K562 cells. Moreover, TH-39 inhibited cell proliferation in a concentration- and time-dependent manner by influencing the morphology of K562 cells and inducing G0/G1 phase arrest. G0/G1 phase arrest was associated with down-regulation of CDK2-cyclin E complex and CDK4/6-cyclin D complex activities. Furthermore, TH-39 also induced cell apoptosis, which was associated with activation of caspase-3, down-regulation of Bcl-2 expression and up-regulation of Bax. TH-39 could also decrease mitochondrial membrane potential (Δψm) and increase reactive oxygen species (ROS) accumulation in K562 cells. The results indicated that TH-39 might induce apoptosis via the ROS-mitochondrial apoptotic pathway. CONCLUSION: This study highlights the potential therapeutic efficacy of the anti-cancer compound TH-39 in treatment-resistant chronic myeloid leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cinnamates/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , NIMA-Related Kinases/metabolism , Nuclear Proteins/metabolism , Resting Phase, Cell Cycle/drug effects , Small Molecule Libraries/pharmacology , Thiazoles/pharmacology , Antineoplastic Agents/chemistry , Cell Cycle Proteins/metabolism , Cell Shape/drug effects , Cell Survival/drug effects , Cinnamates/chemical synthesis , Cinnamates/chemistry , Cytoskeletal Proteins , Humans , Hydrogen-Ion Concentration , K562 Cells , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Small Molecule Libraries/chemistry , Thiazoles/chemical synthesis , Thiazoles/chemistry
12.
Bioorg Med Chem Lett ; 25(7): 1373-6, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25754492

ABSTRACT

Tuberculosis (TB) remains a major human health problem. New therapeutic antitubercular agents are urgent needed to control the global tuberculosis pandemic. We synthesized a new series of 4-carbonyl piperazine substituted 1,3-benzothiazin-4-one derivatives and evaluated their anti-mycobacterial activities against Mycobacterium tuberculosis H37Ra as well as their druggabilities. The results showed that most of these derivatives, especially the compounds with simple alkyl side chains, exhibited good antitubercular activities and favorable aqueous solubilities with no obvious cytotoxicity. It suggested that the 4-carbonyl piperazine substituents in benzothiazinone scaffold were well tolerated, in which the compound 8h, with an antitubercular activity of MIC 0.008 µM, exhibited an excellent aqueous solubility of 104 µg/mL, which was 100-fold better than the potent DprE1 inhibitor Comp.1 (BTZ038), also more soluble than PBTZ169.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Piperazines/pharmacology , Thiazines/pharmacology , Tuberculosis/drug therapy , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Chlorocebus aethiops , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Piperazine , Piperazines/chemical synthesis , Piperazines/chemistry , Structure-Activity Relationship , Thiazines/chemical synthesis , Thiazines/chemistry , Vero Cells
13.
Cell Physiol Biochem ; 34(6): 1863-76, 2014.
Article in English | MEDLINE | ID: mdl-25503322

ABSTRACT

BACKGROUND: Breast cancer is the leading cause of cancer death among women worldwide and metastasis is the major cause of treatment failure. Thus, new treatment options for breast cancer, especially, drugs which could prevent metastasis, are pressingly needed. METHODS: In the present study, we designed and synthesized a novel cinnamide derivative, (E)-N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-3-(3,4,5-trimethoxyphenyl)acrylamide (YLT26), which displayed potent inhibitory effects on breast cancer cells. The cell viability, apoptosis-inducing effect and reactive oxygen species (ROS) production were examined in 4T1 cells following treatment with YLT26. Meanwhile, apoptosis-related proteins levels were determined by western blotting. Finally, we evaluated the effects of YLT26 on breast tumor growth, lung metastases in vivo and the infiltration of myeloid-derived suppressor cells (MDSCs) in lung tissue. RESULTS: Our results showed that the proliferation inhibitory effects of YLT26 were correlated with its apoptosis-inducing effect. Exposure to YLT26 induced mitochondrial transmembrane potential (∆Ψm) change, activated caspase-9, and downregulated the Bcl-2 expression, as well as enhanced ROS accumulation in 4T1 cells. Moreover, YLT26 significantly inhibited tumor growth without obvious side effects in the 4T1 tumor-bearing mice model. Immunohistochemistry analyze revealed YLT26 also induced apoptosis in vivo. More importantly, YLT26 also significantly inhibited lung metastases, which may be associated with the reduction of MDSCs. CONCLUSION: The present study suggested that YLT26 could inhibit breast cancer cells proliferation via ROS-mitochondrial apoptotic pathway, delay breast tumor progression, and suppress lung metastases by impacting on the immunologic microenvironment in vivo. © 2015 S. Karger AG, Basel.


Subject(s)
Anilides/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cinnamates/administration & dosage , Lung Neoplasms/drug therapy , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Neoplasm Proteins/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
14.
Cell Physiol Biochem ; 33(4): 933-44, 2014.
Article in English | MEDLINE | ID: mdl-24713812

ABSTRACT

BACKGROUND: Colorectal cancer continues to be one of the most common causes of cancer death, and the poor survival rates and liver metastases at the time of diagnosis urgently need more effective strategy for colorectal cancer treatment. METHODS: The activities of N-(5-bromopyridin-2-yl)-2-((6-(2-chloroacetamido)benzo[d]thiazol-2-yl)thio)acetamide (YLT205), which is a novel small molecule compound synthesized by us, were investigated using MTT assay, flow cytometry, western blotting and mice tumor xenograft models. RESULTS: YLT205 induced apoptosis of human colorectal cell lines in a dose-dependent manner. The occurrence of apoptosis was associated with activation of caspases-9 and -3, down-regulation of Bcl-2 and up-regulation of Bax in HCT116 cells. Moreover, YLT205 disrupted mitochondrial membranes and induced the release of cytochrome c into cytosol. Impaired phosphorylation of p44/42 mitogen-activated protein kinase was also observed while the expression of phosphorylated protein kinase B (Akt) was not affected. Furthermore, in HCT116 and SW620 tumor-bearing nude mice models, YLT205 dose-dependently inhibited tumor growth without obvious adverse effects. Immunohistochemistry analyses revealed YLT205 also induced apoptosis and inhibited tumor cell proliferation in vivo. CONCLUSION: These studies suggested that YLT205 might be a potential drug candidate for human colorectal cancer therapy.


Subject(s)
Acetamides/pharmacology , Apoptosis/drug effects , Benzothiazoles/pharmacology , Colorectal Neoplasms/pathology , Mitochondria/metabolism , Acetamides/chemistry , Acetamides/therapeutic use , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Benzothiazoles/chemistry , Benzothiazoles/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Cytochromes c/metabolism , Down-Regulation , Flavonoids/pharmacology , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Phosphorylation , Transplantation, Heterologous , bcl-2-Associated X Protein/metabolism
15.
BMC Complement Med Ther ; 24(1): 221, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849817

ABSTRACT

AIMS OF THIS STUDY: This study aims to investigate the potential of Huangqin Tang (HQT), a traditional Chinese medicine formulation, in the treatment of breast cancer (BC) through a comprehensive approach integrating network pharmacology, molecular docking, and experimental validation. METHODS: Chemical composition and target information of HQT were collected using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Disease-related target genes were obtained from the GeneCards database. Network pharmacological analysis, including construction of compound-disease-target networks and protein-protein interaction networks, was performed. Molecular docking simulations were conducted to evaluate the binding affinity between HQT components and key targets. Experimental validation was carried out using cell viability assays, clone formation assays, flow cytometry, Western blotting, and pathway analysis. RESULTS: A total of 210 candidate targets were identified. Network analysis revealed STAT3, AKT1, MAPK3 etc. as central targets. Enrichment analysis suggested HQT may exert anti-tumor effects through regulating lipid metabolism and inflammation related pathways. Molecular docking showed that the key compounds baicalein, wogonin, kaempferol and quercetin all bound effectively to MAPK1. The binding of baicalein to IL6 and naringenin to TNF-α was also relatively stable. The experimental results demonstrated that HQT effectively inhibited the proliferation of breast cancer cells, with IC50 values of 2.334 mg/mL and 1.749 mg/mL in MCF-7 cells at 24 h and 48 h, and IC50 values of 1.286 mg/mL and 1.496 mg/mL in MDA-MB-231 cells at 24 h and 48 h, respectively. Furthermore, HQT induced cell cycle arrest at the G2/M phase in breast cancer cells and downregulated the expression of related proteins including CDK1, Cyclin B1, CDK2, and Cyclin E. Additionally, HQT promoted apoptosis in breast cancer cells by upregulating the expression of Bak and CC-3, while downregulating the expression of Bcl-2. Notably, HQT also exhibited regulatory effects on the HIF-1 signaling pathway. CONCLUSIONS: This study provides insights into the potential multi-component and multi-target mechanisms of HQT against BC, suggesting it may achieve therapeutic effects through regulating inflammatory response and cancer-related pathways via the identified active compounds and targets. The findings highlight the importance of integrating traditional medicine with modern approaches for the development of novel cancer therapies.


Subject(s)
Breast Neoplasms , Drugs, Chinese Herbal , Molecular Docking Simulation , Network Pharmacology , Breast Neoplasms/drug therapy , Humans , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Female , MCF-7 Cells , Cell Line, Tumor , Protein Interaction Maps
16.
Life Sci ; 342: 122538, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38428571

ABSTRACT

Pulmonary disorders, including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), pulmonary hypertension (PH), and lung cancer, seriously impair the quality of lives of patients. A deeper understanding of the occurrence and development of the above diseases may inspire new strategies to remedy the scarcity of treatments. Type I protein arginine methyltransferases (PRMTs) can affect processes of inflammation, airway remodeling, fibroblast proliferation, mitochondrial mass, and epithelial dysfunction through substrate methylation and non-enzymatic activity, thus affecting the occurrence and development of asthma, COPD, lung cancer, PF, and PH. As potential therapeutic targets, inhibitors of type I PRMTs are developed, moreover, representative compounds such as GSK3368715 and MS023 have also been used for early research. Here, we collated structures of type I PRMTs inhibitors and compared their activity. Finally, we highlighted the physiological and pathological associations of type I PRMTs with asthma, COPD, lung cancer, PF, and PH. The developing of type I PRMTs modulators will be beneficial for the treatment of these diseases.


Subject(s)
Asthma , Hypertension, Pulmonary , Lung Neoplasms , Pulmonary Disease, Chronic Obstructive , Pulmonary Fibrosis , Humans , Hypertension, Pulmonary/drug therapy , Lung Neoplasms/drug therapy , Asthma/pathology
17.
Bioorg Med Chem Lett ; 23(8): 2293-7, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23489624

ABSTRACT

Inducing apoptosis is a promising therapeutic approach to overcome cancer. In this study, 30 compounds were synthesized and evaluated for their antiproliferative activity against three tumor cell lines in vitro: A875, H460 and Hela cancer cells by the MTT assay. The most potent analogue 7a, a novel compound was first reported by our group, inhibited the proliferation of A875 cells with an IC50 value of 98 nM. Flow cytometry analysis and morphological analysis suggested that compound 7a had potential anticancer efficacy via G2/M cell cycle arrest, which could be attributed to its proliferation and apoptosis, and also in a concentration-dependent manner. The SAR analysis indicated that the substituents R(2) played a crucial role in the antiproliferation activity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Amides/chemical synthesis , Amides/chemistry , Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Growth Processes/drug effects , Cell Line, Tumor , Chlorobenzenes/chemical synthesis , Chlorobenzenes/chemistry , Chlorobenzenes/pharmacology , Drug Screening Assays, Antitumor , Female , HeLa Cells , Hep G2 Cells , Humans , Neoplasms/pathology , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/chemistry , Thiadiazoles/pharmacology
18.
Biochem Pharmacol ; 210: 115493, 2023 04.
Article in English | MEDLINE | ID: mdl-36898415

ABSTRACT

The incidence and mortality rate of malignant melanoma are increasing worldwide. Metastasis reduces the efficacy of current melanoma therapies and leads to poor prognosis for patients. EZH2 is a methyltransferase that promotes the proliferation, metastasis, and drug resistance of tumor cells by regulating transcriptional activity. EZH2 inhibitors could be effective in melanoma therapies. Herein, we aimed to investigate whether the pharmacological inhibition of EZH2 by ZLD1039, a potent and selective S-adenosyl-l-methionine-EZH2 inhibitor, suppresses tumor growth and pulmonary metastasis in melanoma cells. Results showed that ZLD1039 selectively reduced H3K27 methylation in melanoma cells by inhibiting EZH2 methyltransferase activity. Additionally, ZLD1039 exerted excellent antiproliferative effects on melanoma cells in 2D and 3D culture systems. Administration of ZLD1039 (100 mg/kg) by oral gavage caused antitumor effects in the A375 subcutaneous xenograft mouse model. RNA sequencing and GSEA revealed that the ZLD1039-treated tumors exhibited changes in the gene sets enriched from the "Cell Cycle" and "Oxidative Phosphorylation", whereas the "ECM receptor interaction" gene set had a negative enrichment score. Mechanistically, ZLD1039 induced G0/G1 phase arrest by upregulating p16 and p27 and inhibiting the functions of the cyclin D1/CDK6 and cyclin E/CDK2 complexes. Moreover, ZLD1039 induced apoptosis in melanoma cells via the mitochondrial reactive oxygen species apoptotic pathway, consistent with the changes in transcriptional signatures. ZLD1039 also exhibited excellent antimetastatic effects on melanoma cells in vitro and in vivo. Our data highlight that ZLD1039 may be effective against melanoma growth and pulmonary metastasis and thus could serve as a therapeutic agent for melanoma.


Subject(s)
Lung Neoplasms , Melanoma , Skin Neoplasms , Humans , Animals , Mice , Cell Proliferation , Melanoma/genetics , Skin Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Methyltransferases , Cell Line, Tumor , Apoptosis , Enhancer of Zeste Homolog 2 Protein/metabolism
19.
Expert Opin Ther Pat ; 33(4): 293-308, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37095742

ABSTRACT

INTRODUCTION: EZH2 is an important epigenetic regulator that forms the PRC2 complex with SUZ12, EED and RbAp46/48. As the key catalytic subunit of PRC2, EZH2 regulates the trimethylation of histone H3K27, which in turn promotes chromatin condensation and represses the transcription of relevant target genes. EZH2 overexpression and mutations are strictly related to tumor proliferation, invasion and metastasis. Currently, a large number of highly specific EZH2 inhibitors have been developed and some have already been in clinical trials. AREAS COVERED: The aim of the present review is to provide an overview of the molecular mechanisms of EZH2 inhibitors and to highlight the research advances in the patent literature published from 2017 to date. A search of the literature and patents for EZH2 inhibitors and degraders was performed using the Web of Science, SCIFinder, WIPO, USPTO, EPO and CNIPA databases. EXPERT OPINION: In recent years, a great number of structurally diverse EZH2 inhibitors have been identified, including EZH2 reversible inhibitors, EZH2 irreversible inhibitors, EZH2-based dual inhibitors and EZH2 degraders. Despite the multiple challenges, EZH2 inhibitors offer promising potential for the treatment of various diseases, such as cancers.


Subject(s)
Neoplasms , Humans , Enhancer of Zeste Homolog 2 Protein/genetics , Enzyme Inhibitors , Neoplasms/drug therapy , Neoplasms/genetics , Patents as Topic
20.
Eur J Med Chem ; 245(Pt 1): 114887, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36370549

ABSTRACT

Monopolar spindle kinase 1 (Mps1), a core component of the spindle assembly checkpoint (SAC), plays a crucial role in the transition of cells from mid-to late mitosis. As an attractive therapeutic target, inhibition of Mps1 induces cell cycle arrest and apoptosis in a variety of tumors, including breast cancer. However, early clinical development of Mps1 inhibitors remains unsatisfactory. Here, we designed and synthesized a new class of Mps1 inhibitors with 7H-pyrrolo[2,3-d]pyrimidine structure using a scaffold hopping approach. Structure-activity relationship (SAR) revealed that 12 is a potent Mps1 inhibitor (IC50 = 29 nM), which inhibited phosphorylation of Mps1 in vitro and in vivo. Treatment with 12 not only impeded proliferation of breast cancer cell lines, but also induced cell cycle arrest and apoptosis of MCF-7 and 4T1 cells. 12 suppressed tumor growth in vivo, and no obvious toxicities were observed. These results demonstrated the potential of Mps1 inhibitor 12 for the treatment of breast cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Pyrimidines , Female , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cell Cycle Proteins , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Cell Line, Tumor , Drug Design
SELECTION OF CITATIONS
SEARCH DETAIL