Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.795
Filter
1.
Life Sci ; 348: 122680, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38697280

ABSTRACT

AIMS: Hepatocellular carcinoma (HCC) is still a leading cause of cancer-related death worldwide. But its chemotherapeutic options are far from expectation. We here compared H-ras targeted genetic therapy to a commercial docetaxel formulation (DXT) in inhibiting HCC in rats. MAIN METHODS: After the physicochemical characterization of phosphorothioate-antisense oligomer (PS-ASO) against H-ras mutated gene, the PS-ASO-mediated in vitro hemolysis, in vivo hepatic uptake, its pharmacokinetic profile, tissue distribution in some highly perfused organs, its effect in normal rats, antineoplastic efficacy in carcinogen-induced HCC in rats were evaluated and compared against DXT treatment. Mutated H-ras expression by in situ hybridization, hep-par-I, CK-7, CD-15, p53 expression patterns by immunohistochemical methods, scanning electron microscopic evaluation of hepatic architecture, various hepatic marker enzyme levels and caspase-3/9 apoptotic enzyme activities were also carried out in the experimental rats. KEY FINDINGS: PS-ASO showed low in vitro hemolysis (<3 %), and had a sustained PS-ASO blood residence time in vivo compared to DTX, with a time-dependent hepatic uptake. It showed no toxic manifestations in normal rats. PS-ASO distribution was although initially less in the lung than liver and kidney, but at 8 h it accumulated more in lung than kidney. Antineoplastic potential of PS-ASO (treated for 6 weeks) excelled in inhibiting chemically induced tumorigenesis compared to DTX in rats, by inhibiting H-ras gene expression, some immonohistochemical modulations, and inducing caspase-3/9-mediated apoptosis. It prevented HCC-mediated lung metastatic tumor in the experimental rats. SIGNIFICANCE: PS-ASO genetic therapy showed potential to inhibit HCC far more effectively than DXT in rats.


Subject(s)
Antineoplastic Agents , Docetaxel , Genetic Therapy , Animals , Docetaxel/pharmacology , Rats , Male , Genetic Therapy/methods , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Apoptosis/drug effects , Rats, Sprague-Dawley , Taxoids/pharmacology
2.
Drug Des Devel Ther ; 18: 1321-1338, 2024.
Article in English | MEDLINE | ID: mdl-38681206

ABSTRACT

Purpose: Cinobufotalin injection has obvious curative effects on liver cancer patients with less toxicity and fewer side effects than other therapeutic approaches. However, the core ingredients and mechanism underlying these anti-liver cancer effects have not been fully clarified due to its complex composition. Methods: Multidimensional network analysis was used to screen the core ingredients, key targets and pathways underlying the therapeutic effects of cinobufotalin injection on liver cancer, and in vitro and in vivo experiments were performed to confirm the findings. Results: By construction of ingredient networks and integrated analysis, eight core ingredients and ten key targets were finally identified in cinobufotalin injection, and all of the core ingredients are tightly linked with the key targets, and these key targets are highly associated with the cell cycle-related pathways, supporting that both cinobufotalin injection and its core ingredients exert anti-liver cancer roles by blocking cell cycle-related pathways. Moreover, in vitro and in vivo experiments confirmed that either cinobufotalin injection or one of its core ingredients, cinobufagin, significantly inhibited cell proliferation, colony formation, cell cycle progression and xenograft tumor growth, and the key target molecules involved in the cell cycle pathway such as CDK1, CDK4, CCNB1, CHEK1 and CCNE1, exhibit consistent changes in expression after treatment with cinobufotalin injection or cinobufagin. Interestingly, some key targets CDK1, CDK4, PLK1, CHEK1, TTK were predicted to bind with multiple of core ingredients of cinobufotalin injection, and the affinity between one of the critical ingredients cinobufagin and key target CDK1 was further confirmed by SPR assay. Conclusion: Cinobufotalin injection was confirmed to includes eight core ingredients, and they play therapeutic effects in liver cancer by blocking cell cycle-related pathways, which provides important insights for the mechanism of cinobufotalin injection antagonizing liver cancer and the development of novel small molecule anti-cancer drugs.


Subject(s)
Antineoplastic Agents , Bufanolides , Cell Proliferation , Liver Neoplasms , Bufanolides/pharmacology , Bufanolides/chemistry , Bufanolides/administration & dosage , Humans , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Cell Proliferation/drug effects , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/metabolism , Mice, Inbred BALB C , Cell Cycle/drug effects , Mice, Nude , Dose-Response Relationship, Drug , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/metabolism , Tumor Cells, Cultured , Structure-Activity Relationship , Molecular Structure , Injections
3.
Arch Toxicol ; 96(6): 1829-1843, 2022 06.
Article in English | MEDLINE | ID: mdl-35267068

ABSTRACT

Farnesoid X receptor (FXR) plays an indispensable role in liver homeostasis and has been a promising drug target for hepatic diseases. However, the concerns of undesired biological actions limit the clinical applications of FXR agonists. To reveal the intrinsic mechanism of FXR agonist-induce hepatotoxicity, two typical FXR agonists with different structures (obeticholic acid (OCA) and Px-102) were investigated in the present study. By detecting MMP, ROS, and ATP and analyzing the fate of cells, we found that both OCA and Px-102 reduced the mitochondrial function of hepatocytes and promoted cell apoptosis. Gene ablation or inhibition of FXR or SHP ameliorated the cytotoxicities of OCA and Px-102, which indicated the adverse actions of FXR/SHP activation including down-regulation of phosphorylation of PI3K/AKT and functional hepatic genes. The dose-related injurious effects of OCA (10 mg/kg and 30 mg/kg) and Px-102 (5 mg/kg and 15 mg/kg) on the liver were confirmed on a high-fat diet mouse model. The decrease of hepatocyte-specific genes and augmenter of liver regeneration in the liver caused by OCA or Px-102 suggested an imbalance of liver regeneration and a disruption of hepatic functions. Exploration of intestinally biased FXR agonists or combination of FXR agonist with apoptosis inhibitor may be more beneficial strategies for liver diseases.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Liver Neoplasms, Experimental , Oxazoles , Receptors, Cytoplasmic and Nuclear , Animals , Apoptosis/drug effects , Chenodeoxycholic Acid/pharmacology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Oxazoles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects
4.
J Enzyme Inhib Med Chem ; 37(1): 844-856, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35296193

ABSTRACT

A novel series of aminotrimethylpyridinol and aminodimethylpyrimidinol derivatives were designed and synthesised for FGFR4 inhibitors. Structure-activity relationship on the FGFR4 inhibitory activity of the new compounds was clearly elucidated by an intensive molecular docking study. Anti-cancer activity of the compounds was evaluated using hepatocellular carcinoma (HCC) cell lines and a chick chorioallantoic membrane (CAM) tumour model. Compound 6O showed FGFR4 inhibitory activity over FGFR1 - 3. Compared to the positive control BLU9931, compound 6O exhibited at least 8 times higher FGFR4 selectivity. Strong anti-proliferative activity of compound 6O was observed against Hep3B, an HCC cell line which was a much more sensitive cell line to BLU9931. In vivo anti-tumour activity of compound 6O against Hep3B-xenografted CAM tumour model was almost similar to BLU9931. Overall, compound 6O, a novel derivative of aminodimethylpyrimidinol, was a selective FGFR4 kinase inhibitor blocking HCC tumour growth.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Design , Liver Neoplasms/drug therapy , Pyridines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Chickens , Dose-Response Relationship, Drug , Humans , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Carbohydr Polym ; 278: 118950, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34973766

ABSTRACT

Cancer is a complex disease, and blocking tumor angiogenesis has become one of the most promising approaches in cancer therapy. Here, an exopoly heteropolysaccharide (AQP70-2B) was firstly isolated from Akebia quinata. Monosaccharide composition indicated that the AQP70-2B was composed of rhamnose, glucose, galactose, and arabinose. The backbone of AQP70-2B consisted of →1)-l-Araf, →3)-l-Araf-(1→, →5)-l-Araf-(1→, →3,5)-l-Araf-(1→, →2,5)-l-Araf-(1→, →4)-d-Glcp-(1→, →6)-d-Galp-(1→, and →1)-d-Rhap residues. Based on the close relationship between selenium and anti-tumor activity, AQP70-2B was modified with selenium to obtain selenized polysaccharide Se-AQP70-2B. Then, a series of methods for analysis and characterization, especially scanning electron microscopy coupled with energy dispersive spectrometry (SEM-EDS), indicated that Se-AQP70-2B was successfully synthesized. Furthermore, zebrafish xenografts and anti-angiogenesis experiments indicated that selenization could improve the antitumor activity by inhibiting tumor cell proliferation and migration and blocking angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Fruit/chemistry , Neovascularization, Pathologic/drug therapy , Polysaccharides/pharmacology , Ranunculales/chemistry , Selenium/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/isolation & purification , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Carbohydrate Conformation , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Zebrafish/embryology
6.
ACS Appl Mater Interfaces ; 14(1): 404-416, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-34962752

ABSTRACT

Discoid-reconstituted high-density lipoprotein (d-rHDL) is advantageous for tumor-targeted drug delivery due to its small size, long circulation, and efficient internalization into cancer cells. Nevertheless, an allosteric reaction catalyzed by serum lecithin-cholesterol acyltransferase (LCAT) may cause drug leakage from d-rHDL and reduce its targeting efficiency. Conversely, similar "structural weakening" catalyzed by acyl-coenzyme A-cholesterol acyltransferase (ACAT) inside tumor cells can stimulate precise intracellular drug release. Therefore, we synthesized and characterized a pH-sensitive n-butyraldehyde bi-cholesterol (BCC) to substitute for cholesterol in the d-rHDL particle, and bovine serum albumin (BSA) was used as the targeting agent. This dual pH- and ACAT-sensitive d-rHDL (d-d-rHDL) was small with a disk-like appearance. Morphological transformation observation, in vitro release assays, and differences in internalization upon LCAT treatment confirmed that BCC effectively inhibited the remodeling behavior and enhanced the tumor-targeting efficiency. The accumulation of d-d-rHDL in HepG2 cells was significantly higher than that in LO2 cells, and accumulation was inhibited by free BSA. The pH sensitivity was verified, and d-d-rHDL achieved efficient drug release in vitro and inside tumor cells after exposure to acidic conditions and ACAT. Confocal laser scanning microscopy demonstrated that d-d-rHDL escaped from lysosomes and became distributed evenly throughout cells. Moreover, in vivo imaging assays in a tumor-bearing mouse model demonstrated tumor-targeting properties of d-d-rHDL, and paclitaxel-loaded d-d-rHDL showed strong anticancer activity in these mice. This dual-sensitive d-d-rHDL thus combines structural stability in plasma and an intracellular pH/ACAT-triggered drug release to facilitate inhibition of tumor growth.


Subject(s)
Antineoplastic Agents/pharmacology , Biocompatible Materials/pharmacology , Lipoproteins, HDL/pharmacology , Phosphatidylcholine-Sterol O-Acyltransferase/chemistry , Sterol O-Acyltransferase/chemistry , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Screening Assays, Antitumor , Humans , Hydrogen-Ion Concentration , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Materials Testing , Mice , Molecular Structure , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase/metabolism
7.
Bioorg Chem ; 119: 105509, 2022 02.
Article in English | MEDLINE | ID: mdl-34844768

ABSTRACT

Hepatocellular carcinoma (HCC), the most prevalent liver cancer, is considered one of the most lethal malignancies with a dismal outcome. There is an urgent need to find novel therapeutic approaches to treat HCC. At present, natural products have served as a valuable source for drug discovery. Here, we obtained five known biflavones from the root of Stellera chamaejasme and evaluated their activities against HCC Hep3B cells in vitro. Chamaejasmenin E (CE) exhibited the strongest inhibitory effect among these biflavones. Furthermore, we found that CE could suppress the cell proliferation and colony formation, as well as the migration ability of HCC cells, but there was no significant toxicity on normal liver cells. Additionally, CE induced mitochondrial dysfunction and oxidative stress, eventually leading to cellular apoptosis. Mechanistically, the potential target of CE was predicted by database screening, showing that the compound might exert an inhibitory effect by targeting at c-Met. Next, this result was confirmed by molecular docking, cellular thermal shift assay (CETSA), as well as RT-PCR and Western blot analysis. Meanwhile, CE also reduced the downstream proteins of c-Met in HCC cells. In concordance with above results, CE is efficacious and non-toxic in tumor xenograft model. Taken together, our findings revealed an underlying tumor-suppressive mechanism of CE, which provided a foundation for identifying the target of biflavones.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biflavonoids/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Plant Extracts/pharmacology , Protein Kinase Inhibitors/pharmacology , Thymelaeaceae/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Biflavonoids/chemistry , Biflavonoids/isolation & purification , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Structure-Activity Relationship
8.
Life Sci ; 289: 120192, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34871664

ABSTRACT

AIMS: The number of cancer survivors with cardiovascular disease is increasing. However, the effect of cancer on body fluid regulation remains to be clarified. In this study, we evaluated body osmolyte and water imbalance in rats with hepatocellular carcinoma. MAIN METHODS: Wistar rats were administered diethylnitrosamine, a carcinogenic drug, to establish liver cancer. We analyzed tissue osmolyte and water content, and their associations with aldosterone secretion. KEY FINDINGS: Hepatocellular carcinoma rats had significantly reduced body mass and the amount of total body sodium, potassium, and water. However, these rats had significantly increased relative tissue sodium, potassium, and water content per tissue dry weight. Furthermore, these changes in sodium and water balance in hepatocellular carcinoma rats were significantly associated with increased 24-h urinary aldosterone excretion. Supplementation with 0.25% salt in drinking water improved body weight reduction associated with sodium and water retention in hepatocellular carcinoma rats, which was suppressed by treatment with spironolactone, a mineralocorticoid receptor antagonist. Additionally, the urea-driven water conservation system was activated in hepatocellular carcinoma rats. SIGNIFICANCE: These findings suggest that hepatocellular carcinoma induces body mass loss in parallel with activation of the water conservation system including aldosterone secretion and urea accumulation to retain osmolyte and water. The osmolyte and water retention at the tissue level may be a causative factor for ascites and edema formation in liver failure rats.


Subject(s)
Aldosterone/urine , Carcinoma, Hepatocellular/urine , Diethylnitrosamine/toxicity , Liver Neoplasms, Experimental/urine , Water-Electrolyte Balance , Weight Loss , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Rats , Rats, Inbred WKY , Receptors, Mineralocorticoid/metabolism , Spironolactone/pharmacology
9.
Eur J Med Chem ; 227: 113897, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34649064

ABSTRACT

Microtubule target agents (MTAs) are widely-used clinical anti-cancer drugs for decades, but the acquired drug resistance severely restricted their application. Thioredoxin reductases (TrxR) was reported to be overexpressed in most tumors and closely related to high risk of cancer recurrence and drug resistance, making it a potential target for anticancer drug discovery. Multi-target-directed ligands (MTDLs) by a single molecule provide a logical and alternative approach to drug combinations. In this work, based on the structure-activity relationships obtained in our previous study, some structure modifications were performed. On one hand, the retained skeleton structure of MTAs endowed its tubulin polymerization inhibition activity, on the other hand, the selenium-containing structure and α,ß-unsaturated ketone moiety endowed the TrxR inhibition activity. As results, the newly obtained compounds exhibited superior anti-proliferative activities towards various human cancer cells and drug-resistance cells, and displayed high selectivity towards various human normal cells. The mechanism study revealed that the dual effect of cell cycle arrest triggered by targeting tubulin and the abnormal accumulation of ROS caused by TrxR inhibition eventually lead to cell apoptosis. Notably, compared with the MTA agents CA-4P, and the TrxR inhibitor Ethaselen, the optimized compound 14c, which served as dual-targeting inhibitor of tubulin and TrxR, exerted greatly improved in vivo anti-tumor activity. In summary, 14c deserved further consideration for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcones/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Tubulin Modulators/pharmacology , Tubulin/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indoles/chemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Structure-Activity Relationship , Thioredoxin-Disulfide Reductase/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
10.
Carbohydr Polym ; 275: 118714, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34742439

ABSTRACT

Chemo-photothermal combination therapy has great promise for enhanced tumor treatment. Hereby, we developed a complex nanoparticle using electrostatic absorption method, in which the inner chitosan (CS) NPs loaded polypyrrole (PPy) nanoparticles and 5-fluorouracil (5Fu), the outer shell was carboxymethyl cellulose (CMC) crosslinked with disulfide. The drug loaded polysaccharide complex nanoparticles displayed good photothermal effects, and the drug release would be triggered by multi-model response of NIR irradiation, high glutathione (GSH) and weak acidity in tumor environment. In vitro biological studies indicated the nanopartiles could be effectively internalized by HepG2 cancer cells. Moreover, the remarkable inhibition of the CMC complex PPy and 5Fu loaded CS nanoparticles (CMC/CS@PPy + 5Fu NPs) against tumor growth was achieved in HepG2-bearing mice model, suggesting its great potential for synergetic chemo-photothermal therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carboxymethylcellulose Sodium/chemistry , Chitosan/analogs & derivatives , Fluorouracil/pharmacology , Nanoparticles/chemistry , Photothermal Therapy , Animals , Antimetabolites, Antineoplastic/chemistry , Carbohydrate Conformation , Cell Proliferation/drug effects , Cell Survival/drug effects , Chitosan/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Fluorouracil/chemistry , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Particle Size
11.
Appl Biochem Biotechnol ; 194(1): 368-381, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34792748

ABSTRACT

Biosynthesized silver nanoparticles have a wide range of biological activities and using nanoparticles as one of the novel approaches in cancer therapy. In this present research work, the anti-cancer efficacy of Cucumis melo fruit extract and its silver nanoparticles was explored. Wistar rats were divided into six groups and hepatic cancer was induced with 0.01% DEN (diethylnitrosamine) through drinking water for 16 weeks. Cyclophosphamide was given as the standard drug at the dose of 50 mg/kg body weight. Hematological parameters showed a decrease in the levels of hemoglobin (Hb), packed cell volume (PCV), red blood cells (RBC), mean corpuscular volume (MCV), mean corpuscular Hb (MCH), mean corpuscular Hb concentration (MCHC), and platelets (PLTS) levels except white blood cell (WBC) in DEN-induced cancer animals. Significant alterations in the hematological parameters were observed after treatment which indicate the protective effect of Cucumis melo fruit on the hemopoietic system. The structural integrity of the cells has been damaged in cancer-induced animals, and this results in cytoplasmic leakage of enzyme into the blood stream, leads to the elevated levels of these enzymes in blood with subsequent fall in the tissues. Hence, the levels of liver function markers such as AST ALT, ALP, LDH, GGT, and 5'NT were significantly elevated in serum and the liver of cancer-induced rats. The levels of serum tumor markers, viz., alpha-fetoprotein (AFP) and carcinoembryonic antigen (CEA), elevated in rats induced with DEN, which then were reduced following Cucumis melo fruit treatment, indicating the anti-cancer activity of the drug. Histological evaluation of the liver and kidney was also performed to authenticate the present work. Treatment with crude extract and silver nanoparticles of Cucumis melo fruit indicates that Cucumis melo fruit could have exerted its protective effect.


Subject(s)
Carcinoma, Hepatocellular , Cucumis melo/chemistry , Diethylnitrosamine/toxicity , Fruit/chemistry , Liver Neoplasms, Experimental , Metal Nanoparticles , Plant Extracts/chemistry , Silver , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Rats , Rats, Wistar , Silver/chemistry , Silver/pharmacology
12.
ChemMedChem ; 17(4): e202100517, 2022 02 16.
Article in English | MEDLINE | ID: mdl-34806333

ABSTRACT

Small-molecule inhibitors of MDM2 that block the MDM2-p53 protein-protein interaction have been considered as potential therapeutic agents for the treatment of cancer. Here, we identify five highly potent inhibitors of MDM2 (termed as WY 1-5) that display significant inhibitory effects on MDM2-p53 interaction by using a combined strategy of pharmacophore modeling, virtual screening, and molecular docking studies. Among them, WY-5 is the most active MDM2 inhibitor with an IC50 value of 14.1±2.8 nM. Moreover, WY-5 significantly up-regulate the protein level of p53 in SK-Hep-1 cells harboring wild-type p53. In vitro anticancer study reveals that WY-5 markedly inhibits the survival of SK-Hep-1 cells. In vivo anticancer study suggests that WY-5 significantly inhibits the growth of SK-Hep-1 cells-derived xenograft in nude mice, with no observable toxicity. Our results demonstrate that WY-5 may be a promising candidate for the treatment of cancer harboring wild-type p53.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Structure , Proto-Oncogene Proteins c-mdm2/metabolism , Structure-Activity Relationship
13.
Carbohydr Polym ; 277: 118827, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893244

ABSTRACT

pH-Responsive nanoparticles (NPs) have emerged as an effective antitumor drug delivery system, promoting the drugs accumulation in the tumor and selectively releasing drugs in tumoral acidic microenvironment. Herein, we developed a new amphiphilic modified hydroxyethyl starch (HES) based pH-sensitive nanocarrier of antitumor drug delivery. HES was first modified by hydrophilic imidazole and hydrophobic cholesterol to obtain an amphiphilic polymer (IHC). Then IHC can self-assemble to encapsulate doxorubicin (DOX) and form doxorubicin-loaded nanoparticles (DOX/IHC NPs), which displayed good stability for one week storage and acidic sensitive long-term sustained release of DOX. As a result, cancer cell endocytosed DOX/IHC NPs could continuously release doxorubicin into cytoplasm and nucleus to effectively kill cancer cells. Additionally, DOX/IHC NPs could be effectively enriched in the tumor tissue, showing enhanced tumor growth inhibition effect compared to free doxorubicin. Overall, our amphiphilic modified HES-based NPs possess a great potential as drug delivery system for cancer chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cholesterol/chemistry , Doxorubicin/pharmacology , Hydroxyethyl Starch Derivatives/chemistry , Imidazoles/chemistry , Nanoparticles/chemistry , Surface-Active Agents/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Particle Size , Tumor Cells, Cultured
14.
Carbohydr Polym ; 277: 118891, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893293

ABSTRACT

When organic polymer-based drug nanocarriers become concentrated in macrophages, their influence on macrophage polarization has been rarely reported. This study prepared chitosan-based nanoparticles (CNs, 181.5 nm, +14.83 mV) and detected their impacts on macrophage reprogram. RT-PCR results showed in M1-like RAW264.7 cells (Mφ1), CNs decreased CD86 and iNOS expressions by 53.8% and 57.1%, and increased Arg-1 and IL-10 by 642.9% and 102.1%; in M2-like cells (Mφ2), CNs reduced Arg-1 and MR expressions by 70.7% and 93.0%, but increased CD86, iNOS and TNF-α by 290.4%, 86.2% and 728.6%; these results, consistent with cytokine secretions and surface CD86/CD206 expressions, showed CNs polarized Mφ1 and Mφ2 toward opposite type so as to improve the macrophage polarization homeostasis. In CCl4-induced mouse liver injury model, CNs reduced the hepatic Mφ1/Mφ2 ratio from 1.1 (model group) to 0.3, and then reduced the serum AST and ALT level by 42.3% and 39.0%; in mouse model of hepatocellular carcinoma, CNs decreased the number of CD163-positive cells and increased CD86-positive ones in tumor, and subsequently inhibited the tumor growth and metastasis. This study suggests CNs can improve the phenotype homeostasis of macrophages and subsequently promote the treatment of certain diseases such as liver injury and tumor.


Subject(s)
Antineoplastic Agents/pharmacology , Chitosan/pharmacology , Macrophages/drug effects , Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemistry , Cells, Cultured , Chitosan/administration & dosage , Chitosan/chemistry , Homeostasis/drug effects , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Macrophage Activation/drug effects , Macrophages/metabolism , Mice , Particle Size , Phenotype , RAW 264.7 Cells
15.
Carbohydr Polym ; 277: 118892, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893294

ABSTRACT

Herein, a pH-responsive cyclodextrin derivative (R6H4-CMßCD) with cell-penetrating ability was successfully synthesized, and curcumin-loaded nanoparticles (R6H4-CMßCD@CUR NPs, RCCNPs) were developed to improve its efficacy in hepatoma. RCCNPs could improve the cell uptake compared with CMßCD@CUR NPs (CCNPs) and were internalized into cells mainly through endocytosis mediated by reticulin and macropinocytosis. Furthermore, the accumulation of RCCNPs in hepatoma cells at pH 6.4 was higher than that at pH 7.4, indicating a pH-responsive uptake. Additionally, RCCNPs could escape from the lysosomes via the "proton sponge effect", and a high apoptosis rate was detected. Importantly, in vivo experiments revealed that orally administered RCCNPs could exert excellent anti-cancer effects in tumor-bearing mice. Hematoxylin-eosin staining did not show significant histological changes in the major organs. Thus, our findings indicate the potential of R6H4-CMßCD as a nanopharmaceutical material, and RCCNPs as an effective delivery system for oral curcumin in cancer management.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Cyclodextrins/chemistry , Nanoparticles/chemistry , Peptides/chemistry , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Curcumin/administration & dosage , Drug Screening Assays, Antitumor , Humans , Hydrogen-Ion Concentration , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred ICR
16.
J Ethnopharmacol ; 286: 114930, 2022 Mar 25.
Article in English | MEDLINE | ID: mdl-34952190

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Aidi injection (ADI), a traditional chinese medicine preparation, is widely used in combination with chemotherapy for the treatment of various malignant tumors, such as hepatocellular carcinoma (HCC). Studies have shown that changes in cytochrome P450 (CYP450) activity in disease states would affect the metabolism of drugs in vivo, especially liver diseases. However, the changes of Aidi injection on the activities of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 in normal and HCC states are still unknown. AIM OF THE STUDY: The cocktail probe drugs method was used to investigate the effects of ADI on the activity of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 in normal and HCC rats. MATERIALS AND METHODS: The HCC rats was induced by diethylnitrosamine (DEN). Then, both normal and HCC rats were randomly divided into 2 groups (n = 6). They were given saline or ADI (10 mL/kg/d, i.p) for 2 weeks, respectively. On the fifteenth day, cocktail probe mixing solution, including metoprolol (10 mg/kg), caffeine (1.0 mg/kg), omeprazole (2.0 mg/kg), midazolam (2.0 mg/kg), chlorzoxazone (4.0 mg/kg) and tolbutamide (0.5 mg/kg), was injected into tail vein of all rats in each group. The blood sample was obtained at specified time. After the protein is precipitated, six probe drugs are analyzed by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). RESULTS: Compared with control group, the activity of CYP3A2 and CYP2E1 was significantly lower in the ADI group. Compared with the model group, the activities of CYP1A2, CYP3A2, CYP2E1, and CYP2C11 enzymes in the ADI model group were significantly reduced. Additionally, the activity of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 enzymes in model group was significantly lower than control group. CONCLUSIONS: ADI can inhibit a lot of CYP450 enzyme, so it may reduce the dosage of chemotherapeutic drugs to reach the required plasma concentration of chemotherapeutic drugs, which is of great significance for the combination of anti-tumor chemotherapeutic drugs and is worthy of further in-depth study and clinical attention.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Drugs, Chinese Herbal/pharmacology , Liver Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/enzymology , Chromatography, Liquid , Cytochrome P-450 Enzyme System/drug effects , Diethylnitrosamine , Herb-Drug Interactions , Liver Neoplasms, Experimental/enzymology , Male , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
17.
Eur J Med Chem ; 228: 114037, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34883290

ABSTRACT

Off-target toxicity is one of the main challenges faced by anticancer chemotherapeutics. For tumor targeted and precision chemotherapy, we take the advantages of the ligand directed tumor active targeting of small molecule drug conjugates (SMDCs) and the passive tumor targeting of nanoparticles via the enhanced penetration and retention (EPR) effects, put forward a branched small molecule drug conjugate (BSMDC) nanomedicine design concept. In a proof of concept, we used pentaerythritol as the branched moiety, galactosamine (GalN) as the hepatocellular carcinoma (HCC) directing ligands, PTX as a payload, and a stearoyl moiety as the amphiphilic property adjusting group, designed and synthesized BSMDC 1 and prepared its NPs. In cellular level, the BSMDC 1 NPs targeted asialoglycoprotein receptor (ASGPR)-overexpressing HepG2 cells, were effectively taken up in the cells and released in tumor microenvironments, inhibited the HepG2 cell proliferation, arrested HepG2 cell in G2/M phase and induced tumor cell apoptosis. In HepG2 xenograft nude mice, the BSMDC 1 NPs were high specific to target the tumor and demonstrated a higher antitumor efficiency than BSMDC 1, having no apparent influences on mice body weights and major organs, supporting our BSMDC nanomedicine design concept. Therefore, this new strategy may find applications for cancer targeted and precision chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Galactosamine/pharmacology , Liver Neoplasms/drug therapy , Paclitaxel/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Galactosamine/chemistry , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Nude , Molecular Structure , Nanomedicine , Paclitaxel/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship
18.
ACS Appl Mater Interfaces ; 13(50): 59787-59802, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34894664

ABSTRACT

Developing intelligent and morphology-transformable nanomaterials that can spatiotemporally undergo stimulus-responsive size transformation holds great promise for improving the tumor delivery efficiency of drugs in vivo. Here, we report a smart size-transformable theranostic probe Ce6-Leu consisting of a leucine amino peptidase (LAP) and glutathione (GSH) dual-responsive moiety, an 1,2-aminothiol group, and a clinically used photosensitizer Ce6. This probe tends to self-assemble into uniform nanoparticles with an initial size of ∼80 nm in aqueous solution owing to the amphiphilic feature. Surprisingly, taking advantage of the biocompatible CBT-Cys condensation reaction, the large nanoprobes can be transformed into tiny nanoparticles (∼23 nm) under the joint action of LAP and GSH in a tumor microenvironment, endowing them with great tumor accumulation and deep tissue penetration. Concomitantly, this LAP/GSH-driven disassembly and size shrinkage of Ce6-Leu can also activate the fluorescence/magnetic resonance signals and the photodynamic effect for enhanced multimodal imaging-guided photodynamic therapy of human liver HepG2 tumors in vivo. More excitingly, the Mn2+-chelating probe (Ce6-Leu@Mn2+) was demonstrated to have the capability to catalyze endogenous H2O2 to persistently release O2 at the hypoxic tumor site, as a consequence improving the oxygen supply to boost the radiotherapy effect. We thus believe that this LAP/GSH-driven size-transformable nanosystem would offer a novel advanced technology to improve the drug delivery efficiency for achieving precise tumor diagnosis and treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Biocompatible Materials/pharmacology , Glutathione/metabolism , Leucyl Aminopeptidase/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacology , Theranostic Nanomedicine , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorophyllides/chemistry , Chlorophyllides/pharmacology , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Materials Testing , Molecular Structure , Optical Imaging , Particle Size , Photosensitizing Agents/chemistry
19.
Drug Des Devel Ther ; 15: 4885-4899, 2021.
Article in English | MEDLINE | ID: mdl-34880599

ABSTRACT

OBJECTIVE: Inhibition of tumor metastasis is a useful strategy to improve the efficacy of cancer therapy. Ventilagolin, a natural 1, 4-naphthoquinone derivative extracted from Ventilago leiocarpa Benth, has shown promising antitumor effects in previous studies. However, the effects and underlying mechanisms of Ventilagolin against migration, invasion and epithelial-mesenchymal transition (EMT) of hepatocellular carcinoma (HCC) remain unclear. The present study has examined these effects and determined whether the proto-oncogene Pim-1 is involved. METHODS: The effects of Ventilagolin on migration, invasion, Pim-1 and EMT-related proteins (eg, E-cadherin, N-cadherin, Vimentin) expression were assessed by scratch wound healing, Transwell, qRT-PCR and Western blot assays, respectively. Pim-1 stably overexpressed HepG2 and SMMC-7721 cells were generated to explore whether Ventilagolin inhibited migration, invasion and EMT of HCC cells via regulating Pim-1. Subcutaneous xenograft tumor model in nude mice was established. Histopathological changes of tumor tissues were examined by H&E staining and expressions of Pim-1 and EMT-related proteins were detected by immunohistochemistry. RESULTS: Ventilagolin significantly (P < 0.01) reduced the expression of Pim-1 levels in HepG2 and SMMC-7721 cells. Compared with the control group, the migration and invasion abilities of Pim-1-overexpressing HepG2 and SMMC-7721 cells were significantly (P < 0.05, P < 0.01) enhanced, the expression of E-cadherin was decreased (P < 0.01), and the levels of N-cadherin and Vimentin were upregulated (P < 0.05, P < 0.01). Ventilagolin treatment effectively reversed these effects of Pim-1 overexpression. In vivo experiments showed that Ventilagolin could effectively suppress HCC tumor growth, downregulate Pim-1, N-cadherin and Vimentin expression, and upregulate E-cadherin expression. CONCLUSION: Ventilagolin suppresses HCC cell proliferation, migration and invasion and reverses EMT process by downregulating Pim-1, suggesting Ventilagolin is a potential therapeutic agent for treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/drug therapy , Naphthoquinones/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Naphthoquinones/chemistry , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Wound Healing/drug effects
20.
ACS Appl Mater Interfaces ; 13(46): 54715-54726, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34757716

ABSTRACT

Nowadays, the development of nanoparticles is known to be mainly associated with enhancement of the targeted delivery of the active component to solid tumors. However, the lack of understanding of the nanoparticle morphology restricts the transport efficiency of various nanocarriers, especially offers no consistent mechanism for the delivery. Here, we demonstrate the principles of enhancement of passive delivery utilizing the precise control and analysis of shape-switchable nanomicelles without any functional addition. We successfully regulated the nanomicelle shape with various aspect ratios in the electrospun nanofiber matrix and devised a stretching phase diagram. Using the vascular leakage model, visual laser spectrum, and image analysis in the simulated scene, we found that the deformed nanomicelles with high aspect ratios along with lower equivalent volumes were significantly beneficial to the passive delivery. Further, the enhanced permeability of the shape-variable nanomicelles in the recovering state was up to 4 times of that observed before recovery. Our results challenge the current consensus of passive targeting and provide an important guidance for the design of nanoparticle morphology and active addition in cancer nanomedicine.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Nanofibers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred Strains , Micelles , Molecular Structure , Neoplasms/pathology , Particle Size
SELECTION OF CITATIONS
SEARCH DETAIL
...