Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Small ; 20(21): e2307273, 2024 May.
Article in English | MEDLINE | ID: mdl-38102096

ABSTRACT

In the course of antitumor therapy, the complex tumor microenvironment and drug-mediated changes in cell signaling and biological processes lead to drug resistance. The effect of sorafenib is greatly limited by the specific tumor microenvironment induced by antiangiogenic therapy and ferroptosis resistance induced by the upregulation of nuclear factor erythroid-2 related factor 2 (NRF2). In this study, a pH responsive and amphiphilic hyperbranched polyglycerol, HDP, is synthesized based on a co-graft click chemistry pathway. This nano-scale carrier provides excellent drug-loading capacity, storing stability and pH responsibility, and effectively co-delivery of sorafenib and siRNA. Sorafenib and siNRF2 plays a greatly synergistic effect in inducing reactive oxygen species (ROS), iron overloading, depleting glutathione (GSH), and promoting lipid peroxidation. Importantly, verified in two different animal experiments, HDP-ss (HDP loaded with both siNRF2 and sorafenib) presents a superior anti-tumor effect, by achieving a tumor inhibition rate of ≈94%. Thus, HDP can serve as an excellent targeted delivery nanocarrier with good biocompatibility in antitumor therapy, and combined application of siNRF2 effectively improves the antitumor effect of sorafenib by overcoming NRF2-mediated ferroptosis resistance. Taken together, this study provides a novel therapeutic strategy to combat the drug resistance in antiangiogenic therapy and ferroptosis.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , NF-E2-Related Factor 2 , Sorafenib , Sorafenib/pharmacology , Sorafenib/chemistry , Ferroptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Humans , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Drug Carriers/chemistry , Reactive Oxygen Species/metabolism , Nanoparticles/chemistry , Cell Line, Tumor , Mice , Glutathione/metabolism
2.
J Nanobiotechnology ; 22(1): 533, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39223666

ABSTRACT

The poor prognosis of hepatocellular carcinoma (HCC) is still an urgent challenge to be solved worldwide. Hence, assembling drugs and targeted short peptides together to construct a novel medicine delivery strategy is crucial for targeted and synergy therapy of HCC. Herein, a high-efficiency nanomedicine delivery strategy has been constructed by combining graphdiyne oxide (GDYO) as a drug-loaded platform, specific peptide (SP94-PEG) as a spear to target HCC cells, sorafenib, doxorubicin-Fe2+ (DOX-Fe2+), and siRNA (SLC7A11-i) as weapons to exert a three-path synergistic attack against HCC cells. In this work, SP94-PEG and GDYO form nanosheets with HCC-targeting properties, the chemotherapeutic drug DOX linked to ferrous ions increases the free iron pool in HCC cells and synergizes with sorafenib to induce cell ferroptosis. As a key gene of ferroptosis, interference with the expression of SLC7A11 makes the ferroptosis effect in HCC cells easier, stronger, and more durable. Through gene interference, drug synergy, and short peptide targeting, the toxic side effects of chemotherapy drugs are reduced. The multifunctional nanomedicine GDYO@SP94/DOX-Fe2+/sorafenib/SLC7A11-i (MNMG) possesses the advantages of strong targeting, good stability, the ability to continuously induce tumor cell ferroptosis and has potential clinical application value, which is different from traditional drugs.


Subject(s)
Carcinoma, Hepatocellular , Doxorubicin , Ferroptosis , Liver Neoplasms , Nanomedicine , Peptides , Sorafenib , Ferroptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Humans , Liver Neoplasms/drug therapy , Doxorubicin/pharmacology , Doxorubicin/chemistry , Nanomedicine/methods , Sorafenib/pharmacology , Sorafenib/chemistry , Cell Line, Tumor , Animals , Peptides/chemistry , Peptides/pharmacology , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Synergism , Amino Acid Transport System y+/metabolism , Mice, Nude , RNA, Small Interfering , Mice, Inbred BALB C , Drug Delivery Systems/methods
3.
J Nanobiotechnology ; 22(1): 204, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658948

ABSTRACT

As a famous drug delivery system (DDS), mesoporous organosilica nanoparticles (MON) are degraded slowly in vivo and the degraded components are not useful for cell nutrition or cancer theranostics, and superparamagnetic iron oxide nanoparticles (SPION) are not mesoporous with low drug loading content (DLC). To overcome the problems of MON and SPION, we developed mesoporous SPIONs (MSPIONs) with an average diameter of 70 nm and pore size of 3.9 nm. Sorafenib (SFN) and/or brequinar (BQR) were loaded into the mesopores of MSPION, generating SFN@MSPION, BQR@MSPION and SFN/BQR@MSPION with high DLC of 11.5% (SFN), 10.1% (BQR) and 10.0% (SNF + BQR), demonstrating that our MSPION is a generic DDS. SFN/BQR@MSPION can be used for high performance ferroptosis therapy of tumors because: (1) the released Fe2+/3+ in tumor microenvironment (TME) can produce •OH via Fenton reaction; (2) the released SFN in TME can inhibit the cystine/glutamate reverse transporter, decrease the intracellular glutathione (GSH) and GSH peroxidase 4 levels, and thus enhance reactive oxygen species and lipid peroxide levels; (3) the released BQR in TME can further enhance the intracellular oxidative stress via dihydroorotate dehydrogenase inhibition. The ferroptosis therapeutic mechanism, efficacy and biosafety of MSPION-based DDS were verified on tumor cells and tumor-bearing mice.


Subject(s)
Drug Delivery Systems , Ferroptosis , Magnetic Iron Oxide Nanoparticles , Sorafenib , Ferroptosis/drug effects , Animals , Magnetic Iron Oxide Nanoparticles/chemistry , Mice , Humans , Drug Delivery Systems/methods , Sorafenib/pharmacology , Sorafenib/chemistry , Sorafenib/therapeutic use , Cell Line, Tumor , Tumor Microenvironment/drug effects , Neoplasms/drug therapy , Porosity , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Mice, Inbred BALB C
4.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273340

ABSTRACT

Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer, with a high mortality rate due to the limited therapeutic options. Systemic drug treatments improve the patient's life expectancy by only a few months. Furthermore, the development of novel small molecule chemotherapeutics is time-consuming and costly. Drug repurposing has been a successful strategy for identifying and utilizing new therapeutic options for diseases with limited treatment options. This study aims to identify candidate drug molecules for HCC treatment through repurposing existing compounds, leveraging the machine learning tool MDeePred. The Open Targets Platform, UniProt, ChEMBL, and Expasy databases were used to create a dataset for drug target interaction (DTI) predictions by MDeePred. Enrichment analyses of DTIs were conducted, leading to the selection of 6 out of 380 DTIs identified by MDeePred for further analyses. The physicochemical properties, lipophilicity, water solubility, drug-likeness, and medicinal chemistry properties of the candidate compounds and approved drugs for advanced stage HCC (lenvatinib, regorafenib, and sorafenib) were analyzed in detail. Drug candidates exhibited drug-like properties and demonstrated significant target docking properties. Our findings indicated the binding efficacy of the selected drug compounds to their designated targets associated with HCC. In conclusion, we identified small molecules that can be further exploited experimentally in HCC therapeutics. Our study also demonstrated the use of the MDeePred deep learning tool in in silico drug repurposing efforts for cancer therapeutics.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Drug Repositioning , Liver Neoplasms , Molecular Docking Simulation , Drug Repositioning/methods , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Sorafenib/pharmacology , Sorafenib/therapeutic use , Sorafenib/chemistry , Machine Learning , Phenylurea Compounds/chemistry , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/pharmacology , Pyridines
5.
Molecules ; 29(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339293

ABSTRACT

Ruthenium(II) polypyridyl complexes are being tested as potential anticancer agents in different therapies, which include conventional chemotherapy and light-activated approaches. A mechanistic study on a recently synthesized dual-action Ru(II) complex [Ru(bpy)2(sora)Cl]+ is described here. It is characterized by two mono-dentate leaving ligands, namely, chloride and sorafenib ligands, which make it possible to form a di-aquo complex able to bind DNA. At the same time, while the released sorafenib can induce ferroptosis, the complex is also able to act as a photosensitizer according to type II photodynamic therapy processes, thus generating one of the most harmful cytotoxic species, 1O2. In order to clarify the mechanism of action of the drug, computational strategies based on density functional theory are exploited. The photophysical properties of the complex, which include the absorption spectrum, the kinetics of ISC, and the character of all the excited states potentially involved in 1O2 generation, as well as the pathway providing the di-aquo complex, are fully explored. Interestingly, the outcomes show that light is needed to form the mono-aquo complex, after releasing both chloride and sorafenib ligands, while the second solvent molecule enters the coordination sphere of the metal once the system has come back to the ground-state potential energy surface. In order to simulate the interaction with canonical DNA, the di-aquo complex interaction with a guanine nucleobase as a model has also been studied. The whole study aims to elucidate the intricate details of the photodissociation process, which could help with designing tailored metal complexes as potential anticancer agents.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Ruthenium , Sorafenib , Sorafenib/chemistry , Sorafenib/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ruthenium/chemistry , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Humans , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology
6.
Molecules ; 29(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38893528

ABSTRACT

The existing kinase inhibitors for hepatocellular carcinoma (HCC) have conferred survival benefits but are hampered by adverse effects and drug resistance, necessitating the development of novel agents targeting distinct pathways. To discover potent new anti-HCC compounds, we leveraged scaffold hopping from Sorafenib and introduced morpholine/piperidine moieties to develop ureido-substituted 4-phenylthiazole analogs with optimized physicochemical properties and binding interactions. Notably, compound 27 exhibited potent cytotoxicity against HepG2 cells (IC50 = 0.62 ± 0.34 µM), significantly exceeding Sorafenib (IC50 = 1.62 ± 0.27 µM). Mechanistic investigations revealed that compound 27 potently inhibited HCC cell migration and colony formation, and it induced G2/M arrest and early-stage apoptosis. Kinase profiling revealed IGF1R as a key target, which compound 27 potently inhibited (76.84% at 10 µM). Molecular modeling substantiated compound 27's strong binding to IGF1R via multiple hydrogen bonds. Computational predictions indicate favorable drug-like properties for compound 27. These findings provide a promising drug candidate for the treatment of HCC patients.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Protein Kinase Inhibitors , Receptor, IGF Type 1 , Thiazoles , Humans , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Cell Proliferation/drug effects , Hep G2 Cells , Thiazoles/chemistry , Thiazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Movement/drug effects , Structure-Activity Relationship , Molecular Docking Simulation , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Molecular Structure , Cell Line, Tumor , Sorafenib/pharmacology , Sorafenib/chemistry , Models, Molecular
7.
Molecules ; 29(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38999138

ABSTRACT

Cancer remains a leading cause of death worldwide, often resulting from uncontrolled growth in various organs. Protein kinase inhibitors represent an important class of targeted cancer therapies. Recently, the kinases BRAF and VEGFR-2 have shown synergistic effects on tumor progression. Seeking to develop dual BRAF/VEGFR-2 inhibitors, we synthesized 18 amino-benzothiazole derivatives with structural similarities to reported dual inhibitors. Four compounds-4a, 4f, 4l, and 4r-demonstrated remarkable cytotoxicity, with IC50 values ranging from 3.58 to 15.36 µM, against three cancer cell lines. Furthermore, these compounds showed IC50 values of 38.77-66.22 µM in the case of a normal cell line, which was significantly safer than the reference, sorafenib. Subsequent investigation revealed that compound 4f exhibited the capacity to inhibit the BRAF and VEGFR-2 enzymes, with IC50 values similar to sorafenib (0.071 and 0.194 µM, respectively). Moreover, compound 4f caused G2-M- and S-phase cycle arrest. Molecular modeling demonstrated binding patterns compatible with inhibition for both targets, where 4f exerted the critical interactions in the BRAF site and interacted in the VEGFR-2 site in a manner akin to sorafenib, demonstrating affinity similar to dabrafenib.


Subject(s)
Antineoplastic Agents , Benzothiazoles , Cell Proliferation , Molecular Docking Simulation , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Thiadiazoles , Vascular Endothelial Growth Factor Receptor-2 , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Humans , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Benzothiazoles/chemical synthesis , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Thiadiazoles/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Design , Structure-Activity Relationship , Sorafenib/pharmacology , Sorafenib/chemistry , Molecular Structure , Computer Simulation , Drug Screening Assays, Antitumor
8.
J Cell Mol Med ; 25(22): 10591-10603, 2021 11.
Article in English | MEDLINE | ID: mdl-34655447

ABSTRACT

Sorafenib is a multikinase inhibitor widely used in cancer therapy with an antitumour effect related to biological processes as proliferation, migration or invasion, among others. Initially designed as a Raf inhibitor, Sorafenib was later shown to also block key molecules in tumour progression such as VEGFR and PDGFR. In addition, sorafenib has been connected with key signalling pathways in cancer such as EGFR/EGF. However, no definitive clue about the molecular mechanism linking sorafenib and EGF signalling pathway has been established so far. Our data in HeLa, U2OS, A549 and HEK293T cells, based on in silico, chemical and genetic approaches demonstrate that the MEK5/ERK5 signalling pathway is a novel target of sorafenib. In addition, our data show how sorafenib is able to block MEK5-dependent phosphorylation of ERK5 in the Ser218/Tyr220, affecting the transcriptional activation associated with ERK5. Moreover, we demonstrate that some of the effects of this kinase inhibitor onto EGF biological responses, such as progression through cell cycle or migration, are mediated through the effect exerted onto ERK5 signalling pathway. Therefore, our observations describe a novel target of sorafenib, the ERK5 signalling pathway, and establish new mechanistic insights for the antitumour effect of this multikinase inhibitor.


Subject(s)
MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 7/metabolism , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology , Biomarkers, Tumor , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement , Disease Susceptibility , Epidermal Growth Factor/metabolism , Flow Cytometry , Humans , Mitogen-Activated Protein Kinase 7/chemistry , Molecular Targeted Therapy , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Sorafenib/chemistry , Structure-Activity Relationship
9.
Phys Chem Chem Phys ; 23(8): 4530-4543, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33595579

ABSTRACT

P-glycoprotein (P-gp), a member of ATP-binding cassette (ABC) transporters, is a multidrug resistance pump. Its promiscuous nature is the main cause of multidrug resistance in cancer cells. P-gp can bind multiple drug molecules simultaneously; however, the binding mechanism is still not clear. Furthermore, the upper limit of the number of substrates that can be accommodated by the binding pocket is not fully understood. In this work, we explore the dynamic process of P-gp binding to multiple substrates by using molecular dynamics (MD) simulations. Our results show that P-gp possesses the ability for simultaneous binding, and that the number of substrates has an upper limit. The accommodating ability of P-gp relates to the size of the binding drugs, and conforms to induced fit theory. In the binding process, the residues 339PHE, 982MET and 986GLN are essential. The pocket of P-gp presents strong flexibility and adaptability features according to the mutation results in this work. Drug molecules tend to gather in the pocket during binding, and interactions between these molecules are beneficial to simultaneous binding. These findings provide new insights into the mechanism of the promiscuous nature of P-gp, and may give us a guideline for inhibiting the process of multidrug resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Paclitaxel/chemistry , Sorafenib/chemistry , Amino Acid Sequence , Binding Sites , Drug Resistance, Multiple , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Static Electricity , Thermodynamics
10.
Bioorg Chem ; 116: 105329, 2021 11.
Article in English | MEDLINE | ID: mdl-34544028

ABSTRACT

There are current attempts to find a safe substitute or adjuvant for Sorafenib (Sorf), the standard treatment for advanced hepatocellular carcinoma (HCC), as it triggers very harsh side effects and drug-resistance. The therapeutic properties of Bee Venom (BV) and its active component, Melittin (Mel), make them suitable candidates as potential anti-cancer agents per-se or as adjuvants for cancer chemotherapy. Hence, this study aimed to evaluate the combining effect of BV and Mel with Sorf on HepG2 cells and to investigate their molecular mechanisms of action. Docking between Mel and different tumor-markers was performed. The cytotoxicity of BV, Mel and Sorf on HepG2 and THLE-2 cells was conducted. Combinations of BV/Sorf and Mel/Sorf were performed in non-constant ratios on HepG2. Expression of major cancer-related genes and oxidative stress status was evaluated and the cell cycle was analyzed. The computational analysis showed that Mel can bind to and inhibit XIAP, Bcl2, MDM2, CDK2 and MMP12. Single treatments of BV, Mel and Sorf on HepG2 showed lower IC50than on THLE-2. All combinations revealed a synergistic effect at a combination index (CI) < 1. Significant upregulation (p < 0.05) of p53, Bax, Cas3, Cas7 and PTEN and significant downregulation (p < 0.05) of Bcl-2, Cyclin-D1, Rac1, Nf-κB, HIF-1a, VEGF and MMP9 were observed. The oxidative stress markers including MDA, SOD, CAT and GPx showed insignificant changes, while the cell cycle was arrested at G2/M phase. In conclusion, BV and Mel have a synergistic anticancer effect with Sorf on HepG2 that may represent a new enhancing strategy for HCC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Bee Venoms/pharmacology , Melitten/pharmacology , Sorafenib/pharmacology , Antineoplastic Agents/chemistry , Bee Venoms/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Lipid Peroxidation/drug effects , Melitten/chemistry , Molecular Docking Simulation , Molecular Structure , Sorafenib/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Bioorg Chem ; 112: 104831, 2021 07.
Article in English | MEDLINE | ID: mdl-33831675

ABSTRACT

A series of 1,2,3-triazole-containing Sorafenib analogues, in which the aryl urea moiety of Sorafenib (1) was replaced with a 1,2,3-triazole ring linking a substituted phenoxy fragment, were prepared successfully via Huisgen 1,3-dipolar cycloaddition and nucleophilic aromatic substitution. The studies of cytotoxicity towards human hepatocellular carcinoma (HCC) cell lines, HepG2 and Huh7, indicated that p-tert-butylphenoxy analogue 2m showed significant inhibitory activity against Huh7 with IC50 = 5.67 ± 0.57 µM. More importantly, 2m showed low cytotoxicity against human embryonal lung fibroblast cell line, MRC-5, with IC50 > 100 µM, suggesting its highly selective cytotoxic activity (SI > 17.6) towards Huh7 which is much superior to that of Sorafenib (SI = 6.73). The molecular docking studies revealed that the analogue 2m bound B-RAF near the binding position of Sorafenib, while it interacted VEGFR2 efficiently at the same binding position of Sorafenib. However, 2m exhibited moderate inhibitory activity toward B-RAF, implying that its anti-Huh7 effect might not strictly relate to inhibition of B-RAF. Wound healing and BrdU cell proliferation assays confirmed anti-cell migration and anti-cell proliferative activities towards Huh7. With its inhibitory efficiency and high safety profile, 2m has been identified as a promising candidate for the treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Sorafenib/pharmacology , Triazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Sorafenib/chemical synthesis , Sorafenib/chemistry , Structure-Activity Relationship , Triazoles/chemistry , Wound Healing/drug effects
12.
J Nanobiotechnology ; 19(1): 360, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34749742

ABSTRACT

In addition to early detection, early diagnosis, and early surgery, it is of great significance to use new strategies for the treatment of hepatocellular carcinoma (HCC). Studies showed that the combination of sorafenib (SFN) and triptolide (TPL) could reduce the clinical dose of SFN and maintain good anti-HCC effect. But the solubility of SFN and TPL in water is low and both drugs have certain toxicity. Therefore, we constructed a biomimetic nanosystem based on cancer cell-platelet (PLT) hybrid membrane camouflage to co-deliver SFN and TPL taking advantage of PLT membrane with long circulation functions and tumor cell membrane with homologous targeting. The biomimetic nanosystem, SFN and TPL loaded cancer cell-PLT hybrid membrane-camouflaged liquid crystalline lipid nanoparticles ((SFN + TPL)@CPLCNPs), could simultaneously load SFN and TPL at the molar ratio of SFN to TPL close to 10:1. (SFN + TPL)@CPLCNPs achieved long circulation function and tumor targeting at the same time, promoting tumor cell apoptosis, inhibiting tumor growth, and achieving a better "synergy and attenuation effect", which provided new ideas for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Diterpenes , Liposomes , Liver Neoplasms/metabolism , Nanoparticles , Phenanthrenes , Sorafenib , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomimetic Materials/chemistry , Blood Platelets/chemistry , Cell Line, Tumor , Cell Membrane/chemistry , Diterpenes/chemistry , Diterpenes/pharmacokinetics , Diterpenes/pharmacology , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacokinetics , Epoxy Compounds/pharmacology , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Liposomes/toxicity , Male , Mice , Mice, Inbred BALB C , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/toxicity , Phenanthrenes/chemistry , Phenanthrenes/pharmacokinetics , Phenanthrenes/pharmacology , RAW 264.7 Cells , Sorafenib/chemistry , Sorafenib/pharmacokinetics , Sorafenib/pharmacology
13.
J Enzyme Inhib Med Chem ; 36(1): 1884-1897, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34340602

ABSTRACT

Sorafenib is recommended as the primary therapeutic drug for patients with hepatocellular carcinoma. To discover a new compound that avoids low response rates and toxic side effects that occur in sorafenib therapy, we designed and synthesized new hybrid compounds of sorafenib and 2,4,5-trimethylpyridin-3-ols. Compound 6 was selected as the best of 24 hybrids that inhibit each of the four Raf kinases. The anti-proliferative activity of 6 in HepG2, Hep3B, and Huh7 cell lines was slightly lower than that of sorafenib. However, in H6c7 and CCD841 normal epithelial cell lines, the cytotoxicity of 6 was much lower than that of sorafenib. In addition, similar to sorafenib, compound 6 inhibited spheroid forming ability of Hep3B cells in vitro and tumour growth in a xenograft tumour model of the chick chorioallantoic membrane implanted with Huh7 cells. Compound 6 may be a promising candidate targeting hepatocellular carcinoma with low toxic side effects on normal cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Pyrimidines/chemistry , Sorafenib/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
14.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206574

ABSTRACT

Sorafenib (Sor) is an oral multi-kinase inhibitor, but its water solubility is very low. To improve its solubility, sorafenib hydrochloride hydrate, sorafenib hydrobromide and sorafenib hydrobromide hydrate were prepared in the mixed solvent of the corresponding acid solution, and tetrahydrofuran (THF). The crystal structures of sorafenib hydrochloride trihydrate (Sor·HCl.3H2O), 4-(4-{3-[4-chloro-3-(trifluoro-methyl)phenyl]ureido}phenoxy)-2-(N-methylcarbamoyl) pyridinium hydrochloride trihydrate, C21H17ClF3N4O3+·Cl-.3H2O (I), sorafenib hydrochloride monohydrate (Sor·HCl.H2O), C21H17ClF3N4O3+·Cl-.H2O (II), its solvated form (sorafenib hydrochloride monohydrate monotetrahydrofuran (Sor·HCl.H2O.THF), C21H17ClF3N4O3+·Cl-.H2O.C4H8O (III)), sorafenib hydrobromide (Sor·HBr), 4-(4-{3-[4-chloro-3-(trifluoro-methyl)phenyl]ureido}phenoxy)-2-(N-methylcarbamoyl) pyridinium hydrobromide, C21H17ClF3N4O3+·Br- (IV) and sorafenib hydrobromide monohydrate (Sor·HBr.H2O), C21H17ClF3N4O3+·Br-.H2O (V) were analysed. Their hydrogen bond systems and topologies were investigated. The results showed the distinct roles of water molecules in stabilizing their crystal structures. Moreover, (II) and (V) were isomorphous crystal structures with the same space group P21/n, and similar unit cell dimensions. The predicted morphologies of these forms based on the BFDH model matched well with experimental morphologies. The energy frameworks showed that (I), and (IV) might have better tabletability than (II) and (V). Moreover, the solubility and dissolution rate data exhibited an improvement in the solubility of these salts compared with the free drug.


Subject(s)
Antineoplastic Agents/chemistry , Hydrogen Bonding , Protein Kinase Inhibitors/chemistry , Sorafenib/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Liquid Crystals/chemistry , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/pharmacology , Solubility , Sorafenib/pharmacology , Spectrum Analysis
15.
Molecules ; 26(11)2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34200376

ABSTRACT

The dissolution rate is the rate-limiting step for Biopharmaceutics Classification System (BCS) class II drugs to enhance their in vivo pharmacokinetic behaviors. There are some factors affecting the dissolution rate, such as polymorphism, particle size, and crystal habit. In this study, to improve the dissolution rate and enhance the in vivo pharmacokinetics of sorafenib tosylate (Sor-Tos), a BCS class II drug, two crystal habits of Sor-Tos were prepared. A plate-shaped crystal habit (ST-A) and a needle-shaped crystal habit (ST-B) were harvested by recrystallization from acetone (ACN) and n-butanol (BuOH), respectively. The surface chemistry of the two crystal habits was determined by powder X-ray diffraction (PXRD) data, molecular modeling, and face indexation analysis, and confirmed by X-ray photoelectron spectroscopy (XPS) data. The results showed that ST-B had a larger hydrophilic surface than ST-A, and subsequently a higher dissolution rate and a substantial enhancement of the in vivo pharmacokinetic performance of ST-B.


Subject(s)
Solubility/drug effects , Sorafenib/chemistry , Acetone/chemistry , Biopharmaceutics/methods , Chemistry, Pharmaceutical/methods , Crystallization/methods , Hydrophobic and Hydrophilic Interactions , Particle Size , Powders/chemistry , X-Ray Diffraction/methods
16.
Pharm Res ; 37(7): 124, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32524365

ABSTRACT

PURPOSE: Lipid-based formulations (LBF) have shown oral bioavailability enhancement of lipophilic drugs, but not necessarily in the case of hydrophobic drugs. This study explored the potential of lipid vehicles to improve the bioavailability of the hydrophobic drug nilotinib comparing a chase dosing approach and lipid suspensions. METHODS: Nilotinib in vivo bioavailability in rats was determined after administering an aqueous suspension chase dosed with blank olive oil, Captex 1000, Peceol or Capmul MCM, respectively. Absolute bioavailability was determined (relative to an intravenous formulation). Pharmacokinetic parameters were compared to lipid suspensions. RESULTS: Compared to the lipid suspensions, the chase dosed lipids showed a 2- to 7-fold higher bioavailability. Both long chain chase dosed excipients also significantly increased the bioavailability up to 2-fold compared to the aqueous suspension. Deconvolution of the pharmacokinetic data indicated that chase dosing of nilotinib resulted in prolonged absorption compared to the aqueous suspension. CONCLUSION: Chase dosed LBF enhanced the in vivo bioavailability of nilotinib. Long chain lipids showed superior performance compared to medium chain lipids. Chase dosing appeared to prolong the absorption phase of the drug. Therefore, chase dosing of LBF is favourable compared to lipid suspensions for 'brick dust' molecules such as nilotinib. Graphical Abstract The potential of bio-enabling lipid vehicles, administered via chase dosing and lipid suspensions, has been evaluated as an approach to enhance oral bioavailability of nilotinib.


Subject(s)
Lipids/chemistry , Liposomes/chemistry , Pyrimidines/chemistry , Animals , Biological Availability , Chemistry, Pharmaceutical , Diglycerides/chemistry , Dose-Response Relationship, Drug , Excipients/chemistry , Hydrophobic and Hydrophilic Interactions , Male , Monoglycerides/chemistry , Oleic Acids/chemistry , Olive Oil/chemistry , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility , Sorafenib/chemistry , Sorafenib/pharmacokinetics , Suspensions/chemistry , Water
17.
J Enzyme Inhib Med Chem ; 35(1): 1712-1726, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32962435

ABSTRACT

A series of imidazothiazole derivatives possessing potential activity against melanoma cells were investigated for molecular mechanism of action. The target compounds were tested against V600E-B-RAF and RAF1 kinases. Compound 1zb is the most potent against both kinases with IC50 values 0.978 and 8.2 nM, respectively. It showed relative selectivity against V600E mutant B-RAF kinase. Compound 1zb was also tested against four melanoma cell lines and exerted superior potency (IC50 0.18-0.59 µM) compared to the reference standard drug, sorafenib (IC50 1.95-5.45 µM). Compound 1zb demonstrated also prominent selectivity towards melanoma cells than normal skin cells. It was further tested in whole-cell kinase assay and showed in-cell V600E-B-RAF kinase inhibition with IC50 of 0.19 µM. Compound 1zb induces apoptosis not necrosis in the most sensitive melanoma cell line, UACC-62. Furthermore, molecular dynamic and 3D-QSAR studies were done to investigate the binding mode and understand the pharmacophoric features of this series of compounds.


Subject(s)
Antineoplastic Agents/chemistry , Melanoma/diet therapy , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Thiazoles/chemistry , Antineoplastic Agents/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Molecular Dynamics Simulation , Oximes/chemistry , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , Sorafenib/chemistry , Sorafenib/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiazoles/pharmacology , Vemurafenib/chemistry , Vemurafenib/pharmacology
18.
Int J Mol Sci ; 21(19)2020 Oct 03.
Article in English | MEDLINE | ID: mdl-33022990

ABSTRACT

Dual drug-loaded nanotherapeutics can play an important role against the drug resistance and side effects of the single drugs. Doxorubicin and sorafenib were efficiently co-encapsulated by tailor-made poly([R,S]-3-hydroxybutyrate) (PHB) using an emulsion-solvent evaporation method. Subsequent poly(ethylene glycol) (PEG) conjugation onto nanoparticles was applied to make the nanocarriers stealth and to improve their drug release characteristics. Monodisperse PHB-sorafenib-doxorubicin nanoparticles had an average size of 199.3 nm, which was increased to 250.5 nm after PEGylation. The nanoparticle yield and encapsulation efficiencies of drugs decreased slightly in consequence of PEG conjugation. The drug release of the doxorubicin was beneficial, since it was liberated faster in a tumor-specific acidic environment than in blood plasma. The PEG attachment decelerated the release of both the doxorubicin and the sorafenib, however, the release of the latter drug remained still significantly faster with increased initial burst compared to doxorubicin. Nevertheless, the PEG-PHB copolymer showed more beneficial drug release kinetics in vitro in comparison with our recently developed PEGylated poly(lactic-co-glycolic acid) nanoparticles loaded with the same drugs.


Subject(s)
Colorectal Neoplasms/drug therapy , Doxorubicin/pharmacology , Nanoparticles/chemistry , Sorafenib/pharmacology , Colorectal Neoplasms/pathology , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Delivery Systems/methods , HCT116 Cells , Humans , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Prohibitins , Sorafenib/chemistry
19.
Molecules ; 25(3)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32013003

ABSTRACT

The crystal structure of the sorafenib and B-RAF complex indicates that the binding cavity occupied by the pyridine-2-carboxamide in sorafenib has a large variable space, making it a reasonable modification site. In order to identify novel compounds with anti-cancer activity, better safety and polar groups for further application, five sorafenib analogs with new pyridine-2-amide side chains were designed and synthesized. Preliminary pharmacologic studies showed that these compounds displayed much lower toxicities than that of sorafenib. Among them, compound 10b bearing mercaptoethyl group kept relevant antiproliferation potency compared to sorafenib in Huh7 and Hela cell lines with values of IC50 58.79 and 63.67 M, respectively. As a small molecule inhibitor targeting protein tyrosine kinases, thiol in compound 10b would be an active group to react with maleimide in a mild condition for forming nanoparticles Sorafenib-PEG-DGL, which could be developed as a delivery vehicle to improve the concentration of anti-tumor therapeutic agents in the target cancer tissue and reduce side effects in the next study.


Subject(s)
Amides/chemistry , Polyethylene Glycols/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , Sorafenib/chemistry , Sorafenib/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Conformation , Molecular Structure , Nanoparticles/chemistry , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/chemistry , Sorafenib/administration & dosage , Structure-Activity Relationship
20.
Pharmazie ; 75(1): 13-17, 2020 01 02.
Article in English | MEDLINE | ID: mdl-32033627

ABSTRACT

Sorafenib (SOR) is an important multikinase inhibitor for the treatment of cancers. It is commercially available (Nexavar from Bayer) in the form of sorafenib tosylate (SORt) due to its very low solubility. Studies have been made to further improve the dissolution behavior of the tosylate form (SORt), which could ultimately moderate the currently high daily dose. In the present study, SORt nanoparticles (SORt-NP) were prepared through a process that combined two industrially well-accepted techniques of co-milling and supercritical extraction. SORt was co-milled with hydrophilic polymers and tetradecanol, and the tetradecanol was post-extracted using supercritical carbon dioxide. The process enabled the formation of SORt-NP without using any toxic organic solvents, and the drug/excipient ratio (1:0.38) was substantially higher than determined in other studies (1:5.4-10). The enhanced dissolution behavior of SORt-NP was possible with an optimized number of milling cycles. Combining co-milling and supercritical extraction was able to form overall porous network structures with reduced crystallite size, which accelerated the dissolution of SORt-NP. The current method could be easily extended to other poorly soluble drugs as a general approach to improve their dissolution behaviors.


Subject(s)
Carbon Dioxide/chemistry , Excipients/chemistry , Fatty Alcohols/chemistry , Sorafenib/chemistry , Chemistry, Pharmaceutical/methods , Drug Compounding/methods , Drug Liberation , Hydrophobic and Hydrophilic Interactions , Nanoparticles , Polymers/chemistry , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Solubility , Sorafenib/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL