Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J AOAC Int ; 107(4): 558-570, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38648750

ABSTRACT

BACKGROUND: Estimation of the drug and development of the method is a critical aspect of formulation development and a critical factor for analytical scientists. Gefitinib is a poorly soluble anticancer drug. OBJECTIVE: The present research focuses on the topic of the development of innovative quality by design methods for the estimation of gefitinib (GF) from bulk, pharmaceutical tablet formulation, and complex nanoformulations. METHODS: To simplify the estimation of poorly soluble drugs such as GF, response surface methodology (RSM) was adopted with effective leverages to obtain precise computation design space using the Box-Behnken design (BBD) model. The major three mixed-effect independent factors (percentage of buffer, pH of buffer, and flow rate) were screened with three prominent dependent responses (viz., theoretical plate, retention time, and tailing factor) selected for optimal analysis. Furthermore, co-processed steps were employed for the estimation of the analyte from the complex formulation. RESULTS: The RP-HPLC method uses the quality by design (QbD) approach can effectively estimate the analyte concentration of less than 4.5 min. The developed method was economically robust and sensitive and shows a relative standard deviation (RSD, %) of less than 2% for all the selected validation parameters. The estimated design space suggests the highest desirability (R2-0.998) at 60% of buffer in the mobile phase, pH 4.25, and flow rate of 0.7 mL/min. CONCLUSIONS: The QbD approach was used to design and develop the method by understanding the interaction between dependent and independent variables to get the optimum values. The developed method was validated successfully and can be useful for formulation scientists to estimate drug concentration and drug release profiles from complex nanoformulations. HIGHLIGHTS: The analytical approach was designed and quantified using a quality-by-design approach to make the RP-HPLC method more robust and efficient for the estimation of analytes from complex nanoformulations. The method is also useful to eliminate the interfering molecule during estimation by employing co-processing steps. The developed method saves time and cost of solvent and employs QbD as a requirement of recent regulatory concern.


Subject(s)
Gefitinib , Tablets , Chromatography, High Pressure Liquid/methods , Gefitinib/analysis , Gefitinib/chemistry , Antineoplastic Agents/analysis , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Drug Compounding/methods , Hydrogen-Ion Concentration , Chromatography, Reverse-Phase/methods
2.
Int J Pharm ; 657: 124109, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38626846

ABSTRACT

Breast cancer continues to pose a substantial global health challenge, emphasizing the critical need for the advancement of novel therapeutic approaches. Key players in the regulation of apoptosis, a fundamental process in cell death, are the B-cell lymphoma 2 (Bcl-2) family proteins, namely Bcl-2 and Bax. These proteins have garnered attention as highly promising targets for the treatment of breast cancer. Targeting the overexpressed anti-apoptotic Bcl-2 protein in breast cancer, Gefitinib (GEF), an EGFR (Epidermal Growth Factor Receptor) inhibitor, emerges as a potential solution. This study focuses on designing Gefitinib-loaded polymeric mixed micelles (GPMM) using poloxamer 407 and TPGS (D-alpha tocopherol PEG1000 succinate) for breast cancer therapy. In silico analyses unveil strong interactions between GEF- Bcl-2 and TPGS-Pgp-2 receptors, indicating efficacy against breast cancer. Molecular dynamics simulations offer insights into GEF and TPGS interactions within the micelles. Formulation optimization via Design of Experiment ensures particle size and entrapment efficiency within acceptable ranges. Characterization tools such as zeta sizer, ATR-FTIR, XRD, TEM, AFM, NMR, TGA, and DSC confirms particle size, structure, functional groups, and thermodynamic events. The optimized micelles exhibit a particle size of 22.34 ± 0.18 nm, PDI of 0.038 ± 0.009, and zeta potential of -0.772 ± 0.12 mV. HPLC determines 95.67 ± 0.34% entrapment efficiency and 1.05 ± 0.12% drug loading capacity. In-vitro studies with MDA-MB-231 cell lines demonstrate enhanced cytotoxicity of GPMM compared to free GEF, suggesting its potential in breast cancer therapy. Cell cycle analysis reveals apoptosis induction through key apoptotic proteins. Western blot results confirm GPMM's ability to trigger apoptosis in MDA-MB-231 cells by activating caspase-3, Bax, Bcl-2, and Parp. In conclusion, these polymeric mixed micelles show promise in selectively targeting cancer cells, warranting future in-vivo studies for optimized clinical application against breast cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Gefitinib , Micelles , Poloxamer , Vitamin E , Humans , Poloxamer/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Vitamin E/chemistry , Female , Gefitinib/administration & dosage , Gefitinib/pharmacology , Gefitinib/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Molecular Dynamics Simulation , Cell Line, Tumor , Drug Carriers/chemistry , Computer Simulation , Particle Size , Cell Survival/drug effects , Animals , Proto-Oncogene Proteins c-bcl-2/metabolism , Polyethylene Glycols/chemistry , Drug Liberation , Apoptosis/drug effects
3.
Acta Pharmacol Sin ; 43(1): 167-176, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33772142

ABSTRACT

Recent studies show that intracellular accumulation of cholesterol leads to acquired resistance to gefitinib in non-small cell lung cancer (NSCLC) cells. In this study we investigated how to regulate the cholesterol levels in gefitinib-resistant NSCLC cells. We showed that intracellular cholesterol levels in gefitinib-resistant cell lines (PC-9/GR, H1975, H1650, and A549) were significantly higher than that in gefitinib-sensitive cell line (PC-9). Treatment with gefitinib (5 µM) significantly increased intracellular cholesterol levels in PC-9/GR, H1975, and H1650 cells. Gefitinib treatment downregulated the expression of PPARα, LXRα, and ABCA1, leading to dysregulation of cholesterol efflux pathway. We found that a lipid-lowering drug fenofibrate (20, 40 µM) dose-dependently increased the expression of PPARα, LXRα, and ABCA1, decreased the intracellular cholesterol levels, and enhanced the antiproliferative effects of gefitinib in PC-9/GR, H1975, and H1650 cells. We revealed that fenofibrate increased the gefitinib-induced apoptosis via regulating the key proteins involved in the intrinsic apoptosis pathway. In PC-9/GR, H1975 and H1650 cells, fenofibrate dose-dependently increased the expression of AMPK, FoxO1, and decreased the expression of AKT, which were remarkably weakened by knockdown of PPARα. In PC-9/GR cell xenograft mice, combined administration of gefitinib (25 mg · kg-1 · d-1) and fenofibrate (100 mg · kg-1 · d-1) caused remarkable inhibition on tumor growth as compared to treatment with either drug alone. All the results suggest that fenofibrate relieves acquired resistance to gefitinib in NSCLC by promoting apoptosis via regulating PPARα/AMPK/AKT/FoxO1 pathway. We propose that combination of gefitinib and fenofibrate is a potential strategy for overcoming the gefitinib resistance in NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Fenofibrate/pharmacology , Gefitinib/pharmacology , Hypolipidemic Agents/pharmacology , Lung Neoplasms/drug therapy , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Fenofibrate/chemistry , Forkhead Box Protein O1/metabolism , Gefitinib/chemistry , Humans , Hypolipidemic Agents/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Structure , PPAR alpha/agonists , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
4.
Mol Med Rep ; 24(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34212976

ABSTRACT

The present study aimed to investigate the effects of a gefitinib derivative, LPY­9, on the proliferation, apoptosis and migration of human glioma cell line U251­MG by CCK8, Transwell or flow cytometry, and the effect of LPY­9 on the activity of caspase­3 enzyme and related proteins in the vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) pathways by western blot and ELISA. It was found that LPY­9 exhibited higher a inhibitory effect on the proliferation of U251­MG cell lines compared with gefitinib and it also exhibited a certain dose­dependence. Following LPY­9 treatment, typical apoptotic morphology was observed under the microscope after Giemsa staining. LPY­9 induced apoptosis at low concentration, and the activity of caspase­3 enzyme increased with the increase in drug concentration, significantly inhibiting the secretion of VEGF in a dose­dependent manner. The effect was notably more evident compared with gefitinib at the same concentration. The expression level of caspase­3 and cleaved caspase­3 increased with the increase in LPY­9 concentration; however, expression levels of VEGF, EGFR, phosphorylated AKT and PI3K decreased with the increase of LPY­9 concentration and no change was observed in the expression level of AKT. LPY­9 inhibited the proliferation of the human glioma cell line U251­MG, promoted apoptosis and effectively inhibited the migration of U251­MG cells. The effect of LPY­9 was more noticeable compared with gefitinib. The results of the present study may provide a foundation for further study and clinical research of this as an anti­tumor drug in animal models.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Gefitinib/chemistry , Gefitinib/pharmacology , Glioma/drug therapy , Animals , Apoptosis/drug effects , Caspase 3 , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioma/genetics , Humans , Phosphorylation , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
J Med Chem ; 64(11): 7839-7852, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34038131

ABSTRACT

Inspired by the success of dual-targeting drugs, especially bispecific antibodies, we propose to combine the concept of proteolysis targeting chimera (PROTAC) and dual targeting to design and synthesize dual PROTAC molecules with the function of degrading two completely different types of targets simultaneously. A library of novel dual-targeting PROTAC molecules has been rationally designed and prepared. A convergent synthetic strategy has been utilized to achieve high synthetic efficiency. These dual PROTAC structures are characterized using trifunctional natural amino acids as star-type core linkers to connect two independent inhibitors and E3 ligands together. In this study, gefitinib, olaparib, and CRBN or VHL E3 ligands were used as substrates to synthesize novel dual PROTACs. They successfully degraded both the epidermal growth factor receptor (EGFR) and poly(ADP-ribose) polymerase (PARP) simultaneously in cancer cells. Being the first successful example of dual PROTACs, this technique will greatly widen the range of application of the PROTAC method and open up a new field for drug discovery.


Subject(s)
Drug Design , ErbB Receptors/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Gefitinib/chemistry , Humans , Ligands , Phthalazines/chemistry , Piperazines/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
6.
Int J Biol Macromol ; 174: 185-197, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33516855

ABSTRACT

PAMAM dendrimers (PAMs) are a group of polymeric macromolecules with distinctive physicochemical features, which can make them multifunctional theranostic nanoparticles (NPs). This study was designed to examine the impact of mucin-1 aptamer-conjugated NPs which were engineered using PAM for image-guided delivery of gefitinib (GEF) in the breast cancer cells/tumor. For this, PAMAM was conjugated with diethylenetriaminepentaacetic acid (DTPA) and modified with PEG2000 to prepare a multi-functionalized NPs. Subsequently, GEF was loaded onto the DTPA-PAM-PEG NPs, which were then armed with MUC-1 aptamer to form the DTPA-PAM-PEG/GEF@MUC-1 nanosystem. Finally, aptamer-conjugated NPs were radiolabeled by gallium-67 as an imaging agent to construct image-guided nanoplatforms. The prepared NPs were characterized by different techniques. The kinetic release models of gefitinib from radiolabeled NPs offer the sustained-release mechanism of the encapsulated drug for over 7 days. In vitro evaluation showed higher cytotoxicity and enhanced uptake of the mucin-grafted NPs in MCF-7 cells. Nuclear medicine imaging and in vivo investigations revealed significant accumulation of 67Ga-DTPA-PAM-PEG/GEF@MUC-1 in the tumor site of the animal models. These data suggest that the engineered NPs are a promising image-guided nanosystem for mucin-expressing breast cells/tumors with the assistance of nuclear medicine.


Subject(s)
Gefitinib/chemistry , Mucin-1/chemistry , Polyamines/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Dendrimers/chemistry , Dendrimers/pharmacology , Drug Delivery Systems/methods , Female , Gefitinib/administration & dosage , Gefitinib/therapeutic use , Humans , MCF-7 Cells , Mucin-1/metabolism , Mucin-1/pharmacology , Nanoparticles/chemistry , Polyethylene Glycols/chemistry
7.
Bioorg Med Chem ; 29: 115889, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33260051

ABSTRACT

Various radiosensitizers are being developed to increase the radiation sensitivity of hypoxic cancer cells, which show resistance to radiation. Previously, we demonstrated that an acetyl glucose-modified nitroimidazole derivative showed a high radiosensitizing effect by inhibiting glucose uptake and glycolysis. Based on this finding, we designed and synthesized novel sugar hybrid radiosensitizers, wherein acetyl glucose was introduced into gefitinib. Among them, UTX-114 had higher autophosphorylation and radiosensitizing activity than gefitinib and inhibited glucose uptake. This result supports our hypothesis that an acetyl glucose moiety improves the radiosensitizing effect of the drug, and UTX-114 can be expected to be a leading compound with a radiosensitizing effect.


Subject(s)
Antineoplastic Agents/chemistry , Gefitinib/chemistry , Glucose/chemistry , Nitroimidazoles/chemistry , Radiation-Sensitizing Agents/chemistry , Antineoplastic Agents/pharmacology , Biomedical Enhancement , Cell Line, Tumor , Cell Membrane Permeability , ErbB Receptors/metabolism , Gefitinib/pharmacology , Glucose Transport Proteins, Facilitative/metabolism , Glycolysis/drug effects , Humans , Monosaccharide Transport Proteins/metabolism , Phosphorylation , Radiation-Sensitizing Agents/pharmacology
8.
Cell Cycle ; 19(24): 3581-3594, 2020 12.
Article in English | MEDLINE | ID: mdl-33300430

ABSTRACT

Gefitinib (GEB) is one of the drugs used for patients with epidermal growth factor receptor (EGFR)-positive mutations in non-small cell lung cancer (NSCLC). However, application of GEB is limited by its low water solubility, stability, and utilization rate, especially the side effects while GEB is given by oral. In this study, nanoliposome was used as a carrier to prepare nanoliposome compound drug (GL) by embedding GEB in the nanoliposome perfectly combined with green nontoxic solvent and thin-film dispersion method. The nanoliposome structure was expected to improve the water solubility and biocompatibility of GEB, thus improving the effect of cancer treatment. The surface electronegative nanoliposomes can effectively avoid protein adsorption and prolong the circulation time in vivo. Meanwhile, the ratio of lecithin to cholesterol (LE/CH) was explored to maximize the encapsulation efficiency of nanoliposome. Subsequent test results showed that GL exhibited better stability, smaller particle size and higher encapsulation efficiency. In addition, in vitro drug release curve also further confirmed that GL had a promising drug sustained-release effect. In particular, a series of in vitro tests such as cell activity, apoptosis, colony formation, scratch, invasion, and cell cycle assays were performed. The results indicated that GL significantly enhanced the pro-apoptotic effect on A549 cells. Most cell cycles of A549 cells were blocked in the G0/G1 phase influenced by GL, thus inhibiting the proliferation of cancer cells. In vivo anti-tumor studies showed that compared with pure GEB, GL had a significant inhibiting effect on NSCLC. In conclusion, the GL which was synthesized by a simple method in this study significantly improved the treatment effect of cancer cells, which proved that the nanoliposome carrier had an excellent application prospect in the treatment of lung cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Delivery Systems/methods , Gefitinib/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Nanostructures/chemistry , A549 Cells , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Capsules , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Liberation , Drug Stability , Gefitinib/chemistry , Humans , Liposomes , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Particle Size , Solubility , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Drug Des Devel Ther ; 14: 4503-4510, 2020.
Article in English | MEDLINE | ID: mdl-33122893

ABSTRACT

INTRODUCTION: This study aims to develop a novel co-delivery gefitinib and quercetin system loaded with PLGA-PEG nanoparticles and evaluate their antitumor activity in vitro and in vivo. METHODS: Gef/Qur NPs were prepared and characterized. The release of drugs, stability, cellular uptake and cytotoxicity were evaluated in vitro. The antitumor effects and systemic toxicity of different formulations were also investigated. RESULTS: Gef/Qur NPs displayed a smaller particle size and a PDI and zeta potential of 0.11 and -23.5 mV, respectively. The hydrophobic Gef and Qur content in NPs reached up to 65.2% and 56.4%, respectively, and their high entrapment efficiencies recorded 83.7% and 82.3%, respectively. The in vitro release of Gef/Qur from the NPs was sustained for 12 h. Compared with control groups, Gef/Qur NPs showed higher cellular uptake and cell inhibition rates. In vivo studies identified the lungs as the target tissue and the region of maximum drug release. Through pharmacodynamics analysis, we found that two drugs (Gef and Qur) were incorporated into one nanoparticle carrier, which played a good role in generating synergistic effect. DISCUSSION: It is concluded that PLGA-PEG is an ideal drug carrier for the co-delivery of Gef/Qur to treat lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Gefitinib/pharmacology , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Quercetin/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Gefitinib/chemistry , Humans , Lung Neoplasms/pathology , Mice , Molecular Structure , Particle Size , Quercetin/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
10.
ACS Appl Mater Interfaces ; 12(41): 45714-45727, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-32927941

ABSTRACT

Despite the effective targeting of the epidermal growth factor receptor (EGFR), the use of gefitinib (GFT) for nonsmall cell lung cancer (NSCLC) treatment meets a failure because of the insufficient drug accumulation in the tumor region. Therefore, developing chemosensitizers of GFT with synergistic therapeutic effects is urgently needed for advanced cancer therapy. Herein, a natural chemosensitizer, natural borneol (NB), is reformulated as an oil-in-water nanoemulsion to enhance its solubility, distribution, and to ultimately increase the therapeutic index with GFT. The nanolization of NB (NBNPs) displays stronger targeted delivery and cytotoxicity than NB by selectively identifying eight specific protein targets in A549 NSCLC cells as revealed by the proteomic studies. Consistently, NBNPs realize stronger chemosensitization effects than NB with GFT by effectively regulating EGFR/EHD1-mediated apoptosis in A549 NSCLC cells. Owing to the satisfying synergistic effect between NBNPs and GFT, the combined therapy not only enhances the anticancer ability of GFT against NSCLC proliferation but also avoids heavy double toxicity in vivo. This finding demonstrates the effective synergism between NBNPs and GFT with clear mechanistic investigation and is expected to extend the application of NBNPs as a novel chemosensitizer for advanced cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Camphanes/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Gefitinib/pharmacology , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Vesicular Transport Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Camphanes/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Emulsions/chemistry , Female , Gefitinib/chemistry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Particle Size , Surface Properties , Vesicular Transport Proteins/metabolism
11.
PLoS One ; 15(7): e0236523, 2020.
Article in English | MEDLINE | ID: mdl-32702075

ABSTRACT

BACKGROUND: Gefitinib is an orally potent and selective ATP-competitive inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase and is commonly used to treat locally advanced or metastatic non-small-cell lung cancer (NSCLC) with sensitive EGFR mutations. Multiple adverse effects associated with gefitinib, including liver and lung injuries, severe nausea, and diarrhea, have limited its clinical application. Xenobiotic-induced bioactivation is thought to be an important reason for gefitinib toxicity, which encouraged us to clarify the metabolism of gefitinib in NSCLC patients. MATERIALS AND METHODS: An ultra-performance liquid chromatography coupled with triple quadrupole time-of-flight mass spectrometry (UPLCQ-TOF-MS) method was established to tentatively identify the metabolites of gefitinib in human plasma. The extracted ion chromatogram peak intensity threshold was set at 1500 cps with minimum MS and MS/MS peak intensities of 400 and 100 cps, respectively. RESULTS: A total of 18 tentative metabolites were identified. Eight novel tentative metabolites with metabolic changes in dechlorination, defluorination, and hydrogenation on the quinazoline skeleton; removal of a partial or complete 3-chloro-4-fluoroaniline-substituted group; and sulfate conjugation and taurine conjugation were newly discovered in human plasma. Based on structural analysis of the tentative metabolites, the metabolic pathways were proposed. In addition, the pathways of dechlorination, defluorination, and hydrogenation on the quinazoline skeleton; removal of partial or complete 3-chloro-4-fluoroaniline-substituted groups; and sulfate conjugation and taurine conjugation in humans in vivo indicate that novel metabolic pathways exist in humans. CONCLUSIONS: In summary, the metabolism of gefitinib in humans in vivo is extensive and complex. Based on in vivo evidence, the propoxy-morpholine ring side chain and O-methyl group are the critical metabolic regions of gefitinib in humans. The novel metabolic pathways differ from those of in vitro studies, suggesting that intestinal floral metabolism might be involved.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood , Gefitinib/chemistry , Lung Neoplasms/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Chromatography, High Pressure Liquid , Gefitinib/blood , Gefitinib/metabolism , Gefitinib/therapeutic use , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Tandem Mass Spectrometry
12.
Clin Cancer Res ; 26(20): 5424-5437, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32723835

ABSTRACT

PURPOSE: Small-molecule inhibitors have had a major impact on cancer care. While treatments have demonstrated clinically promising results, they suffer from dose-limiting toxicities and the emergence of refractory disease. Considerable efforts made to address these issues have more recently focused on strategies implementing particle-based probes that improve drug delivery and accumulation at target sites, while reducing off-target effects. EXPERIMENTAL DESIGN: Ultrasmall (<8 nm) core-shell silica nanoparticles, C' dots, were molecularly engineered to function as multivalent drug delivery vehicles for significantly improving key in vivo biological and therapeutic properties of a prototype epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, gefitinib. Novel surface chemical components were used to conjugate gefitinib-dipeptide drug-linkers and deferoxamine (DFO) chelators for therapeutic delivery and PET imaging labels, respectively. RESULTS: Gefitinib-bound C' dots (DFO-Gef-C' dots), synthesized using the gefitinib analogue, APdMG, at a range of drug-to-particle ratios (DPR; DPR = 11-56), demonstrated high stability for DPR values≤ 40, bulk renal clearance, and enhanced in vitro cytotoxicity relative to gefitinib (LD50 = 6.21 nmol/L vs. 3 µmol/L, respectively). In human non-small cell lung cancer mice, efficacious Gef-C' dot doses were at least 200-fold lower than that needed for gefitinib (360 nmoles vs. 78 µmoles, respectively), noting fairly equivalent tumor growth inhibition and prolonged survival. Gef-C' dot-treated tumors also exhibited low phosphorylated EFGR levels, with no appreciable wild-type EGFR target inhibition, unlike free drug. CONCLUSIONS: Results underscore the clinical potential of DFO-Gef-C' dots to effectively manage disease and minimize off-target effects at a fraction of the native drug dose.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Gefitinib/pharmacology , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Small Molecule Libraries/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deferoxamine/chemistry , Deferoxamine/pharmacology , Drug Delivery Systems , Gefitinib/chemistry , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Mice , Positron-Emission Tomography , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Silicon Dioxide/chemistry , Small Molecule Libraries/chemistry
13.
Drug Dev Ind Pharm ; 46(8): 1265-1277, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32594775

ABSTRACT

Gefitinib as an epidermal growth factor receptor tyrosine kinase inhibitor has strong potential in lung cancer therapy. However, a major challenge of using gefitinib is its toxicities. In the present study, we developed a dry powder inhaler dosage form containing gefitinib loaded glucosamine targeted solid lipid nanopaticles (Gef-G-SLNs) to locally transfer anticancer agent to the lung tumor. The Gef-G-SLNs were prepared by emulsion-solvent diffusion and evaporation method and optimized with irregular factorial design. The optimized nanoformulation was tested for action against A549 cells. Mannitol or lactose based dry powders were obtained from Gef-G-SLNs after spray drying and characterized using Anderson Cascade Impactor. The optimized formulation had drug loading of 33.29%, encapsulation efficiency of 97.31 ± 0.23%, zeta potential of -15.53 ± 0.47 mV, particle size of 187.23 ± 14.08 nm, polydispersity index of 0.28 ± 0.02 and release efficiency of 35.46 ± 2.25%. The Gef-G-SLNs showed superior anticancer effect compared to free gefitinib. The increased cellular uptake of G-SLNs in A549 cells was demonstrated compared with non-targeted SLNs using flow cytometry and fluorescence microscopy. The produced mannitol based microparticles showed suitable aerodynamic properties with an acceptable mass median aerodynamic diameter of 4.48 µm and fine particle fraction of 44.41%. Therefore, it can be concluded that this formulation represents promising drug delivery to treatment of lung cancer.


Subject(s)
Gefitinib/therapeutic use , Glucosamine/administration & dosage , Lung Neoplasms , Nanoparticles , Administration, Inhalation , Dry Powder Inhalers , Gefitinib/chemistry , Glucosamine/chemistry , Humans , Lung Neoplasms/drug therapy , Particle Size , Powders
14.
ChemMedChem ; 15(9): 794-798, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32162481

ABSTRACT

Attractive results have been achieved with small-molecule target-based drugs in the anticancer field; however, enhancing their treatment effect and solving the problem of drug resistance remain key concerns worldwide. Inspired by the specific affinity of gefitinib for tumour cells and the strong oxidation capacity of singlet oxygen, we combined a chemically generated singlet oxygen moiety with the small-molecule targeted drug gefitinib to improve its anticancer effect. We designed and synthesised a novel compound (Y5-1), in which a small-molecule targeted therapy agent (gefitinib) and a singlet oxygen (provided by an in vitro photodynamic reaction) thermally controlled releasing moiety are covalently conjugated. We demonstrated that the introduction of the singlet oxygen thermally controlled releasing moiety enhanced the anticancer activities of gefitinib. The results of this study are expected to provide a novel strategy to enhance the effect of chemotherapy drugs on drug-resistant cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Gefitinib/pharmacology , Singlet Oxygen/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Gefitinib/chemical synthesis , Gefitinib/chemistry , Humans , Molecular Structure
15.
J Mater Chem B ; 8(11): 2307-2320, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32100786

ABSTRACT

In this study, a new type of ß-1,3-d-glucan porous microcapsule (GPM)-enveloped and folate conjugated chitosan-functional liposome (FCL), FCL@GPM, was developed for the potential oral co-delivery of chemotherapeutic drugs and quantum dots (QDs) with facilitated drug absorption and antitumor efficacy. In this dual-particulate system, multiple FCLs serve as the cores for effective loading, folate-mediated tumor-targeting, facilitated intracellular accumulation, and pH-responsive controlled release of chemotherapeutic agents, while a GPM acts as the shell for affording macrophage-mediated tumor selectivity. Gefitinib (GEF) was selected as a chemotherapeutic agent, while acid degradable ZnO QDs were selected due to their dual role as an anticancer agent for synergistic chemotherapy and as a fluorescent probe for potential cancer cellular imaging. The GEF and ZnO QD co-loaded FCL@GPMs (GEF/ZnO-FCL@GPMs) exhibited a prolonged release manner with limited release before uptake by intestinal cells. Furthermore, Peyer's patch uptake, macrophage uptake, cytotoxicity, and biodistribution of FCL@GPMs were tested. In addition, GEF and ZnO QD co-loaded FCLs (GEF/ZnO-FCLs) not only have a tumor acidity responsive release property, but also induce a superior cytotoxicity on cancer cells as compared to GEF. Moreover, a 1.75-fold increase in the bioavailability of GEF delivered from GEF/ZnO-FCL@GPMs as compared to its trademarked drug (Iressa®). As a result, GEF/ZnO-FCL@GPMs exerted a superior antitumor efficacy (1.47-fold) as compared to the trademarked drug in mice. Considered together, the developed FCL@GPMs, combining the unique physicochemical and biological benefits of FCLs and GPMs, possess great potential as an efficient delivery system for the co-delivery of chemotherapeutic agents and quantum dots.


Subject(s)
Antineoplastic Agents/chemistry , Chitosan/chemistry , Folic Acid/chemistry , Gefitinib/chemistry , Liposomes/chemistry , Mouth Neoplasms/diagnostic imaging , Mouth Neoplasms/drug therapy , Proteoglycans/chemistry , Quantum Dots/chemistry , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Biological Transport , Cell Survival/drug effects , Drug Compounding/methods , Drug Liberation , Drug Synergism , Fluorescent Dyes/chemistry , Gefitinib/administration & dosage , Gefitinib/pharmacokinetics , Humans , Male , Mice , Mice, Nude , Porosity , Rats, Sprague-Dawley , Tissue Distribution , Zinc Oxide/chemistry
16.
Drug Deliv Transl Res ; 10(4): 862-877, 2020 08.
Article in English | MEDLINE | ID: mdl-32103449

ABSTRACT

The major objective of the present investigation was to assess the targeting potential of a designed system for breast cancer at metastatic phases with imaging ability. In a nutshell, we have developed surface-engineered graphene oxide (GO) nanosheets by covalent linking with amine-functionalized iron oxide nanoparticles (IONPs) (GOIOIs). Gefitinib (Gf) was selected as a model drug and entrapped in between exfoliated GO sheets (GOIGF) via π-π* stacking before functionalization with IONPs. Preliminary characterization of GO, IONPs, GOIOI, and GOIGF was performed using UV-visible and Fourier transform infrared spectroscopy. Scanning and transmission electron microscopy studies confirmed successful surface engineering of GO with IONPs. The in vitro drug release study demonstrated sustained release of Gf. The magnetic behavior of IONPs and GOIOI demonstrated a sigmoidal-shaped hysteresis loop with superparamagnetic properties. The in vitro cell cytotoxicity assay was carried out on MDA-MB-231 breast cancer adenocarcinoma cell lines. The cell cytotoxicity assay showed 61.18% inhibition of cell growth with 30 ppm concentration containing 64% of the drug, whereas 100% of the pure drug revealed only 56% of inhibition. In the near future, GOIOI could be tailored further for theranostic research, especially for metastatic cancers. Graphical abstract.


Subject(s)
Amines , Antineoplastic Agents , Ferric Compounds , Gefitinib , Graphite , Methylamines , Nanoparticles , Amines/administration & dosage , Amines/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Liberation , Erythrocytes/drug effects , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Gefitinib/administration & dosage , Gefitinib/chemistry , Graphite/administration & dosage , Graphite/chemistry , Hemolysis/drug effects , Humans , Magnetic Phenomena , Methylamines/administration & dosage , Methylamines/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/pathology
17.
Anticancer Res ; 39(12): 6585-6593, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31810924

ABSTRACT

BACKGROUND/AIM: Human lung adenocarcinoma PC14 cells without mutations in the epidermal growth factor receptor (EGFR) are less sensitive to gefitinib than PC9 cells with EGFR mutations. We report the involvement of tetrandrine in autophagy flux as a mechanism that enhances the sensitivity of PC14 cells to gefitinib. MATERIALS AND METHODS: Sensitivity to gefitinib was determined by a growth inhibition assay, and quantitative real-time PCR, western blotting, and fluorescent immunostaining were used to detect autophagy. RESULTS: In PC14 cells, combined treatment with gefitinib and tetrandrine caused a significant increase in gefitinib sensitivity and autophagy-related mRNAs and proteins (LC3, etc.), and the LC3 protein accumulated in lysosomes. Furthermore, an autophagy flux assay revealed that tetrandrine inhibited lysosomes and that gefitinib promoted autophagy. Finally, the sensitivity of PC14 cells to gefitinib was enhanced with chloroquine. CONCLUSION: Tetrandrine possibly increases the susceptibility of PC14 cells to gefitinib by lysosomal inhibition.


Subject(s)
Adenocarcinoma of Lung/metabolism , Benzylisoquinolines/pharmacology , Gefitinib/pharmacology , Lung Neoplasms/metabolism , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Autophagy , Benzylisoquinolines/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/genetics , Gefitinib/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lysosomes/drug effects , Microtubule-Associated Proteins/genetics
18.
Nanoscale ; 11(34): 15958-15970, 2019 Aug 29.
Article in English | MEDLINE | ID: mdl-31418432

ABSTRACT

Oral drug delivery systems (ODDSs) have attracted considerable attention in relation to orthotopic colon cancer therapy due to certain popular advantages. Unfortunately, their clinical applications are generally limited by the side-effects caused by systemic drug exposure and poor real-time monitoring capabilities. Inspired by the characteristics of pH changes of the gastrointestinal tract (GIT) and specific enzymes secreted by the colonic microflora, we anchored polyacrylic acid (PAA) and chitosan (CS) on Gd3+-doped mesoporous hydroxyapatite nanoparticles (Gd-MHAp NPs) to realize programmed drug release and magnetic resonance imaging (MRI) at the tumor sites. In particular, the grafted PAA, as a pH-responsive switch, could effect controlled drug release in the colon. Further, CS is functionalized as the enzyme-sensitive moiety, which could be degraded by ß-glycosidase in the colon. Gadolinium is a paramagnetic lanthanide element used in chelates, working as a contrast medium agent for an MRI system. Interestingly, after oral administration, CS and PAA could protect the drug-loaded nanoparticles (NPs) against variable physiological conditions in the GIT, allowing the drug to reach the colon tumor sites, preventing premature drug release. Enhanced drug concentrations at the colon tumor sites were achieved via this programmed drug release, which subsequently ameliorated the therapeutic effect. In addition, encapsulating both chemotherapeutic (5-fluorouracil, 5-FU) and targeted therapy drug (gefitinib, Gef) within Gd-MHAp NPs produced a synergistic therapeutic effect. In summary, this study demonstrated that such a novel drug system (Gd-MHAp/5-FU/Gef/CS/PAA NPs) could protect, transport, and program drug release locally within the colonic environment; further, this system exhibited a worthwhile therapeutic effect, providing a promising novel treatment strategy for orthotopic colon cancer.


Subject(s)
Colonic Neoplasms , Contrast Media , Fluorouracil , Gadolinium , Gefitinib , Magnetic Resonance Imaging , Nanoparticles , Acrylic Resins/chemistry , Acrylic Resins/pharmacokinetics , Acrylic Resins/pharmacology , Administration, Oral , Animals , Chitosan/chemistry , Chitosan/pharmacokinetics , Chitosan/pharmacology , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Durapatite/chemistry , Durapatite/pharmacokinetics , Durapatite/pharmacology , Fluorouracil/chemistry , Fluorouracil/pharmacokinetics , Fluorouracil/pharmacology , Gadolinium/chemistry , Gadolinium/pharmacokinetics , Gadolinium/pharmacology , Gefitinib/chemistry , Gefitinib/pharmacokinetics , Gefitinib/pharmacology , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use
19.
Arch Pharm (Weinheim) ; 352(5): e1800381, 2019 May.
Article in English | MEDLINE | ID: mdl-31012144

ABSTRACT

Drug latentiation is a process of modifying a drug molecule structurally to improve its binding affinity as well as increasing the drug-receptor interactions and potentiate its therapeutic potential. In the quest for discovering more potent epidermal growth factor receptor (EGFR) inhibitors, gefitinib-based derivatives were designed by simple structural modification at the secondary amine of gefitinib by N-alkylation. Three gefitinib derivatives (gefitinib-NB, -NP, and -NIP) were synthesized by N-alkylation and phase transfer catalysis. Structural characterization, physicochemical parameters such as solubility, log P, and p K a were determined. Molecular docking studies were carried out to investigate the binding interactions at the active site. Further drug-bovine serum albumin (BSA) protein and drug-calf thymus (CT) DNA interactions were performed to understand the pharmacokinetics of the synthesized derivatives. All the compounds were screened for preliminary in vitro cytotoxic activity against A549, A431 lung, and MDA-MB-231 breast cancer cell lines by MTT assay. The gefitinib-NP and gefitinib-NB derivatives exhibited strong cytotoxic activity compared with gefitinib. They also showed higher drug-BSA and drug-DNA interactions. Molecular docking studies showed the orientation and binding interactions with the EGFR as well as with BSA and CT DNA. The results establish a strong correlation between the experimental and molecular docking studies. EGFR inhibition studies were also carried out for the derivatives and we identified the NP derivative of gefitinib as a potential lead compound. The gefitinib-based derivatives reported herein are cytotoxic agents and can be tested for further pharmacokinetic profiles and toxicity studies which might be helpful for designing more potent gefitinib-based derivatives in the future.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Gefitinib/analogs & derivatives , Gefitinib/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Gefitinib/chemical synthesis , Gefitinib/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
20.
J Colloid Interface Sci ; 548: 131-144, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-30991180

ABSTRACT

The construction of multifunctional theranostic nanoplatforms to integrate accurate imaging and enhanced therapy to treat tumors is highly attractive but remains a challenge. Here, we developed a molybdenum disulfide (MoS2)-based hyaluronic acid (HA)-functionalized nanoplatform capable of achieving the targeted co-delivery of the gadolinium (Gd)-based contrast agents (CAs) and the anticancer drug gefitinib (Gef) for magnetic resonance imaging (MRI) and synergetic chemo-photothermal therapy of tumors. Gd3+ ions were coupled to HA-grafted MoS2 nanosheets with diethylenetriaminepentaacetic acid (DTPA) as a linker, followed by the incorporation of Gef. The resulting MoS2-HA-DTPA-Gd/Gef exhibited enhanced relaxivity, 3.3 times greater than that of the commercial CA DTPA-Gd, which facilitated the MRI in vivo. Moreover, the nanoplatform effectively converted the absorbed near-infrared (NIR) light into heat, which not only induced the photothermal ablation of cancer cells but also triggered the release of Gef from MoS2-HA-DTPA-Gd/Gef, enabling the synergetic chemo-photothermal therapy. The results of in vitro and in vivo experiments revealed that MoS2-HA-DTPA-Gd/Gef upon NIR irradiation effectively blocked the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) signaling pathway and activated apoptosis-related proteins to induce cell apoptosis and suppress cell proliferation, thus inhibiting the tumor growth in lung cancer cell-bearing mice. Taken together, this multifunctional theranostic nanoplatform has significant promise for the diagnosis and treatment of cancer.


Subject(s)
Disulfides/chemistry , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Molybdenum/chemistry , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biological Transport , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Contrast Media/chemistry , Drug Liberation , Drug Therapy/methods , Gadolinium/chemistry , Gefitinib/chemistry , Gefitinib/pharmacology , Humans , Infrared Rays , Magnetic Resonance Imaging/methods , Mice , Particle Size , Phototherapy/methods , Signal Transduction , Surface Properties , Theranostic Nanomedicine
SELECTION OF CITATIONS
SEARCH DETAIL
...