Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Korean Med Sci ; 36(40): e259, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34664799

ABSTRACT

BACKGROUND: Titanium dioxide films exhibit good biocompatibility and may be effective as drug-binding matrices for drug-eluting stents. We conducted a mid-term evaluation of a novel polymer-free everolimus-eluting stent using nitrogen-doped titanium dioxide film deposition (TIGEREVOLUTION®) in comparison with a commercial durable polymer everolimus-eluting stent (XIENCE Alpine®) in a porcine coronary restenosis model. METHODS: Twenty-eight coronary arteries from 14 mini-pigs were randomly allocated to TIGEREVOLUTION® stent and XIENCE Alpine® stent groups. The stents were implanted in the coronary artery at a 1.1-1.2:1 stent-to-artery ratio. Eleven stented coronary arteries in each group were finally analyzed using coronary angiography, optical coherence tomography, and histopathologic evaluation 6 months after stenting. RESULTS: Quantitative coronary analysis showed no significant differences in the pre-procedural, post-procedural, and 6-month lumen diameters between the groups. In the volumetric analysis of optical coherence tomography at 6 months, no significant differences were observed in stent volume, lumen volume, and percent area stenosis between the groups. There were no significant differences in injury score, inflammation score, or fibrin score between the groups, although the fibrin score was zero in the TIGEREVOLUTION® stent group (0 vs. 0.07 ± 0.11, P = 0.180). CONCLUSION: Preclinical evaluation, including optical coherence tomographic findings 6 months after stenting, demonstrated that the TIGEREVOLUTION® stent exhibited efficacy and safety comparable with the XIENCE Alpine® stent, supporting the need for further clinical studies on the TIGEREVOLUTION® stent.


Subject(s)
Coronary Restenosis/drug therapy , Drug-Eluting Stents , Everolimus/therapeutic use , Animals , Coronary Angiography , Coronary Restenosis/pathology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Everolimus/chemistry , Polymers/chemistry , Swine , Swine, Miniature , Titanium/chemistry , Tomography, Optical Coherence
2.
Int J Mol Sci ; 22(14)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34299359

ABSTRACT

Chronic lung allograft dysfunction (CLAD) and interstitial lung disease associated with collagen tissue diseases (CTD-ILD) are two end-stage lung disorders in which different chronic triggers induce activation of myo-/fibroblasts (LFs). Everolimus, an mTOR inhibitor, can be adopted as a potential strategy for CLAD and CTD-ILD, however it exerts important side effects. This study aims to exploit nanomedicine to reduce everolimus side effects encapsulating it inside liposomes targeted against LFs, expressing a high rate of CD44. PEGylated liposomes were modified with high molecular weight hyaluronic acid and loaded with everolimus (PEG-LIP(ev)-HA400kDa). Liposomes were tested by in vitro experiments using LFs derived from broncholveolar lavage (BAL) of patients affected by CLAD and CTD-ILD, and on alveolar macrophages (AM) and lymphocytes isolated, respectively, from BAL and peripheral blood. PEG-LIP-HA400kDa demonstrated to be specific for LFs, but not for CD44-negative cells, and after loading everolimus, PEG-LIP(ev)-HA400kDa were able to arrest cell cycle arrest and to decrease phospho-mTOR level. PEG-LIP(ev)-HA400kDa showed anti-inflammatory effect on immune cells. This study opens the possibility to use everolimus in lung fibrotic diseases, demonstrating that our lipids-based vehicles can vehicle everolimus inside cells exerting the same drug molecular effect, not only in LFs, but also in immune cells.


Subject(s)
Everolimus/pharmacology , Hyaluronic Acid/pharmacology , Liposomes/chemistry , Pulmonary Fibrosis/drug therapy , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage/methods , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Drug Delivery Systems/methods , Everolimus/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Lung/drug effects , Lung/metabolism , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Polyethylene Glycols/chemistry , Pulmonary Fibrosis/metabolism
3.
Biomed Mater ; 16(4)2021 06 16.
Article in English | MEDLINE | ID: mdl-34020430

ABSTRACT

Drug-eluting bioresorbable vascular scaffolds (BVSs) have emerged as a potential breakthrough for the treatment of coronary artery stenosis, providing mechanical support and drug delivery followed by complete resorption. Restenosis and thrombosis remain the primary limitations in clinical use. The study aimed to identify potential markers of restenosis and thrombosis analyzing the vascular wall cell transcriptomic profile modulation triggered by BVS at different values of shear stress (SS). Human coronary artery endothelial cells and smooth muscle cells were cultured under SS (1 and 20 dyne cm-2) for 6 h without and with application of BVS and everolimus 600 nM. Cell RNA-Seq and bioinformatics analysis identified modulated genes by direct comparison of SS conditions and Gene Ontology (GO). The results of different experimental conditions and GO analysis highlighted the modulation of specific genes as semaphorin 3E, mesenchyme homeobox 2, bone morphogenetic protein 4, (heme oxygenase 1) and selectin E, with different roles in pathological evolution of disease. Transcriptomic analysis of dynamic vascular cell cultures identifies candidate genes related to pro-restenotic and pro-thrombotic mechanisms in anin-vitrosetting of BVS, which are not adequately contrasted by everolimus addition.


Subject(s)
Absorbable Implants , Biocompatible Materials , Biomarkers/metabolism , Thrombosis/metabolism , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cells, Cultured , Coronary Restenosis/metabolism , Coronary Vessels/cytology , Everolimus/chemistry , Everolimus/pharmacology , Humans , Shear Strength , Transcriptome/drug effects
4.
Molecules ; 25(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887218

ABSTRACT

Natural products remain a popular alternative treatment for many ailments in various countries. This study aimed to screen for potential mammalian target of rapamycin (mTOR) inhibitors from Malaysian natural substance, using the Natural Product Discovery database, and to determine the IC50 of the selected mTOR inhibitors against UMB1949 cell line. The crystallographic structure of the molecular target (mTOR) was obtained from Protein Data Bank, with Protein Data Bank (PDB) ID: 4DRI. Everolimus, an mTOR inhibitor, was used as a standard compound for the comparative analysis. Computational docking approach was performed, using AutoDock Vina (screening) and AutoDock 4.2.6 (analysis). Based on our analysis, asiaticoside and its derivative, asiatic acid, both from Centella asiatica, revealed optimum-binding affinities with mTOR that were comparable to our standard compound. The effect of asiaticoside and asiatic acid on mTOR inhibition was validated with UMB1949 cell line, and their IC50 values were 300 and 60 µM, respectively, compared to everolimus (29.5 µM). Interestingly, this is the first study of asiaticoside and asiatic acid against tuberous sclerosis complex (TSC) disease model by targeting mTOR. These results, coupled with our in silico findings, should prompt further studies, to clarify the mode of action, safety, and efficacy of these compounds as mTOR inhibitors.


Subject(s)
Computer Simulation , Pentacyclic Triterpenes/pharmacology , Plants/chemistry , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triterpenes/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Everolimus/chemistry , Everolimus/pharmacology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Malaysia , Molecular Docking Simulation , Pentacyclic Triterpenes/chemistry , Protein Kinase Inhibitors/chemistry , TOR Serine-Threonine Kinases/metabolism , Triterpenes/chemistry
5.
Expert Rev Med Devices ; 17(7): 671-682, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32543934

ABSTRACT

INTRODUCTION: Coronary percutaneous interventions have evolved from plain old balloon angioplasty (POBA) to stent implantation, which itself evolved from bare-metal stents (BMS) to the new biodegradable stents which try to restore endothelial function. Currently, the most commonly used stent is the everolimus-eluting stent. AREAS COVERED: This review will cover the current status of durable polymer everolimus-eluting stent, its history, and future perspectives. Nowadays, the everolimus-eluting stent is the most used device in the acute and chronic settings due to its safety and efficacy. EXPERT OPINION: Durable polymer everolimus-eluting stent, supported by much evidence, has demonstrated its efficacy and safety, not only in de novo artery lesions, but in multiples scenarios, such as the acute setting and diabetic population, becoming one of the most polyvalent stents available. Nowadays, research is focused on the reduction of antiplatelet treatment duration. Similar rates of stent thrombosis with short dual antiplatelet treatment regimens of 1 to 3 months compared to pronged treatment have been observed. However, specific studies should be performed to evaluate this possibility.


Subject(s)
Drug-Eluting Stents/trends , Everolimus/therapeutic use , Polymers/chemistry , Acute Coronary Syndrome/drug therapy , Drug-Eluting Stents/adverse effects , Everolimus/adverse effects , Everolimus/chemistry , Humans , Treatment Outcome
6.
Pharm Res ; 37(3): 39, 2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31965330

ABSTRACT

PURPOSE: The intratumoral heterogeneity observed in breast cancer (BC), in particular with regard to cell surface receptor expression, can hinder the success of many targeted cancer therapies. The development of novel therapeutic agents that target more than one receptor can overcome this inherent property of tumors and can facilitate their selective internalization in cancer cells. The goal of this study is to develop a drug combination-loaded nanoparticle (NP) formulation that is actively-targeted to HER2 and EGFR receptors on BC cells. METHODS: A polymeric NP formulation was prepared which co-encapsulated a synergistic combination of the chemotherapeutic agent, paclitaxel (PTX), and the mTOR inhibitor, everolimus (EVER), and is targeted to HER2 and EGFR receptors on BC cells using antibody Fab fragments as the targeting moieties. The physicochemical characteristics of the dual-targeted formulation (Dual-NP) were evaluated, along with its cytotoxic profile (in both, monolayer and 3D BC models), as well as the degree of cellular uptake in HER2high/EGFRmod and HER2neg/EGFRlow BC cells. RESULTS: Dual-NPs were found to have significantly higher cytotoxicity relative to HER2 mono-targeted (T-NPs) and untargeted NPs (UT-NPs) in HER2high/EGFRmod monolayer BC cells after 72 h exposure, while no significant difference was observed in HER2neg/EGFRlow cells. However, in the HER2high/EGFRmod spheroids, the cytotoxicity of Dual-NPs was comparable to that of T-NPs. This was thought to be attributed to the previously reported downregulation of EGFR in 3D in comparison to 2D BC models. Dual-NPs had significantly higher cellular uptake relative to UT-NPs and T-NPs in HER2high/EGFRmod BC cells after 24 h exposure, whereas in the HER2neg/EGFRlow cells, the increase in cellular uptake of the Dual-NPs was not as high as the level achieved in the HER2high/EGFRmod cells. Blocking HER2 and EGFR significantly reduced the uptake of T-NPs and Dual-NPs in the HER2high/EGFRmod BC cells, demonstrating specific binding to both EGFR and HER2. CONCLUSIONS: The dual-targeting strategy developed in this study in conjunction with a potentially promising delivery vector for a synergistic combination therapy can overcome receptor heterogeneity, yielding significant improvements in the cytotoxicity and cellular uptake in BC cells.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Everolimus/chemistry , Nanocapsules/chemistry , Paclitaxel/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Survival , Drug Compounding/methods , Drug Liberation , ErbB Receptors/metabolism , Everolimus/pharmacology , Female , Humans , Molecular Targeted Therapy/methods , Paclitaxel/pharmacology , Panitumumab/chemistry , Panitumumab/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Receptor, ErbB-2/metabolism , Surface Properties , Trastuzumab/chemistry , Trastuzumab/metabolism
7.
Biomacromolecules ; 20(12): 4546-4562, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31697482

ABSTRACT

Macromolecular architecture plays an important role in the self-assembly process of block copolymer amphiphiles. Herein, two series of stimuli-responsive amphiphilic 3-miktoarm star hybrid terpolypeptides and their corresponding linear analogues were synthesized exhibiting the same overall composition and molecular weight but different macromolecular architecture. The macromolecular architecture was found to be a key parameter in defining the morphology of the nanostructures formed in aqueous solutions as well as to alter the self-assembly behavior of the polymers independently of their composition. In addition, it was found that the assemblies prepared from the star-shaped polymers showed superior tolerance against enzymatic degradation due to the increased corona block density on the outer surface of the nanoparticles. Encapsulation of the hydrophobic anticancer drug Everolimus resulted in the formation of intriguing non-spherical and non-symmetric pH-responsive nanostructures, such as "stomatocytes" and "multi-compartmentalized suprapolymersomes", while the pH-triggered release of the drug was also investigated. Owing to the similarities of the developed "stomatocytes" with red blood cells, in combination with their pH-responsiveness and superior stability over enzymatic degradation, they are expected to present advanced drug delivery properties and have the ability to bypass several extra- and intracellular barriers to reach and effectively treat cancer cells.


Subject(s)
Antineoplastic Agents , Everolimus , Hydrogels , Nanoparticles/chemistry , Peptides , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Everolimus/chemistry , Everolimus/pharmacokinetics , Humans , Hydrogels/chemical synthesis , Hydrogels/chemistry , Hydrogels/pharmacokinetics , Hydrogen-Ion Concentration , Neoplasms/drug therapy , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacokinetics
8.
J Control Release ; 307: 272-281, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31260753

ABSTRACT

Mutations in the tumor protein (TP53) and the mammalian target of rapamycin (mTOR) pathway have been elucidated as driver mutations in ovarian carcinomas that transform into an invasive phenotype under hypoxic conditions. Chetomin (CHE) targets the hypoxic pathway while Everolimus (EVR) acts on the mTOR pathway. Poor aqueous solubilities of both compounds limit their clinical applications. Diblock copolymer nanoplatforms of methoxy poly(ethylene glycol)2000-block-poly (lactic acid)1800 (mPEG2000-b-PLA1800) and (mPEG4000-b-PLA2200) were used to formulate individual and dual drug loaded micelles (DDM) using the solvent evaporation method. The CHE micelles (CHE-M) had a size of 21 nm with CHE loading of 0.5 mg/mL while the EVR micelles (EVR-M) and the DDM had a size around 35 and 39 nm, respectively, with EVR loading up to 2.3 mg/mL. The anti-proliferative effects of these micelles have been tested in vitro in three ovarian cell lines (ES2, OVCAR3 and TOV21G) with the DDM exhibiting a strong synergistic anti-proliferative effect in the ES2 and the TOV21G cells. The DDM were able to significantly induce tumor regression in ES2 ovarian xenograft mouse models by inhibiting angiogenesis and inducing apoptosis when compared to the individual micelles. The inhibition of hypoxia inducible factor (HIF) and the mTOR pathways has been elucidated using immunohistochemistry studies. In conclusion, we have developed a mPEG-b-PLA based micellar nanoplatform that could prevent drug resistance by delivering multiple drugs at therapeutically relevant concentrations for effectively treating ovarian carcinomas.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Disulfides/administration & dosage , Drug Carriers/administration & dosage , Everolimus/administration & dosage , Indole Alkaloids/administration & dosage , Micelles , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Disulfides/chemistry , Drug Carriers/chemistry , Drug Combinations , Drug Liberation , Drug Synergism , Everolimus/chemistry , Female , Humans , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Indole Alkaloids/chemistry , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Polyesters/administration & dosage , Polyesters/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
9.
J Korean Med Sci ; 34(19): e145, 2019 May 20.
Article in English | MEDLINE | ID: mdl-31099195

ABSTRACT

BACKGROUND: Patients with acute myocardial infarction (AMI) have worse clinical outcomes than those with stable coronary artery disease despite revascularization. Non-culprit lesions of AMI also involve more adverse cardiovascular events. This study aimed to investigate the influence of AMI on endothelial function, neointimal progression, and inflammation in target and non-target vessels. METHODS: In castrated male pigs, AMI was induced by balloon occlusion and reperfusion into the left anterior descending artery (LAD). Everolimus-eluting stents (EES) were implanted in the LAD and left circumflex (LCX) artery 2 days after AMI induction. In the control group, EES were implanted in the LAD and LCX in a similar fashion without AMI induction. Endothelial function was assessed using acetylcholine infusion before enrollment, after the AMI or sham operation, and at 1 month follow-up. A histological examination was conducted 1 month after stenting. RESULTS: A total of 10 pigs implanted with 20 EES in the LAD and LCX were included. Significant paradoxical vasoconstriction was assessed after acetylcholine challenge in the AMI group compared with the control group. In the histologic analysis, the AMI group showed a larger neointimal area and larger area of stenosis than the control group after EES implantation. Peri-strut inflammation and fibrin formation were significant in the AMI group without differences in injury score. The non-target vessel of the AMI also showed similar findings to the target vessel compared with the control group. CONCLUSION: In the pig model, AMI events induced endothelial dysfunction, inflammation, and neointimal progression in the target and non-target vessels.


Subject(s)
Endothelium/physiology , Immunosuppressive Agents/therapeutic use , Inflammation/pathology , Myocardial Infarction/drug therapy , Neointima/pathology , Acute Disease , Animals , Arteries/pathology , Blood Cell Count , Disease Models, Animal , Drug-Eluting Stents , Everolimus/chemistry , Everolimus/therapeutic use , Immunosuppressive Agents/chemistry , Myocardial Infarction/pathology , Myocardium/pathology , Swine , Treatment Outcome
10.
J Biomater Appl ; 33(9): 1232-1241, 2019 04.
Article in English | MEDLINE | ID: mdl-30776991

ABSTRACT

Everolimus inhibits stent restenosis and the WKYMV (fluorescein isothiocyanate) peptide promotes endothelial homing. Dextran is a natural polymer that is widely used as a pharmaceutical agent. The purpose of this study was to develop a double-drug-coated stent using a bidirectional coating system and to examine the surface shape with in vitro experiments. Stent length was 16 mm and strut thickness was 70 µm (Chonnam National University Hospital Tiger stent). Optical and scanning electron microscopy showed good coating without cracks or bubbles. Fluorescein isothiocyanate-peptide was dip-coated on the lumen and the abluminal surface was coated with everolimus and dextran. Stents were coated with dextran, everolimus, or everolimus-dextran. The radial force and flexibility were measured to determine the mechanical properties. Contact angle testing was performed in all groups. Dextran and peptide as hydrophilic substances and everolimus as a hydrophobic substance were each coated on cover glasses (cobalt-chromium). A10 and human umbilical vein endothelial cells were used in the experiments. Water and dimethyl sulfoxide served as a control, and three drug groups were tested: peptide-everolimus, everolimus-dextran, and peptide-everolimus-dextran. Immunocytochemistry was performed to assess cell adhesion. Light intensity was plotted according to the average on nuclear staining. Experiments were conducted using 5-bromo-2'-deoxyuridine to investigate A10 and human umbilical vein endothelial cell proliferation. Cell adhesion and proliferation of peptide-everolimus-dextran were inhibited at A10, and human umbilical vein endothelial cell was found to proliferate with cell adhesion. On conclusion, dextran and peptide-everolimus bidirectional stent is effective in re-endothelialization and inhibition of cell proliferation.


Subject(s)
Dextrans/administration & dosage , Drug-Eluting Stents , Everolimus/administration & dosage , Oligopeptides/administration & dosage , Animals , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Dextrans/chemistry , Dextrans/pharmacology , Drug Delivery Systems , Everolimus/chemistry , Everolimus/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Hydrophobic and Hydrophilic Interactions , Oligopeptides/chemistry , Oligopeptides/pharmacology , Rats
11.
PLoS One ; 14(1): e0209841, 2019.
Article in English | MEDLINE | ID: mdl-30629613

ABSTRACT

Current drug-eluting stents have abluminal polymer coating; however, thrombus formation in these compared with that in uniformly coated stents remains controversial. We evaluated thrombus formation and early endothelialization after using abluminal biodegradable polymer-coated sirolimus- (BP-SES), and everolimus-eluting stents (BP-EES) versus a durable polymer-coated everolimus-eluting stent (DP-EES) in an in vivo setting. BP-SES, BP-EES, and DP-EES (n = 6 each) were implanted in coronary arteries of 12 mini-pigs that were then sacrificed after 7 and 10 days. Stents were stained with hematoxylin and eosin, and a combined Verhoeff and Masson trichrome stain. Areas of fibrin deposition were digitally detected and measured with off-line morphometric software. Stents were investigated for re-endothelialization by transmission electron microscopy. At 7 days, histological analysis revealed the lowest area of fibrin deposition in BP-SES (BP-SES vs. BP-EES vs. DP-EES; 0.10 ± 0.06 mm2 vs. 0.15 ± 0.07 mm2 vs. 0.19 ± 0.06 mm2, p = 0.0004). At 10 days, the area of fibrin deposition was significantly greater in DP-EES (0.13 ± 0.04 mm2 vs. 0.14 ± 0.05 mm2 vs. 0.19 ± 0.08 mm2, p = 0.007). Endothelial cells in BP-SES demonstrated a significantly greater number of tight junctions than those in DP-EES according to by transmission electron microscopy for both days (p<0.05). Various parameters, including an inflammatory reaction and neointimal formation, were comparable among the groups at 7 and 10 days. An abluminal biodegradable polymer-coated SES showed the least fibrin deposition and greatest endothelial cell recovery at an early stage following implantation in the coronary arteries of mini-pigs.


Subject(s)
Absorbable Implants , Coronary Vessels/surgery , Drug-Eluting Stents , Polymers/chemistry , Animals , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible/administration & dosage , Coated Materials, Biocompatible/chemistry , Coronary Angiography/methods , Coronary Artery Disease/surgery , Endothelial Cells/metabolism , Everolimus/administration & dosage , Everolimus/chemistry , Fibrin/metabolism , Models, Animal , Percutaneous Coronary Intervention , Polymers/administration & dosage , Polymers/adverse effects , Prosthesis Design , Sirolimus/administration & dosage , Sirolimus/chemistry , Swine , Swine, Miniature , Thrombosis/etiology , Thrombosis/metabolism , Treatment Outcome
12.
PLoS One ; 13(10): e0205046, 2018.
Article in English | MEDLINE | ID: mdl-30289945

ABSTRACT

OBJECTIVE: Data concerning the effect of current smoking on solely new-generation drug-eluting stents (DES) are limited. We investigated the impact of current smoking on 2-year clinical outcomes between durable-polymer (DP)-coated DES (zotarolimus-eluting [ZES] and everolimus eluting [EES]) and biodegradable-polymer (BP)-coated biolimus-eluting stent (BES) in acute myocardial infarction (AMI) patients after successful percutaneous coronary intervention (PCI). METHODS: Finally, a total of 8357 AMI patients with current smoking underwent successful PCI with new-generation DES (ZES, EES, and BES) were enrolled and divided into three groups as ZES (n = 3199), EES (n = 3987), and BES group (n = 1171). The primary endpoint was the occurrence of major adverse cardiac events (MACE) defined as all-cause death (cardiac death [CD] or non-cardiac death), recurrent AMI (re-MI), any revascularization (target lesion revascularization [TLR], target vessel revascularization [TVR], and non-TVR). The secondary endpoint was the incidence of definite or probable stent thrombosis (ST). RESULTS: The 2-year adjusted hazard ratio (HR) of MACE for ZES vs. EES (1.055; 95% confidence interval [CI], 0.843-1.321; p = 0.638), ZES vs. BES (HR, 0.885; 95% CI, 0.626-1.251; p = 0.488), EES vs. BES (HR, 0.889; 95% CI, 0.633-1.250; p = 0.499), and ZES/EES vs. BES (HR, 0.891; 95% CI, 0.648-1.126; p = 0.480) were similar. The occurrence of ST after adjustment were also comparable. In addition, the 2-year adjusted HR for all-cause death, CD, re-MI, TLR, TVR, and non-TVR were not different. CONCLUSIONS: In this study, DP-DES and BP-DES showed comparable safety and efficacy during 2-year follow-up periods. Therefore, DP-DES or BP-DES are equally acceptable in AMI patients with current smoking undergoing PCI.


Subject(s)
Drug-Eluting Stents , Myocardial Infarction/surgery , Percutaneous Coronary Intervention , Polymers/chemistry , Smoking/adverse effects , Everolimus/chemistry , Everolimus/pharmacology , Female , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Polymers/metabolism , Sirolimus/analogs & derivatives , Sirolimus/chemistry , Sirolimus/pharmacology , Thrombosis/etiology , Thrombosis/prevention & control
13.
Mol Pharm ; 15(9): 3672-3681, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29863881

ABSTRACT

Clinical studies examining the combination of paclitaxel (PTX) and everolimus (EVER), an mTOR inhibitor, have failed to result in significant improvements in efficacy and toxicity in patients with breast cancer (BC), relative to treatment with PTX alone. These disappointing clinical trial results have been attributed to poorly designed preclinical studies using the combination of PTX and EVER as well as the significantly different pharmacokinetic profiles of the two drugs. In the current work, the potential synergy between PTX and EVER was examined in a panel of six BC cell lines that differ in terms of their molecular subtype and drug sensitivity. Polymeric nanoparticles (NPs) were used to encapsulate PTX and EVER at an optimal synergistic ratio to achieve specific, colocalized delivery of the combination therapy in BC cell lines. Combinations of PTX and EVER (especially at relatively high doses of EVER) resulted in pronounced synergy in all BC cell lines evaluated. The optimal molar ratio of PTX:EVER was determined to be 1:0.5. The combination was delivered to BC cells at the synergistic ratio via encapsulation within polymeric NPs formed from the poly(ethylene glycol)- b-poly(lactide- co-glycolide) (PEG- b-PLGA) copolymer. The NPs had an average diameter of less than 100 nm and were capable of in vitro retention of the encapsulated PTX and EVER at the optimal synergistic molar ratio for over 7 days. Cytotoxicity data demonstrated that PTX+EVER-loaded NPs were significantly less cytotoxic than the free drug combination in MCF-7 and SKBR3 BC cell lines following 72 h, suggesting that PTX+EVER-loaded NPs remain stable and retain the drug combination loaded within the core after 72 h. The uptake of FITC-labeled NPs in SKBR3 cells was evaluated by flow cytometry, with approximately 41% of cells demonstrating detectable fluorescence after 24 h of exposure. The thorough and systematic approach used in this study to determine and evaluate a synergistic PTX:EVER ratio in conjunction with a potentially promising delivery vector for the drug combination could offer a future clinical benefit for patients with BC.


Subject(s)
Breast Neoplasms/drug therapy , Everolimus/chemistry , Everolimus/therapeutic use , Nanoparticles/chemistry , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Delivery Systems/methods , Drug Synergism , Female , Flow Cytometry , Humans , MCF-7 Cells , Polymers/chemistry
14.
Drug Des Devel Ther ; 12: 911-920, 2018.
Article in English | MEDLINE | ID: mdl-29719377

ABSTRACT

AIM: The mammalian target of rapamycin (mTOR) pathway is a critical target for cancer treatment and the mTOR inhibitor everolimus (RAD001) has been approved for treatment of renal cell carcinoma (RCC). However, the limited efficacy of RAD001 has led to the development of drug resistance. Autophagy is closely related to cell survival and death, which may be activated under RAD001 stimulation. The aim of the present study was to identify the underlying mechanisms of RAD001 resistance in RCC cells through cytoprotective autophagy involving activation of the extracellular signal-regulated kinase (ERK) pathway. METHODS AND RESULTS: RAD001 strongly induced autophagy of RCC cells in a dose- and time-dependent manner, as confirmed by Western blot analysis. Importantly, suppression of autophagy by the pharmacological inhibitor chloroquine effectively enhanced RAD001-induced apoptotic cytotoxicity, as demonstrated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Western blot analysis, indicating a cytoprotective role for RAD001-induced autophagy. In addition, as was shown by the MTT assay, flow cytometry, and Western blot analysis, RAD001 robustly activated ERK, but not c-Jun N-terminal kinase and p38. Activation of ERK was inhibited by the pharmacological inhibitor selumetinib (AZD6244), which effectively promoted RAD001-induced cell death. Moreover, employing AZD6244 markedly attenuated RAD001-induced autophagy and enhanced RAD001-induced apoptosis, which play a central role in RAD001-induced cell death. Furthermore, RAD001-induced autophagy is regulated by ERK-mediated phosphorylation of Beclin-1 and B-cell lymphoma 2, as confirmed by Western blot analysis. CONCLUSION: These results suggest that RAD001-induced autophagy involves activation of the ERK, which may impair cytotoxicity of RAD001 in RCC cells. Thus, inhibition of the activation of ERK pathway-mediated autophagy may be useful to overcome chemoresistance to RAD001.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Renal Cell/drug therapy , Everolimus/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Benzimidazoles/pharmacology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Everolimus/chemistry , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flow Cytometry , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
15.
J Biomed Mater Res B Appl Biomater ; 106(5): 1721-1729, 2018 07.
Article in English | MEDLINE | ID: mdl-28884951

ABSTRACT

The polymers poly(vinylidene fluoride-co-hexafluoropropylene) (PVDF-HFP) and poly(n-butyl methacrylate) (PBMA) are employed in manufacturing the XIENCE family of coronary stents. PBMA serves as a primer and adheres to both the stent and the drug coating. PVDF-HFP is employed in the drug matrix layer to hold the drug everolimus on the stent and control its release. Chemical stability of the polymers of XIENCE stents in the in-vivo environment was evaluated by pyrolysis-gas chromatography with mass spectrometry (Py-GC/MS) detection. For this evaluation, XIENCE stents explanted from porcine coronary arteries and from human coronary artery specimens at autopsy after 2-4 and 5-7 years of implantation, respectively, were compared to freshly manufactured XIENCE stents (controls). The comparison of pyrograms of explanted stent samples and controls showed identical fragmentation fingerprints of polymers, indicating that PVDF-HFP and PBMA maintained their chemical integrity after multiple years of XIENCE coronary stent implantation. The findings of the present study demonstrate the chemical stability of PVDF-HFP and PBMA polymers of the XIENCE family of coronary stents in the in-vivo environment, and constitute a further proof of the suitability of PVDF-HFP as a drug carrier for the drug eluting stent applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1721-1729, 2018.


Subject(s)
Coronary Vessels , Drug-Eluting Stents , Everolimus , Materials Testing , Animals , Coronary Vessels/metabolism , Coronary Vessels/pathology , Coronary Vessels/surgery , Everolimus/chemistry , Everolimus/pharmacokinetics , Everolimus/pharmacology , Female , Humans , Male , Swine
16.
J Pharm Biomed Anal ; 149: 106-113, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29112898

ABSTRACT

While the therapeutic drug monitoring (TDM) of everolimus has been routinely performed for over 10 years in solid organ transplantation medicine, in order to optimize the balance between effectiveness and toxicity, it is yet uncommon in the treatment of malignancies. The aim of this study was to develop and validate a bioanalytical method to quantify everolimus in dried blood spots (DBS) to facilitate TDM for the oncology outpatient setting. The hematocrit effect of everolimus was investigated. An 7.5mm disk from the central part of the DBS was punched, followed by the extraction of everolimus from the DBS by methanol/acetonitrile (80/20%) spiked with deuterium-labelled everolimus as internal standard. Subsequently, everolimus was separated and analyzed using ultra performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). The UPLC-MS/MS method was validated according to the European Medicine Agency (EMA) guideline. Everolimus concentrations could be quantified over the range of 3-75µg/L. The intra- and inter-assay precision and accuracy of the method were shown to be acceptable (coefficient of variation ≤10.7% and relative error ≤4.4%, respectively). The matrix effects appeared to be influenced by the hematocrit effect. The hematocrit effect was tested in a range of 0.20-0.50L/L, at which hematocrit accuracy and precision were satisfactory at values ≥0.25L/L. However, at 0.20L/L hematocrit in combination with high everolimus concentrations of 20 and 40µg/L, the precision was adequate (≤7.4%), but the accuracy was >15% of the nominal concentration. Everolimus was stable in DBS for at least 80days at 2-8°C. Given these results, the everolimus DBS method has been successfully developed and validated. Special attention is necessary for cancer patients with both a 0.20L/L hematocrit in combination with everolimus concentrations ≥20µg/L. A clinical validation for the use of everolimus DBS in cancer patients is currently being undertaken.


Subject(s)
Antineoplastic Agents/blood , Drug Monitoring/methods , Everolimus/blood , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Dried Blood Spot Testing/instrumentation , Dried Blood Spot Testing/methods , Dried Blood Spot Testing/standards , Drug Monitoring/instrumentation , Drug Monitoring/standards , Drug Stability , European Union , Everolimus/chemistry , Everolimus/therapeutic use , Guidelines as Topic , Hematocrit , Humans , Neoplasms/blood , Reproducibility of Results , Tandem Mass Spectrometry/instrumentation , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards , Treatment Outcome
17.
J Cell Physiol ; 232(8): 2063-2074, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27607841

ABSTRACT

The aim of this study is based on the evaluation of anticancer, anti-inflammatory activities, and cellular uptake of hyaluronic acid nanohydrogel of quercetin tested alone and in combination to a macrolide derivative of rapamycin RAD001 (everolimus) on hormone-responsive breast cancer cell line MCF-7. Biological investigations were focused on the receptor mediated cellular internalization of the nanohydrogel and its abilities to reduce secretion of several cytokines (IL-8, IL-6, IL-19), VEGF, and metalloproteases (MMP-2, MMP-9) under pro-inflammatory conditions. Nanohydrogel show a CD44 dependent endocytosis with evident time dependent cytoplasmatic accumulation with abilities to reduce secretion of all cytokines of ∼60% compared to untreated cells. Combination of formulated quercetin and everolimus leads to a synergistic cytotoxic effects with a Combination Index of 0.38. These results highlights the importance of synergistic effect of the hyaluronic acid nanohydrogel of quercetin with everolimus in the regulation of human breast cancer cell proliferation and emphasize the antitumor and anti-inflammatory properties of the nanocarrier. J. Cell. Physiol. 232: 2063-2074, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Carriers , Everolimus/pharmacology , Hyaluronic Acid/pharmacology , Nanoparticles , Neoplasms, Hormone-Dependent/drug therapy , Quercetin/pharmacology , Anti-Inflammatory Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Compounding , Drug Synergism , Everolimus/chemistry , Female , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Hydrogels , Interleukins/metabolism , MCF-7 Cells , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Necrosis , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Quercetin/chemistry , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
18.
Cancer Treat Rev ; 50: 247-263, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27776286

ABSTRACT

Dissolution from the pharmaceutical formulation is a prerequisite for complete and consistent absorption of any orally administered drug, including anticancer agents (oncolytics). Poor dissolution of an oncolytic can result in low oral bioavailability, high variability in blood concentrations and with that suboptimal or even failing therapy. This review discusses pharmaceutical formulation aspects and absorption pharmacokinetics of currently licensed orally administered oncolytics. In nearly half of orally dosed oncolytics poor dissolution is likely to play a major role in low and unpredictable absorption. Dissolution-limited drug absorption can be improved with a solid dispersion which is a formulation method that induces super-saturated drug dissolution and with that it enhances in vivo absorption. This review discusses formulation principles with focus on the solid dispersion technology and how it works to enhance drug absorption. There are currently three licensed orally dosed oncolytics formulated as a solid dispersion (everolimus, vemurafenib and regorafenib) and these formulations result in remarkably improved dissolution and absorption compared to what can be achieved with conventional formulations of the respective oncolytics. Because of the successful implementation of these three solid dispersion formulations, we encourage the application of this formulation method for poorly soluble oral oncolytics.


Subject(s)
Antineoplastic Agents/administration & dosage , Everolimus/administration & dosage , Indoles/administration & dosage , Phenylurea Compounds/administration & dosage , Pyridines/administration & dosage , Sulfonamides/administration & dosage , Administration, Oral , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Dosage Forms , Everolimus/chemistry , Everolimus/pharmacokinetics , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Solubility , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Vemurafenib
19.
Chem Pharm Bull (Tokyo) ; 64(10): 1445-1449, 2016.
Article in English | MEDLINE | ID: mdl-27725499

ABSTRACT

The aims of this study were to improve in vitro dissolution property of poorly water-soluble everolimus (EVR) for enhanced bioavailability without using organic solvents and characterize the effects of microfluidization and freeze-drying on physicochemical properties of EVR nanosuspension and nanoparticle, respectively. EVR nanosuspension was prepared using microfluidization with various types and concentrations of stabilizers. After that, it was solidified into nanoparticle using freeze-drying with various concentrations of xylitol, a cryoprotectant. The particle size, zeta potential, physical stability, and chemical stability of EVR nanosuspension and nanoparticle were measured. In vitro release of EVR nanoparticle was also measured and compared with that of physical mixture. Zero point five percent (w/w) poloxamer 407 (P407) was chosen as the stabilizer considering particle size, zeta potential, and yield of EVR nanosuspension. Freeze-drying with 1% (w/w) xylitol improved both physical and chemical stability of EVR nanoparticle. In vitro release test showed improved dissolution property compared to that of physical mixture, implying enhanced bioavailability.


Subject(s)
Everolimus/chemistry , Microfluidics/methods , Nanoparticles/chemistry , Freeze Drying , Particle Size , Solubility , Surface Properties
20.
Br J Pharmacol ; 173(23): 3359-3371, 2016 12.
Article in English | MEDLINE | ID: mdl-27638766

ABSTRACT

BACKGROUND AND PURPOSE: Everolimus is an allosteric inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1) widely known for its potent autophagy stimulating properties. Because everolimus shows poor solubility and stability in aqueous solutions, long-term in vivo administration in preclinical models is challenging. The aim of the present study was to evaluate the effects of short-term and long-term everolimus administration on mTORC1 inhibition and autophagy induction in mice. EXPERIMENTAL APPROACH: We developed a vehicle in which everolimus was solubilized and stable at 37°C for at least 1 month. Using osmotic minipumps, GFP microtubule-associated protein light chain 3 transgenic mice were treated continuously either with vehicle or everolimus (1.5 mg·kg-1 per day) for 3 or 28 days. Alternatively, a regimen consisting of intermittent everolimus administration (every other day) for 56 days by oral gavage was used. Autophagy markers and mTORC1 activation status were investigated in the liver. KEY RESULTS: As expected, everolimus inhibited mTORC1 and stimulated autophagy in the liver after 3 days of treatment. However, continuous administration for 28 days resulted in hyperactivation of the Akt1-mTORC1 pathway accompanied by a remarkable decrease in autophagy markers. Everolimus given intermittently for 56 days partially rescued mTORC1 sensitivity to the drug but without inducing autophagy. The failure to induce autophagy following long-term everolimus administration was due to uncoupling of the mTORC1 substrate unc-51 like autophagy activating kinase 1. CONCLUSIONS AND IMPLICATIONS: Our data encourage the use of intermittent everolimus regimens to prevent tolerance and to extend its activity.


Subject(s)
Autophagy/drug effects , Drug Tolerance , Everolimus/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Animals , Drug Administration Schedule , Drug Stability , Everolimus/administration & dosage , Everolimus/chemistry , Female , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, Transgenic , Solubility , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...