Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Publication year range
1.
Prostate ; 83(6): 534-546, 2023 05.
Article in English | MEDLINE | ID: mdl-36597263

ABSTRACT

We have shown that decursin, a coumarin compound, induces cell cycle arrest and apoptosis in human prostate cancer cells (PCa); however, its molecular mechanisms are largely unexplored. We studied the mechanisms associated with its anticancer activity in advanced human prostate carcinoma cells. We found that decursin inhibited epidermal growth factor receptor (EGFR) signaling by inhibiting its activating phosphorylation at tyrosine 1068 residue in DU145 and 22Rv1 cells. This inhibition of EGFR was associated with the downregulation of ERK1/2 phosphorylation. Both EGFR and ERK1/2 are known to be deregulated/activated in many human malignancies. Consistent with our earlier study, decursin (25-100 µM) treatment for 24-72 h inhibited DU145 cell proliferation by 49%-87% (p < 0.001) which was associated with strong G1 phase arrest and cell death. It also decreased (p < 0.001) the number of surviving colonies. Decursin moderately increased the expression of Rb-related proteins p107 and p130 but decreased the levels of E2F family transcription factors including E2F-3, E2F-4 and E2F-5. Further, decursin strongly inhibited the growth of androgen-dependent prostate carcinoma 22Rv1 cells from 61% to 79% (p < 0.001) and arrested these cells at G1 phase via induction of cyclin-dependent kinase inhibitor p27/Kip1 and downregulation of CDK2 and CDK4 protein expression. Additionally, EGFR inhibitor erlotinib- and EGF ligand-modulated EGFR activation validated EGFR signaling as a target of decursin-mediated cell growth inhibition and cytotoxicity. Decursin decreased EGF ligand-induced phosphorylation of EGFR (Y-1068) as well as activation of its downstream mediator, ERK1/2. Furthermore, inhibitory targeting of EGFR-ERK1/2 axis by combinatorial treatment of decursin and erlotinib further sensitized DU145 cells for the decursin-induced growth inhibition and cell death. Overall, these findings strongly suggest that anticancer efficacy of decursin against human PCa involves inhibitory targeting of EGFR-ERK1/2 signaling axis, a pathway constitutively active in advanced PCa.


Subject(s)
Carcinoma , Prostatic Neoplasms , Male , Humans , Epidermal Growth Factor , MAP Kinase Signaling System , Prostate/pathology , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/metabolism , Ligands , ErbB Receptors/metabolism , Phosphorylation , Prostatic Neoplasms/pathology , Carcinoma/metabolism
2.
Int J Med Sci ; 19(10): 1510-1518, 2022.
Article in English | MEDLINE | ID: mdl-36185336

ABSTRACT

Background: The most common spinal disorder in elderly is lumbar spinal canal stenosis (LSCS). Previous studies showed that ligamentum flavum hypertrophy (LFH) with fibrosis as the main pathological change is one of the pathogenic factors leading to LSCS. Epidermal Growth Factor (EGF) is known to have an intimate relationship with fibrosis in various tissues. Nevertheless, currently, there are few studies regarding EGF in LFH. The effect of EGF on the development of LFH is unknown, and the underlying pathomechanism remains unclear. In this study, we investigated the role of EGF in LFH and its potential molecular mechanism. Methods: First, the expression levels of EGF, phosphorylation of EGF receptor (pEGFR), Transforming growth factor-ß1 (TGF-ß1), Phosphorylated Smad3 (pSmad3), collagen I and collagen III were examined via immunohistochemistry and Western blot in LF tissues from patients with LSCS or Non-LSCS. Second, primary LF cells were isolated from adults with normal LF thickness and were cultured with different concentrations of exogenous EGF with or without erlotinib/TGF-ß1-neutralizing antibody. Results: The results showed that EGF, pEGFR, TGF-ß1, pSmad3, collagen I and collagen III protein expression in the LSCS group was significantly higher than that in the Non-LSCS group. Meanwhile, pEGFR, TGF-ß1, pSmad3, collagen I and collagen III protein expression was significantly enhanced in LF cells after exogenous EGF exposure, which can be notably blocked by erlotinib. In addition, pSmad3, collagen I and collagen III protein expression was blocked by TGF-ß1-neutralizing antibody. Conclusions: EGF promotes the synthesis of collagen I and collagen III via the TGF-ß1/Smad3 signaling pathway, which eventually contributes to LFH.


Subject(s)
Ligamentum Flavum , Spinal Stenosis , Adult , Aged , Antibodies, Neutralizing/metabolism , Collagen/metabolism , Collagen Type I/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Erlotinib Hydrochloride/metabolism , Fibrosis , Humans , Hypertrophy/metabolism , Ligamentum Flavum/metabolism , Ligamentum Flavum/pathology , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Spinal Stenosis/metabolism , Spinal Stenosis/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
3.
Cell Mol Life Sci ; 78(1): 207-225, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32140747

ABSTRACT

NAD(P)-dependent steroid dehydrogenase-like (NSDHL), an essential enzyme in human cholesterol synthesis and a regulator of epidermal growth factor receptor (EGFR) trafficking pathways, has attracted interest as a therapeutic target due to its crucial relevance to cholesterol-related diseases and carcinomas. However, the development of pharmacological agents for targeting NSDHL has been hindered by the absence of the atomic details of NSDHL. In this study, we reported two X-ray crystal structures of human NSDHL, which revealed a detailed description of the coenzyme-binding site and the unique conformational change upon the binding of a coenzyme. A structure-based virtual screening and biochemical evaluation were performed and identified a novel inhibitor for NSDHL harboring suppressive activity towards EGFR. In EGFR-driven human cancer cells, treatment with the potent NSDHL inhibitor enhanced the antitumor effect of an EGFR kinase inhibitor. Overall, these findings could serve as good platforms for the development of therapeutic agents against NSDHL-related diseases.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Enzyme Inhibitors/metabolism , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/genetics , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Kinetics , Molecular Docking Simulation , Mutagenesis, Site-Directed , NAD/chemistry , NAD/metabolism , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Signal Transduction
4.
J Pharmacol Exp Ther ; 374(2): 295-307, 2020 08.
Article in English | MEDLINE | ID: mdl-32393528

ABSTRACT

Gefitinib and erlotinib are epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) with activity against metastatic non-small cell lung cancer. Aldehyde oxidase-1 (AOX1) is a cytosolic drug-metabolizing enzyme. We conducted an experimental and molecular docking study on the effect of gefitinib, erlotinib, and select metabolites on the in vitro catalytic activity of AOX1, as assessed by carbazeran 4-oxidation, and determined the impact of AOX1 inhibition on hepatic metabolism of zaleplon and methotrexate. Gefitinib, desmorpholinopropylgefitinib, erlotinib, desmethylerlotinib, and didesmethylerlotinib inhibited human hepatic cytosolic carbazeran 4-oxidation by a competitive mode, with inhibition constants in submicromolar or low micromolar concentrations. Desmethylgefitinib did not affect AOX1 catalytic activity. A similar pattern was obtained when investigated with human kidney cytosol or recombinant AOX1. The differential effect of gefitinib on human, rat, and mouse hepatic AOX1 catalytic activity suggests species-dependent chemical inhibition of AOX1. Erlotinib was considerably more potent than gefitinib in decreasing hepatic cytosolic zaleplon 5-oxidation and methotrexate 7-oxidation. Molecular docking analyses provided structural insights into the interaction between EGFR-TKIs and AOX1, with key residues and bonds identified, which provided favorable comparison and ranking of potential inhibitors. Based on the US Food and Drug Administration guidance to assess the risk of drug-drug interactions, the calculated R1 values indicate that further investigations are warranted to determine whether gefitinib and erlotinib impact AOX1-mediated drug metabolism in vivo. Overall, erlotinib desmethylerlotinib, didesmethylerlotinib, gefitinib, and desmorpholinopropylgefitinib are potent inhibitors of human AOX1 catalytic function and hepatic metabolism of zaleplon and methotrexate, potentially affecting drug efficacy or toxicity. SIGNIFICANCE STATEMENT: As epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), gefitinib and erlotinib are first-line pharmacotherapy for metastatic non-small cell lung cancer. Our experimental findings indicate that clinically relevant concentrations of gefitinib, desmorpholinopropylgefitinib, erlotinib, desmethylerlotinib, and didesmethylerlotinib, but not desmethylgefitinib, inhibit human aldehyde oxidase (AOX1) catalytic activity and hepatic cytosolic metabolism of zaleplon and methotrexate. Molecular docking analysis provide structural insights into the key AOX1 interactions with these EGFR-TKIs. Our findings may trigger improved strategies for new EGFR-TKI design and development.


Subject(s)
Acetamides/metabolism , Aldehyde Oxidase/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Gefitinib/pharmacology , Liver/drug effects , Methotrexate/metabolism , Molecular Docking Simulation , Pyrimidines/metabolism , Aldehyde Oxidase/chemistry , Aldehyde Oxidase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Erlotinib Hydrochloride/metabolism , Gefitinib/metabolism , Humans , Liver/metabolism , Protein Conformation
5.
Bioorg Med Chem Lett ; 30(23): 127557, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32949719

ABSTRACT

Epidermal growth factor receptor tyrosine kinase (EGFR-TK) has been proved as a target for the treatment of non-small cell lung cancer (NSCLC) with specific gene mutations. However, EGFR-TK inhibitors (EGFR-TKIs) need to enter cancer cells and then competitively interact with the active site of tyrosine kinase receptors to suppress the downstream signaling pathway to inhibit tumor proliferation. In this study, in order to improve the tumor cell targeting ability of EGFR-TKI, EGFR-TKI erlotinib was conjugated with the cancer cell-targeting heptamethine cyanine dyes to form seventeen novel erlotinib-dye conjugates. The efficiency of tumor targeting properties of conjugates against cancer cell growth and EGFR-TK inhibition was evaluated in vitro. The result revealed that most erlotinib-dye conjugates exhibited stronger inhibitory effect on A549, H460, H1299 and MDA-MB-231 cell lines than the parent drug erlotinib. Meanwhile, representative compounds exhibited weak cytotoxicity on human normal mammary epithelial MCF-10A cells. Moreover, the conjugate CE17 also showed ~14-fold higher EGFR-TK inhibition activity (IC50 = 0.124 µM) than erlotinib (IC50 = 5.182 µM) in A549 cell line. Finally, molecular docking analysis verified that the erlotinib moiety of compound CE17 could form hydrogen bond with Met-769 and occupy active cavity of EGFR-TK. Therefore, we believed the integration strategy between heptamethine cyanine dyes and EGFR-TKI will contribute to enhancing the therapeutic effect of EGFR-TKI for NSCLC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemical synthesis , Erlotinib Hydrochloride/metabolism , Humans , Indoles/chemical synthesis , Indoles/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Structure-Activity Relationship
6.
J Biol Chem ; 293(50): 19211-19212, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552114

ABSTRACT

The cytochromes P450 (CYPs) oxidatively transform a huge number of substrates in both prokaryotic and eukaryotic organisms, but the mechanisms by which they accommodate these diverse molecules remain unclear. A new study by Bart and Scott reports two co-crystal structures of CYP1A1 that reveal structural rearrangements and flexible interaction networks that explain how the active site cavity shapes itself around new ligands. These data open the door to an increased understanding of fundamental enzyme behavior and improved searches for anti-cancer compounds.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Enzyme Inhibitors/metabolism , Erlotinib Hydrochloride/metabolism , Furocoumarins/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytochrome P-450 CYP1A1/chemistry , Enzyme Inhibitors/chemistry , Erlotinib Hydrochloride/chemistry , Furocoumarins/chemistry , Humans , Ligands , Protein Binding , Substrate Specificity
7.
J Biol Chem ; 293(50): 19201-19210, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30254074

ABSTRACT

Human cytochrome P450 1A1 (CYP1A1) is an extrahepatic enzyme involved in the monooxygenation of structurally diverse compounds ranging from natural products to drugs and protoxins. Because CYP1A1 has a role in human carcinogenesis, inhibiting its activity may potentially aid in cancer chemoprevention, whereas utilizing CYP1A1's oxidative activity could help selectively activate anticancer prodrugs. Such potential therapeutic purposes require detailed knowledge of CYP1A1's interactions with potential ligands. Known CYP1A1 ligands also vary substantially in size, and it has not been apparent from a single existing CYP1A1 structure how larger, structurally diverse ligands are accommodated within the enclosed active site. Here, two new X-ray structures with the natural product furanocoumarin bergamottin (at 2.85 Å resolution) and the lung cancer drug erlotinib (3.0 Å) revealed binding orientations consistent with the formation of innocuous metabolites and of toxic metabolites, respectively. They also disclosed local changes in the roof of the active site that enlarge the active site and ultimately form a channel to the protein exterior. Although further structural modifications would be required to accommodate the largest CYP1A1 ligands, knowing which components of the active site are malleable provides powerful information for those attempting to use computational approaches to predict compound binding and substrate metabolism by this clinically relevant monooxygenase.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Enzyme Inhibitors/metabolism , Erlotinib Hydrochloride/metabolism , Furocoumarins/metabolism , Catalytic Domain , Crystallography, X-Ray , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/chemistry , Enzyme Assays , Enzyme Inhibitors/chemistry , Erlotinib Hydrochloride/chemistry , Furocoumarins/chemistry , Humans , Ligands , Protein Binding , Substrate Specificity
8.
Anal Chem ; 91(15): 10026-10032, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31282657

ABSTRACT

Low-abundance phosphotyrosine (pTyr)-mediated signaling protein complexes play critical roles in cancer signaling. The precise and comprehensive profiling of these pTyr-mediated protein complexes remains challenging because of their dynamic nature and weak binding affinity. Taking advantage of the SH2 domains modified with trifunctional chemical probes and genetic mutations (termed Photo-pTyr-scaffold), we developed a Photo-pTyr-scaffold-based forward-phase protein array that can be used to specifically capture complexes by developing an engineered SH2 domain, photoaffinity cross-linking, and antibody-based measuring weak pTyr-mediated protein complexes from complex biological samples in a 96-well microplate format. This platform demonstrated good precision for quantitation (R2 = 0.99) and high sensitivity by which only 5 µg of whole cell lysates is needed. We successfully applied the technology for profiling the dynamic EGF-stimulation-dependent EGFR signaling protein complexes across four different time courses (i.e., 0, 2, 5, 10, and 30 min) in a high-throughput manner. We further evaluated the modulation of EGFR-GRB2-SHC1 protein complexes by FDA-approved EGFR kinase inhibitor erlotinib, demonstrating the feasibility of this approach for high-throughput drug screening. The Photo-pTyr-scaffold-based forward-phase protein array could be generically applicable for exploring the dynamic pTyr signaling complexes in various biological systems and screening for related drugs in a high-throughput manner.


Subject(s)
Phosphotyrosine/metabolism , Protein Array Analysis/methods , Ultraviolet Rays , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/metabolism , High-Throughput Screening Assays , Humans , Phosphotyrosine/chemistry , Protein Binding , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1/chemistry , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , src Homology Domains
9.
Mol Pharm ; 16(3): 1282-1293, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30694684

ABSTRACT

P-Glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are two efflux transporters at the blood-brain barrier (BBB), which effectively restrict brain distribution of diverse drugs, such as tyrosine kinase inhibitors. There is a crucial need for pharmacological ABCB1 and ABCG2 inhibition protocols for a more effective treatment of brain diseases. In the present study, seven marketed drugs (osimertinib, erlotinib, nilotinib, imatinib, lapatinib, pazopanib, and cyclosporine A) and one nonmarketed drug (tariquidar), with known in vitro ABCB1/ABCG2 inhibitory properties, were screened for their inhibitory potency at the BBB in vivo. Positron emission tomography (PET) using the model ABCB1/ABCG2 substrate [11C]erlotinib was performed in mice. Tested inhibitors were administered as i.v. bolus injections at 30 min before the start of the PET scan, followed by a continuous i.v. infusion for the duration of the PET scan. Five of the tested drugs increased total distribution volume of [11C]erlotinib in the brain ( VT,brain) compared to vehicle-treated animals (tariquidar, + 69%; erlotinib, + 19% and +23% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 22%; lapatinib, + 25%; and cyclosporine A, + 49%). For all drugs, increases in [11C]erlotinib brain distribution were lower than in Abcb1a/b(-/-)Abcg2(-/-) mice (+149%), which suggested that only partial ABCB1/ABCG2 inhibition was reached at the mouse BBB. The plasma concentrations of the tested drugs at the time of the PET scan were higher than clinically achievable plasma concentrations. Some of the tested drugs led to significant increases in blood radioactivity concentrations measured at the end of the PET scan (erlotinib, + 103% and +113% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 125%; and cyclosporine A, + 101%), which was most likely caused by decreased hepatobiliary excretion of radioactivity. Taken together, our data suggest that some marketed tyrosine kinase inhibitors may be repurposed to inhibit ABCB1 and ABCG2 at the BBB. From a clinical perspective, moderate increases in brain delivery despite the administration of high i.v. doses as well as peripheral drug-drug interactions due to transporter inhibition in clearance organs question the translatability of this concept.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Erlotinib Hydrochloride/metabolism , Protein Kinase Inhibitors/metabolism , Radiopharmaceuticals/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Animals , Capillary Permeability/physiology , Cyclosporine/administration & dosage , Cyclosporine/blood , Cyclosporine/metabolism , Cyclosporine/pharmacology , Drug Interactions , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/blood , Erlotinib Hydrochloride/pharmacology , Female , Mice , Models, Animal , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacology , Quinolines/administration & dosage , Quinolines/blood , Quinolines/metabolism , Quinolines/pharmacology , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacology , Solubility , Tissue Distribution
10.
Bioorg Chem ; 93: 103314, 2019 12.
Article in English | MEDLINE | ID: mdl-31590041

ABSTRACT

A series of pyrazolo[1,5-c]quinazolines as EGFR inhibitors was designed and synthesized by highly efficient and novel multicomponent route involving Pd-catalyzed tandem one-pot four-component reaction. The reaction proceeds with good functional group tolerance under a simple condition with excellent regioselectivity and high efficiency. Target compounds were screened against cancer cell lines MDA-MB-231, A549 and H1299. Of these, 9b and 10b exhibited superior anticancer activity (IC50 < 2.5 µM) to erlotinib and gefitinib. Synthetics were able to inhibit EGFR mediated kinase activity, induced ROS in cancer cells promoting mitochondrial mediated apoptosis via halting cell cycle progression at G1 phase.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Palladium/chemistry , Protein Kinase Inhibitors/chemical synthesis , Pyrazoles/chemistry , Quinazolines/chemistry , Apoptosis/drug effects , Binding Sites , Catalysis , Catalytic Domain , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrazoles/metabolism , Pyrazoles/pharmacology , Quinazolines/metabolism , Quinazolines/pharmacology , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
11.
Bioorg Chem ; 89: 102985, 2019 08.
Article in English | MEDLINE | ID: mdl-31121559

ABSTRACT

derivatives of benzo[g]indazole 5a, b, benzo[h]quinazoline 7, 12a-c, 13a-c and 15a-c and benzo[h]quinoline 17a-c and 19a-c were synthesized from 6-methoxy-3,4-dihydronaphthalen-1(2H)-one (1). Anticancer activity of all the synthesized compounds was evaluated against four cancerous cell lines; HepG2, MCF-7, HCT116 and Caco-2. MCF-7 cells emerged as the most sensitive cell line against the target compounds. All the examined compounds, except 5a and 5b, displayed potent to moderate anticancer activity against MCF-7 cells with an IC50 values ranging from 7.21 to 21.55 µM. In particular, compounds 15c and 19b emerged as the most potent derivatives against EGFR-expressing MCF-7 cells with IC50 values = 7.70 ±â€¯0.39 and 7.21 ±â€¯0.43 µM, respectively. Additionally, both compounds did not display any significant cytotoxicity towards normal BHK-21 fibroblast cells (IC50 value > 200 µM), thereby providing a good safety profile as anticancer agents. Furthermore, compounds 15c and 19b displayed potent inhibitory activity towards EGFR in the sub-micromolar range (IC50 = 0.13 ±â€¯0.01 and 0.14 ±â€¯0.01 µM, respectively), compared to that of Erlotinib (IC50 = 0.11 ±â€¯0.01 µM). Docking studies for 15c and 19b into EGFR active site was carried out to explore their potential binding modes. Therefore, compounds 15c and 19b can be considered as interesting candidates for further development of more potent anticancer agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Indazoles/chemistry , Protein Kinase Inhibitors/chemical synthesis , Quinazolines/chemistry , Quinolines/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
12.
Nurs Res ; 68(2): 110-126, 2019.
Article in English | MEDLINE | ID: mdl-30540703

ABSTRACT

BACKGROUND: Therapies targeting the epidermal growth factor receptor (EGFR) result in a painful rash, the most common and debilitating toxicity among patients with non-small cell lung cancer (NSCLC) who take EGFR tyrosine kinase inhibitor (TKI) therapy; however, predicting the development and the severity of the rash is difficult. OBJECTIVE: The aim of this study was to examine how erlotinib-an EGFR TKI that NSCLC patients take to stop or slow tumor growth-altered the transcriptome of dermal fibroblasts. METHODS: Dermal fibroblasts (ATCC PCS-201-012) were seeded in cell culture flasks, grown under standard conditions, and transferred to cell culture dishes. Cells were treated once daily for 3 days with erlotinib 100 nM (n = 5), erlotinib 1 µM (n = 5), vehicle 1 µM (dimethyl sulfoxide) (n = 5), or no treatment (n = 5). Total RNA was extracted using a standard TRIzol method and hybridized using Affymetrix GeneChip Human Genome U133 Plus 2.0 arrays. Raw intensities generated from the arrays were normalized using a Robust Multiarray Average method and analyzed using analysis of variance in Limma R software. Differentially expressed genes were analyzed using Ingenuity Pathway Analysis to identify canonical or noncanonical signaling pathways enriched in this dataset. RESULTS: We selected genes for investigation based on their potential role in wound healing (AQP3), rash development (CCL2), fibroblast activation (PALLD), cancer and cancer progression (GDF-15, SLC7A11, MMP12, and DIRAS3), and cell cycle control (CDC6). We were able to validate four of these genes by both Western blot analysis and quantitative polymerase chain reaction (MMP12, CCL2, CDC6, and SLC7A11). DISCUSSION: If found predictive of rash in future studies using patient samples, our findings may help to identify those at risk for severe rash so that (a) the dose of EGFR TKI therapy may be adjusted; (b) additional treatments for the rash can be developed; and/or (c) precise, patient-centered interventions can be developed so that patients with cancer can better self-manage their rash and adhere to EGFR TKI treatment.


Subject(s)
Antineoplastic Agents/metabolism , Erlotinib Hydrochloride/metabolism , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Protein Kinase Inhibitors/metabolism , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor/drug effects , Erlotinib Hydrochloride/administration & dosage , Gene Expression Profiling , Humans , Protein Kinase Inhibitors/administration & dosage
13.
Drug Metab Dispos ; 46(4): 442-450, 2018 04.
Article in English | MEDLINE | ID: mdl-29352069

ABSTRACT

Erlotinib (ELT), a tyrosine kinase inhibitor, is widely used for the treatment of nonsmall cell lung cancer in clinic. Unfortunately, severe drug-induced liver injury and other adverse effects occurred during the treatment. Meanwhile, ELT has been reported to be a mechanism-based inactivator of cytochrome P450(CYPs) 3A4 and 3A5. The objectives of this study were to identify ketene intermediate of ELT and investigate the association of the acetylenic bioactivation with the enzyme inactivation caused by ELT. A ketene intermediate was detected in human microsomal incubations of ELT, using 4-bromobenzylamine as a trapping agent. CYPs 3A4 and 3A5 mainly contributed to the bioactivation of ELT. Microsomal incubation study showed that the ketene intermediate covalently modified the enzyme protein at lysine residues and destroyed the structure of heme. The vinyl and ethyl analogs of ELT showed minor enzyme inhibitory effect (less than 20%), whereas ELT inactivated more than 60% of the enzyme. The present study provided a novel bioactivation pathway of ELT and facilitated the understanding of the mechanisms of ELT-induced mechanism-based enzyme inactivation and liver injury.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Erlotinib Hydrochloride/metabolism , Ethylenes/metabolism , Ketones/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Heme/metabolism , Humans , Microsomes, Liver/metabolism , Protein Binding/physiology
14.
Drug Metab Dispos ; 45(10): 1093-1100, 2017 10.
Article in English | MEDLINE | ID: mdl-28790147

ABSTRACT

The epidermal growth factor receptor (EGFR) regulates cellular expression levels of breast cancer resistance protein (humans: ABCG2, rodents: Abcg2) via its downstream signaling pathways. Drugs that inhibit EGFR signaling (e.g., tyrosine kinase inhibitors, antibodies) may lead to ABCG2-mediated drug-drug interactions (DDIs) by changing the disposition of concomitantly administered ABCG2 substrate drugs. In this study, we used positron emission tomography and magnetic resonance imaging to compare disposition of the model Abcg2 substrate [11C]erlotinib in a mouse model of hepatocyte-specific deletion of EGFR (EGFR∆hep mice, n = 5) with EGFRfl/fl control mice (n = 6), which have normal EGFR expression levels in all tissues. Integration plot analysis was used to estimate the rate constants for transfer of radioactivity from the liver into bile (kbile) and from the kidney into urine (kurine). EGFR∆hep mice showed significantly lower radioactivity concentrations in the intestine (1.6-fold) and higher radioactivity concentrations in the urinary bladder (3.2-fold) compared with EGFRfl/fl mice. Kbile was significantly decreased (3.0-fold) in EGFR∆hep mice, whereas kurine was by 2.2-fold increased. Western blot analysis of liver tissue confirmed deletion of EGFR and showed significant decreases in Abcg2 and increases in P-glycoprotein (Abcb1a/b) expression levels in EGFR∆hep versus EGFRfl/fl mice. Our data show that EGFR deletion in hepatocytes leads to a reduction in Abcg2-mediated hepatobiliary clearance of a probe substrate accompanied by a shift to renal excretion of the drug, which raises the possibility that EGFR-inhibiting drugs may cause ABCG2-mediated DDIs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ErbB Receptors/metabolism , Erlotinib Hydrochloride/metabolism , Hepatocytes/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Carbon Isotopes/metabolism , Drug Interactions/physiology , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Mice , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/metabolism , Renal Elimination/physiology , Signal Transduction/physiology
15.
Chembiochem ; 17(11): 990-4, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26991964

ABSTRACT

The receptor tyrosine kinase EGFR is regulated by complex conformational changes, and this conformational control is disturbed in certain types of cancer. Many ligands are known to bind EGFR in its active conformation, thereby preventing ATP from binding. Only a few ligands are known to stabilize EGFR in its inactive conformation, thus providing novel strategies for perturbing EGFR activity. We report a direct binding assay that enables the identification of novel ligands that bind to and stabilize the inactive conformation of EGFR.


Subject(s)
ErbB Receptors/metabolism , Protein Kinase Inhibitors/metabolism , Binding Sites , ErbB Receptors/chemistry , ErbB Receptors/genetics , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Lapatinib , Ligands , Mutagenesis, Site-Directed , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Structure, Tertiary , Quinazolines/chemistry , Quinazolines/metabolism , Spectrometry, Fluorescence
17.
Fundam Clin Pharmacol ; 37(5): 983-993, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37143385

ABSTRACT

BACKGROUND: Erlotinib is a selective epidermal growth factor receptor inhibitor that is used for the treatment of non-small cell lung cancer and pancreatic cancer. Its metabolism is mainly mediated by cytochrome P450 3A (CYP 3A). Resveratrol, a natural compound found in many plants and supplements, is known to inhibit CYP3A enzyme, therefore, it may act as an inhibitor for the metabolism of erlotinib. OBJECTIVE: Development of a rapid high performance liquid chromatography with photodiode array detection (HPLC-PDA) method for the quantification of erlotinib in liver microsomes and cancer cells and its application to study resveratrol effect on metabolism and cellular uptake of erlotinib. METHODS: HPLC-PDA was used to develop an efficient bioanalytical method with a 2.5-min runtime preceded by a simple protein precipitation step. The method was validated according to the European Medicines Agency guidelines. Erlotinib metabolic stability and resveratrol effect on erlotinib metabolite formation were evaluated in rat liver microsomes. Furthermore, the method was used to measure the intracellular concentrations of erlotinib in cancer colorectal cells and investigating resveratrol effect on the cellular uptake of erlotinib. RESULTS: A rapid HPLC-PDA method was developed and validated for the first time to address potential drug interaction of erlotinib with resveratrol. Resveratrol was a strong inhibitor of erlotinib metabolism in vitro with IC50 = 4.03 µM. Resveratrol, however, had no effect on erlotinib cellular uptake after 1 h incubation in human colorectal cancer cells. CONCLUSION: The study suggests that resveratrol may produce a potential herb-drug interaction with erlotinib at the metabolism level and should be investigated in patients in the clinic.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Rats , Animals , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/therapeutic use , Chromatography, High Pressure Liquid , Resveratrol/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/metabolism , Cytochrome P-450 CYP3A/metabolism , Microsomes, Liver/metabolism
18.
Chem Biol Drug Des ; 99(3): 456-469, 2022 03.
Article in English | MEDLINE | ID: mdl-34923743

ABSTRACT

Epidermal growth factor receptor tyrosine kinase domain (EGFR-TK) has been one of the prominent targets for therapeutics of several human cancers, in particular non-small cell lung cancer. Although several small chemical compounds targeting EGFR-TK have been approved by FDA for treatment of such a cancer, the discovery of a new class of EGFR-TK inhibitors, for example, small peptides, is still desired. In this study, using molecular docking-based virtual screening, we selected five small peptides with high docking scores from eight thousand peptides as candidate compounds against EGFR-TK. Among five, the tripeptide WFF had the most potency to suppress the survival of non-small cell lung cancer cells but had the least toxicity to human liver cancer cells. Our in vitro kinase assays showed that WFF exhibited much lower inhibitory activity against purified EGFR-TK than the drug erlotinib (i.e., IC50  values of ≈ 0.62 µM vs ≈ 7.57 nM, respectively). The relative free binding energies estimated from molecular dynamic simulations were consistent with the in vitro experiments in which the WFF bound had a lower affinity than erlotinib bound to EGFR-TK (i.e., ΔGbind values of -20.3 kJ/mol vs ≈ -126.8 kJ/mol, respectively). In addition, the simulation analyses demonstrated the difference in EGFR binding preference between the drug and tripeptide in which erlotinib was stably bound in the ATP-binding pocket for 4-anilinoquinazoline class of inhibitors, while WFF moved out of that pocket to interact with polar amino acid residues on the αC-helix, activation loop, and substrate-binding region. Our findings suggest preferable interactions of the potential tripeptide on enzyme inhibition that are useful for further development of a new class of inhibitors targeting EGFR-TK.


Subject(s)
ErbB Receptors/metabolism , Oligopeptides/chemistry , Protein Kinase Inhibitors/chemistry , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Docking Simulation , Oligopeptides/metabolism , Oligopeptides/pharmacology , Protein Domains , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Thermodynamics
19.
Clin Transl Med ; 12(5): e876, 2022 05.
Article in English | MEDLINE | ID: mdl-35605028

ABSTRACT

OBJECTIVE: Apurinic endonuclease 1 (APE1) has been suggested as an oncogene of lung tumours and our bioinformatics analysis identified the association between Erlotinib resistance and interleukin-6 (IL-6). Thus, we performed this work to delineate the mechanistic actions of APE1/IL-6 signalling in Erlotinib resistance of non-small cell lung cancer (NSCLC). METHODS: We selected human NSCLC cell lines HCC827 and PC9 to establish Erlotinib-resistant HCC827R and PC9R cells. Cancer stem cells (CSCs) were isolated from Erlotinib-sensitive HCC827P and PC9P cells (PCSCs) and from HCC827R and PC9R cells (RCSCs). Further, extracellular vesicles (EVs) were separated from PCSCs (PCSC-EVs) and RCSCs (RCSC-EVs) and co-cultured with RCSCs with or without short hairpin RNA (shRNA)-targeting APE1 (APE1 shRNA) transduction. In addition, functional assays were conducted to determine the effect of APE1 shRNA on malignant phenotypes of cancer cells in vitro and in vivo and the activation of IL-6/STAT3 signalling. RESULTS: It was found that NSCLC cells could internalize both RCSC-EVs and PCSC-EVs. RCSC-EVs augmented the resistance of NSCLC cells to Erlotinib. The overexpression of APE1 occurred in NSCLC tissues, and IL-6 was enriched in serum samples of patients with NSCLC. APE1 shRNA was demonstrated to restrict the Erlotinib resistance of NSCLC cells by inactivating the IL-6/STAT3 signalling. Additionally, shAPE1-loaded RCSC-EVs suppressed the Erlotinib resistance of NSCLC via the IL-6/STAT3 axis both in vitro and in vivo, as reflected by impeded malignant phenotypes and xenograft tumour formation. CONCLUSIONS: Collectively, these data indicate that APE1 confers Erlotinib resistance by activating the IL-6/STAT3 signalling, suggesting targeting APE1 as a possible therapeutic target in Erlotinib-resistant NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Extracellular Vesicles , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/therapeutic use , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Humans , Interleukin-6/metabolism , Interleukin-6/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/therapeutic use
20.
J Mater Chem B ; 8(8): 1728-1738, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32022097

ABSTRACT

Polymer microspheres are attracting wide attention in localized cancer therapy owing to the excellent biocompatibility and drug loading capacity, controllable biodegradation speeds, and minimized systemic toxicity. Herein, we presented poly(ester-thioether) microspheres, porous and nonporous, as drug depots for localized therapy of non-small cell lung cancer (NSCLC). Specifically, erlotinib and α-tocopheryl succinate (α-TOS), which are respectively an epidermal growth factor receptor (EGFR) inhibitor and mitochondria destabilizer, were efficiently loaded into porous and nonporous poly(ester-thioether) microspheres for the treatment of EGFR-overexpressing NSCLC (A549 cells). The poly(ester-thioether) microspheres significantly improved the bioavailability of both erlotinib and α-TOS in comparison to the free drug combination, realizing synergistic inhibition of A549 cells both in vitro and in vivo. The porous microspheres displayed faster degradation and drug release than the nonporous counterpart, thereby showing better anticancer efficacy. Overall, our study reported a new anticancer strategy of erlotinib and α-TOS combination for therapy of NSCLC, and established that poly(ester-thioether) microspheres could be a robust and biodegradable reservoir for drug delivery and localized cancer therapy.


Subject(s)
Erlotinib Hydrochloride/chemistry , Microspheres , Polymers/chemistry , Protein Kinase Inhibitors/chemistry , alpha-Tocopherol/chemistry , A549 Cells , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/drug effects , Drug Carriers/chemistry , Drug Therapy, Combination , Erlotinib Hydrochloride/metabolism , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Porosity , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism , alpha-Tocopherol/pharmacology , alpha-Tocopherol/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL