Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.002
Filter
1.
Respir Res ; 25(1): 233, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840238

ABSTRACT

BACKGROUND: There is inconclusive evidence to suggest that the expression of programmed cell death ligand 1 (PD-L1) is a putative predictor of response to EGFR-TKI therapy in advanced EGFR-mutant non-small cell lung cancer (NSCLC). We evaluated the heterogeneity in PD-L1 expression in the primary lung site and metastatic lymph nodes to analyze the association between PD-L1 expression and response for patients treated with EGFR-TKI. METHODS: This study reviewed 184 advanced NSCLC patients with EGFR mutations who received first-generation EGFR-TKI as first-line treatment from 2020 to 2021 at Shanghai Chest Hospital. The patients were divided into the primary lung site group (n = 100) and the metastatic lymph nodes group (n = 84) according to the biopsy site. The patients in each group were divided into TPS < 1%, TPS 1-49%, and TPS ≥ 50% groups according to PD-L1 expression. RESULTS: The median PFS was 7 (95% CI: 5.7-8.3) months, and the median OS was 26 (95% CI: 23.5-28.5) months for all patients. No correlation existed between PFS or OS and PD-L1 expression. The median PFS in the primary lung site group was 11 months (95% CI: 9.6-12.4) in the TPS < 1% group, 8 months (95% CI: 6.6-9.4) in TPS 1-49% group, and 4 months (95% CI: 3.2-4.8) in TPS ≥ 50% group, with statistically significant differences (p = 0.000). The median OS of the TPS < 1% group and TPS ≥ 50% group showed a statistically significant difference (p = 0.008) in the primary lung site group. In contrast, PD-L1 expression in the lymph nodes of EGFR-mutant patients was unrelated to PFS or OS after EGFR-TKI therapy. CONCLUSION: PD-L1 expression from the primary lung site might predict clinical benefit from EGFR-TKI, whereas PD-L1 from metastatic lymph nodes did not. TRIAL REGISTRATION: This retrospective study was approved by the Ethics Committee of Shanghai Chest Hospital (ID: IS23060) and performed following the Helsinki Declaration of 1964 (revised 2008).


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Lymphatic Metastasis , Protein Kinase Inhibitors , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Female , Male , Middle Aged , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Aged , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Lymph Nodes/pathology , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Adult , Aged, 80 and over , Treatment Outcome , Predictive Value of Tests , Mutation , Biomarkers, Tumor/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis
2.
Oncol Rep ; 48(2)2022 08.
Article in English | MEDLINE | ID: mdl-35775375

ABSTRACT

The clinical introduction of molecular imaging for the management of oropharyngeal squamous cell carcinoma (OPSCC) relies on the identification of relevant cancer­specific biomarkers. The application of three membrane­bound receptors, namely urokinase­type plasminogen activator receptor (uPAR), tissue factor (TF) and EGFR have been previously explored for targeted imaging and therapeutic strategies in a broad range of solid cancers. The present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and prognostic value in OPSCC. In a retrospective cohort of 93 patients with primary OPSCC, who were balanced into the 45 human papillomavirus (HPV)­positive and 48 HPV­negative groups, the IHC­determined expression profiles of uPAR, TF and EGFR in large biopsy or tumor resection specimens were analyzed. Using the follow­up data, overall survival (OS) and recurrence­free survival were measured. Specifically, associations between survival outcome, biomarker expression and clinicopathological factors were examined using Cox proportional hazards model and log­rank test following Kaplan­Meier statistics. After comparing the expression pattern of biomarkers within the tumor compartment with that in the adjacent normal tissues, uPAR and TF exhibited a highly tumor­specific expression pattern, whereas EGFR showed a homogeneous expression within the tumor compartment as well as a consistent expression in the normal mucosal epithelium and salivary gland tissues. The positive expression rate of uPAR, TF and EGFR in the tumors was 98.9, 76.3 and 98.9%, respectively. No statistically significant association between biomarker expression and survival outcome could be detected. Higher uPAR expression levels had a trend towards reduced OS according to results from univariate analysis (P=0.07; hazard ratio=2.01; 95% CI=0.92­4.37). Taken together, these results suggest that uPAR, TF and EGFR may be suitable targets for molecular imaging and therapy in OPSCC. In particular, uPAR may be an attractive target owing to their high positive expression rates in tumors and a highly tumor­specific expression pattern.


Subject(s)
Oropharyngeal Neoplasms , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Biomarkers, Tumor/biosynthesis , ErbB Receptors/biosynthesis , Humans , Molecular Imaging , Molecular Targeted Therapy , Oropharyngeal Neoplasms/diagnostic imaging , Oropharyngeal Neoplasms/drug therapy , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomaviridae , Papillomavirus Infections/diagnostic imaging , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Prognosis , Receptors, Urokinase Plasminogen Activator/biosynthesis , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Squamous Cell Carcinoma of Head and Neck/drug therapy , Thromboplastin/biosynthesis
3.
Molecules ; 27(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35208960

ABSTRACT

Melanoma is the most dangerous skin malignancy due to its strong metastatic potential with high mortality. Activation of crucial signaling pathways enforcing melanoma progression depends on phosphorylation of distinct tyrosine kinases and oxidative stress. We here investigated the effect of a bis-coumarin derivative [3, 3'- ((3″, 5'-Dichlorophenyl) methylene) bis (4-hydroxy-2H-chromen-2-one)] [3, 3'- (3, 5-DCPBC)] on human melanoma cell survival, growth, proliferation, migration, intracellular redox state, and deciphered associated signaling pathways. This derivative is toxic for melanoma cells and non-toxic for melanocytes, their benign counterpart, and fibroblasts. 3, 3'- (3, 5-DCPBC) inhibits cell survival, migration, and proliferation of different metastatic and non-metastatic melanoma cell lines through profound suppression of the phosphorylation of Epidermal Growth Factor receptor (EGFR) and proto-oncogene cellular sarcoma (c-SRC) related downstream pathways. Thus, 3, 3'- (3, 5-DCPBC) endowed with the unique property to simultaneously suppress phosphorylation of multiple downstream kinases, such as EGFR/JAK/STAT and EGFR/SRC and their corresponding transcription factors.


Subject(s)
Coumarins , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Melanoma , Neoplasm Proteins/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Signal Transduction/drug effects , Cell Line, Tumor , Coumarins/chemistry , Coumarins/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Humans , Melanoma/drug therapy , Melanoma/enzymology , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics
4.
Anal Biochem ; 643: 114544, 2022 04 15.
Article in English | MEDLINE | ID: mdl-34973938

ABSTRACT

This work developed an electrochemical impedance spectroscopy (EIS) sensor for detection of EGFR (epidermal growth factor receptor)-overexpressing tumor cell and preliminary estimation of EGFR expression. Here, EGFR antibodies as the specific antibodies for cancer cells were conjugated on magnetic gold-decorated graphene oxide nanocomposites, which were used to capture the EGFR-overexpressing tumor cells. The magnetically responsive tumor cells were enriched and immobilized on a magnetic glassy carbon electrode (mGCE) surface, leading to increased electron-transfer resistance (Ret) utilized for determination of cells and preliminary evaluation of EGFR expression level. This strategy enables the enrichment, fixation and detection of tumor cells to be accomplished in a facile way. An excellent linearity in the range of 2.0 × 102 - 3.0 × 105 cell mL-1 with the detection limit of 152 cell mL-1 for MDA-MB-231 cells was obtained. Investigation on the expression levels of EGFR on various types of cells was conducted. MDA-MB-231 cells showed a distinctly higher EGFR expression, compared with MHCC97-L and L02 cells, providing the possibility for the EGFR-targeted therapy of the tumors. It is expected that the proposed sensor has the potential to be applied for cancer monitoring.


Subject(s)
Biosensing Techniques , Electrochemical Techniques , Nanocomposites/chemistry , Carbon/chemistry , Electrodes , ErbB Receptors/biosynthesis , ErbB Receptors/metabolism , Gold/chemistry , Graphite/chemistry , Humans , Magnetic Phenomena , Tumor Cells, Cultured
5.
J Biochem Mol Toxicol ; 36(1): e22947, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34726804

ABSTRACT

Cervical cancer is a major human papillomavirus-related disease and is the fourth leading cause of death by cancer among women. Plants are an important source of anticancer compounds and many of them are currently used in the treatment of cancer. Several reports suggest the efficacy of plant-derived compounds increases when used in combination. This study was carried out to evaluate the effect of four plant-derived compounds such as curcumin (C), ellagic acid (E), quercetin (Q), and resveratrol (R) when used alone or in combinations using HeLa cervical cancer cells. All four phytocompounds showed effective cytotoxic activities in targeting HeLa cervical cancer cells as determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay. The selected phytocompound combinations C + E, C + Q, and Q + R work synergistically while the combination C + R shows additive effects. All four phytocompounds reduce cell migration as determined by in vitro wound-healing assay. The expression level of the epidermal growth factor receptor is significantly downregulated both in individual and combination. The flow cytometry analysis of cell cycle indicates that individual drugs curcumin, ellagic acid, quercetin, and resveratrol, each with 20 µM effectively arrested cell cycle at the S-phase while the combination of drugs (10 + 10 µM) at the G2/M phase.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Checkpoints/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Uterine Cervical Neoplasms/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Drug Screening Assays, Antitumor , ErbB Receptors/biosynthesis , Female , HeLa Cells , Humans , Uterine Cervical Neoplasms/drug therapy
6.
Oxid Med Cell Longev ; 2021: 9306760, 2021.
Article in English | MEDLINE | ID: mdl-34873433

ABSTRACT

Diabetic foot ulcers (DFUs) are caused by impairments in peripheral blood vessel angiogenesis and represent a great clinical challenge. Although various innovative techniques and drugs have been developed for treating DFUs, therapeutic outcomes remain unsatisfactory. Using the GEO database, we obtained transcriptomic microarray data for DFUs and control wounds and detected a significant downregulation of epidermal growth factor receptor (EGFR) in DFUs. We cultured human umbilical vein endothelial cells (HUVECs) and noted downregulated EGFR expression following high-glucose exposure in vitro. Further, we observed decreased HUVEC proliferation and migration and increased apoptosis after shRNA-mediated EGFR silencing in these cells. In mice, EGFR inhibition via focal EGFR-shRNA injection delayed wound healing. Target prediction analysis followed by dual-luciferase reporter assays indicated that microRNA-133b (miR-133b) is a putative upstream regulator of EGFR expression. Increased miR-133b expression was observed in both glucose-treated HUVECs and wounds from diabetes patients, but no such change was observed in controls. miR-133b suppression enhanced the proliferation and angiogenic potential of cultured HUVECs and also accelerated wound healing. Although angiogenesis is not the sole mechanism affected in DFU, these findings suggest that the miR-133b-induced downregulation of EGFR may contribute to delayed wound healing in diabetes. Hence, miR-133b inhibition may be a useful strategy for treating diabetic wounds.


Subject(s)
Diabetic Foot/metabolism , ErbB Receptors/biosynthesis , MicroRNAs/antagonists & inhibitors , Wound Healing/genetics , Animals , Case-Control Studies , Cell Proliferation , Diabetic Foot/genetics , Diabetic Foot/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Int J Mol Sci ; 22(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34948100

ABSTRACT

Neurofibromatosis type 1 (NF1) is an autosomal dominant human genetic disorder. The progression of benign plexiform neurofibromas to malignant peripheral nerve sheet tumors (MPNSTs) is a major cause of mortality in patients with NF1. Although elevated epidermal growth factor receptor (EGFR) expression plays a crucial role in the pathogenesis of MPNST, the cause of EGFR overexpression remains unclear. Here, we assessed EGFR expression levels in MPNST tissues of NF1 patients and NF1 patient-derived MPNST cells. We found that the expression of EGFR was upregulated in MPNST tissues and MPNST cells, while the expression of neurofibromin was significantly decreased. Manipulation of NF1 expression by NF1 siRNA treatment or NF1-GAP-related domain overexpression demonstrated that EGFR expression levels were closely and inversely correlated with neurofibromin levels. Notably, knockdown of the NF1 gene by siRNA treatment augmented the nuclear localization of phosphorylated SP1 (pSP1) and enhanced pSP1 binding to the EGFR gene promoter region. Our results suggest that neurofibromin deficiency in NF1-associated MPNSTs enhances the Ras/ERK/SP1 signaling pathway, which in turn may lead to the upregulation of EGFR expression. This study provides insight into the progression of benign tumors and novel therapeutic approaches for treatment of NF1-associated MPNSTs.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Neurofibromatosis 1/metabolism , Neurofibromin 1/metabolism , Sp1 Transcription Factor/metabolism , Up-Regulation , ras Proteins/metabolism , Cell Line, Tumor , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Sp1 Transcription Factor/genetics , ras Proteins/genetics
8.
Molecules ; 26(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34299635

ABSTRACT

Lung cancer is one of the most commonly occurring cancer mortality worldwide. The epidermal growth factor receptor (EGFR) plays an important role in cellular functions and has become the new promising target. Natural products and their derivatives with various structures, unique biological activities, and specific selectivity have served as lead compounds for EGFR. D-glucose and EGCG were used as starting materials. A series of glucoside derivatives of EGCG (7-12) were synthesized and evaluated for their in vitro anticancer activity against five human cancer cell lines, including HL-60, SMMC-7721, A-549, MCF-7, and SW480. In addition, we investigated the structure-activity relationship and physicochemical property-activity relationship of EGCG derivatives. Compounds 11 and 12 showed better growth inhibition than others in four cancer cell lines (HL-60, SMMC-7721, A-549, and MCF), with IC50 values in the range of 22.90-37.87 µM. Compounds 11 and 12 decreased phosphorylation of EGFR and downstream signaling protein, which also have more hydrophobic interactions than EGCG by docking study. The most active compounds 11 and 12, both having perbutyrylated glucose residue, we found that perbutyrylation of the glucose residue leads to increased cytotoxic activity and suggested that their potential as anticancer agents for further development.


Subject(s)
Antineoplastic Agents , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Cytotoxins , Glucose , Molecular Docking Simulation , Neoplasm Proteins , Neoplasms , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catechin/chemical synthesis , Catechin/chemistry , Catechin/pharmacology , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/chemistry , Glucose/analogs & derivatives , Glucose/chemical synthesis , Glucose/chemistry , Glucose/pharmacology , HL-60 Cells , Humans , MCF-7 Cells , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/chemistry , Neoplasms/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation/drug effects
9.
Int J Biol Sci ; 17(10): 2590-2605, 2021.
Article in English | MEDLINE | ID: mdl-34326696

ABSTRACT

Pancreatic adenosquamous carcinoma (PASC) - a rare pathological pancreatic cancer (PC) type - has a poor prognosis due to high malignancy. To examine the heterogeneity of PASC, we performed single-cell RNA sequencing (scRNA-seq) profiling with sample tissues from a healthy donor pancreas, an intraductal papillary mucinous neoplasm, and a patient with PASC. Of 9,887 individual cells, ten cell subpopulations were identified, including myeloid, immune, ductal, fibroblast, acinar, stellate, endothelial, and cancer cells. Cancer cells were divided into five clusters. Notably, cluster 1 exhibited stem-like phenotypes expressing UBE2C, ASPM, and TOP2A. We found that S100A2 is a potential biomarker for cancer cells. LGALS1, NPM1, RACK1, and PERP were upregulated from ductal to cancer cells. Furthermore, the copy number variations in ductal and cancer cells were greater than in the reference cells. The expression of EREG, FCGR2A, CCL4L2, and CTSC increased in myeloid cells from the normal pancreas to PASC. The gene sets expressed by cancer-associated fibroblasts were enriched in the immunosuppressive pathways. We demonstrate that EGFR-associated ligand-receptor pairs are activated in ductal-stromal cell communications. Hence, this study revealed the heterogeneous variations of ductal and stromal cells, defined cancer-associated signaling pathways, and deciphered intercellular interactions following PASC progression.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Adenosquamous/genetics , Pancreatic Neoplasms/genetics , Transcriptome/genetics , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Biomarkers, Tumor/genetics , Carcinoma, Adenosquamous/metabolism , Carcinoma, Adenosquamous/pathology , Chemotactic Factors/genetics , DNA Copy Number Variations , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Genetic Heterogeneity , Humans , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , S100 Proteins/genetics , Sequence Analysis, RNA , Single-Cell Analysis
10.
Biochem Pharmacol ; 192: 114702, 2021 10.
Article in English | MEDLINE | ID: mdl-34324869

ABSTRACT

Platinum complexes are currently used for breast cancer therapy, but, as with other drug classes, a series of intrinsic and acquired resistance mechanisms hinder their efficacy. To better understand the mechanisms underlying platinum complexes resistance in breast cancer, we generated a [Pt(O,O'-acac)(γ-acac)(DMS)]-resistant MCF-7, denoted as [Pt(acac)2]R. [Pt(O,O'-acac)(γ-acac)(DMS)] was chosen as previous works showed that it has distinct mechanisms of action from cisplatin, especially with regard to cellular targets. [Pt(acac)2]R cells are characterized by increased proliferation rates and aggressiveness with higher PKC-δ, BCL-2, MMP-9 and EGFR protein expressions and also by increased expression of various genes covering cell cycle regulation, invasion, survival, and hormone receptors. These [Pt(acac)2]R cells also displayed high levels of activated signaling kinases Src, AKT and ERK/2. [Pt(acac)2]R cells incubated with [Pt(O,O'-acac)(γ-acac)(DMS)], showed a relevant EGFR activation due to PKC-δ and Src phosphorylation that provoked proliferation and survival through MERK1/2/ERK1/2 and PI3K/Akt pathways. In addition, EGFR shuttled from the plasma membrane to the nucleus maybe acting as co-transcriptional factor. The data suggest that growth and survival of resistant cells rely upon a remarkable increase in EGFR level which, in collaboration with an enhanced role of PKC-δ and Src kinases support [Pt(acac)2]R cell. It could therefore be assumed that combination treatments targeting both EGFR and PKC-δ/Src can improve therapy for breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Platinum Compounds/pharmacology , Signal Transduction/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Humans , MCF-7 Cells , Platinum Compounds/therapeutic use , Signal Transduction/drug effects
11.
Medicine (Baltimore) ; 100(14): e25344, 2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33832110

ABSTRACT

ABSTRACT: Excision repair cross complementing 1 (ERCC1), ribonucleotide reductase M1 (RRM1), ß-tubulin III (TUBB3), thymidylate synthetase (TYMS), and topoisomerase IIα (TOP2A) genes have been shown to be associated with the pathogenesis and prognosis of various types of carcinomas; however, their roles in breast cancer have not been fully validated. In this study, we evaluated the correlations among these biomarkers and the associations between their expression intensity and the clinicopathological characteristics to investigate whether the above genes are underlying biomarkers for patients with breast cancer.Ninety-seven tissue specimens collected from breast cancer patients. The expression levels of these biomarkers were measured by the multiplex branched DNA liquidchip (MBL) technology and clinicopathological characteristics were collected simultaneously.The expression levels of ERCC1, TUBB3, TYMS, and TOP2A were significantly associated with the characteristics of menopausal status, tumor size, lymph node metastasis, hormone receptor status, triple-negative status, Ki-67 index, and epidermal growth factor receptor. The expression intensity of ERCC1 negatively associated with that of TUBB3 and TYMS, and positively associated with that of RRM1. The expression intensity of TOP2A positively associated with that of TYMS. Hierarchical clustering analysis and difference test indicated that breast cancer with higher levels of TUBB3, TYMS, and TOP2A, as well as lower levels of ERCC1 and RRM1 tended to have higher histological grade and Ki-67 index.Our studies showed that ERCC1, TYMS, TUBB3, and TOP2A may be potential biomarkers for prognosis and individualized chemotherapy guidance, while there may be interactions between ERCC1 and RRM1, or TUBB3, or TYMS, as well as between TOP2A and TYMS in pathogenesis and development of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , ErbB Receptors/biosynthesis , Female , Gene Expression , Humans , Ki-67 Antigen/biosynthesis , Lymphatic Metastasis/pathology , Menopause/physiology , Middle Aged , Ribonucleotide Reductases/genetics , Thymidylate Synthase/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Burden
12.
Biochem Pharmacol ; 188: 114516, 2021 06.
Article in English | MEDLINE | ID: mdl-33713643

ABSTRACT

The overexpression of the human ATP-binding cassette (ABC) drug transporter ABCB1 (P-glycoprotein, P-gp) or ABCG2 (breast cancer resistance protein, BCRP) in cancer cells often contributes significantly to the development of multidrug resistance (MDR) in cancer patients. Previous reports have demonstrated that some epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) could modulate the activity of ABCB1 and/or ABCG2 in human cancer cells, whereas some EGFR TKIs are transport substrates of these transporters. Almonertinib (HS-10296) is a promising, orally available third-generation EGFR TKI for the treatment of EGFR T790M mutation-positive non-small cell lung cancer (NSCLC) in patients who have progressed on or after other EGFR TKI therapies. Additional clinical trials are currently in progress to study almonertinib as monotherapy and in combination with other agents in patients with NSCLC. In the present work, we found that neither ABCB1 nor ABCG2 confers significant resistance to almonertinib. More importantly, we discovered that almonertinib was able to reverse MDR mediated by ABCB1, but not ABCG2, in multidrug-resistant cancer cells at submicromolar concentrations by inhibiting the drug transport activity of ABCB1 without affecting its expression level. These findings are further supported by in silico docking of almonertinib in the drug-binding pocket of ABCB1. In summary, our study revealed an additional activity of almonertinib to re-sensitize ABCB1-overexpressing multidrug-resistant cancer cells to conventional chemotherapeutic drugs, which may be beneficial for cancer patients and warrant further investigation.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Pyrimidines/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/physiology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Protein Structure, Secondary
13.
Appl Immunohistochem Mol Morphol ; 29(8): 576-584, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33758141

ABSTRACT

The membrane EGFR (mEGFR) protein overexpression in the head and neck squamous cell carcinoma (SCC) is considered to cause increased EGFR activity which adds to tumorigenicity and therapy resistance. The mEGFR upon stimulation can translocate to the nucleus nuclear EGFR (nEGFR) where it has been associated with poor prognosis and worse survival in many cancers. The relevance of differentially located EGFR proteins in laryngeal lesions has not been studied enough and remains unclear. Aim of our study was to examine nEGFR and mEGFR protein expression as well as EGFR gene status and cell cycle proliferation markers in the laryngeal polyps, dysplasia, and SCC using immunohistochemistry and in situ hybridization. There was significantly higher frequency of strong nEGFR between SCC, dysplasia, and polyps (P<0.0001), and strong mEGFR in the SCC and laryngeal dysplasia comparing to polyps (P<0.0001). Gene amplification was confirmed only in relatively small number of SCC but not in non-neoplastic lesions. In dysplasia the statistically significant positive correlations between nEGFR, and Ki-67 (P=0.029), p53 (P=0.001), and cyclin D1 (P=0.031) were found. nEGFR and mEGFR expression showed statistically significant inverse correlation in the SCC (P=0.004) as well as nEGFR and cyclin D1 (P=0.032). Univariate statistical analysis showed statistically significant correlation between strong nEGFR protein expression and worse overall survival in laryngeal SCC, alone or in coexpression with strong cyclin D1 and high Ki-67 (P=0.025, P=0.046, P=0.043, respectively). Our data show that nEGFR cellular localization might influence biology of the laryngeal carcinogenesis and is indicator of poor survival.


Subject(s)
Cell Nucleus , Gene Expression Regulation, Neoplastic , Laryngeal Neoplasms , Aged , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cyclin D1/biosynthesis , Disease-Free Survival , ErbB Receptors/biosynthesis , Female , Humans , Immunohistochemistry , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/mortality , Laryngeal Neoplasms/pathology , Male , Middle Aged , Survival Rate
14.
Biomolecules ; 11(2)2021 01 26.
Article in English | MEDLINE | ID: mdl-33530424

ABSTRACT

The anticancer activities of Withaferin-A (Wi-A) and Withanone (Wi-N) from Ashwagandha and Caffeic Acid Phenethyl Ester (CAPE) from honeybee propolis have been well documented. Here, we examined the binding potential of these natural compounds to inhibit the constitutive phosphorylation of epidermal growth factor receptors (EGFRs). Exon 20 insertion mutants of EGFR, which show resistance to various FDA approved drugs and are linked to poor prognosis of lung cancer patients, were the primary focus of this study. Apart from exon 20 insertion mutants, the potential of natural compounds to serve as ATP competitive inhibitors of wildtype protein and other common mutants of EGFR, namely L858R and exon19del, were also examined. The potential of natural compounds was compared to the positive controls such as erlotinib, TAS6417 and poziotinib. Similar to known inhibitors, Wi-A and Wi-N could displace and binds at the ATP orthosteric site of exon19del, L858R and exon20, while CAPE was limited to wildtype EGFR and exon 20 insertion mutants only. Moreover, the binding free energy of the natural drugs against EGFRs was also comparable to the positive controls. This computational study suggests that Wi-A and Wi-N have potential against multiple mutated EGFRs, warranting further in vitro and in vivo experiments.


Subject(s)
Antineoplastic Agents/pharmacology , Caffeic Acids/pharmacology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Phenylethyl Alcohol/analogs & derivatives , Withanolides/pharmacology , Adenosine Triphosphate/chemistry , Computational Biology/methods , Computer Simulation , Dimerization , ErbB Receptors/biosynthesis , Erlotinib Hydrochloride/pharmacology , Exons , Gene Deletion , Humans , Indolizines/pharmacology , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Phenylethyl Alcohol/pharmacology , Quinazolines/pharmacology
15.
Mol Cell Biochem ; 476(4): 1673-1690, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33420898

ABSTRACT

Accumulating evidence suggests that specific non-coding RNAs exist in many types of malignant tissues, and are involved in cancer invasion and metastasis. However, little is known about the precise roles of non-coding RNAs in squamous cell carcinoma (SQCC) invasion and migration. Recently, the dentin matrix protein-1 (DMP-1) gene locus was identified as a transcriptionally active site in squamous cell carcinoma (SQCC) tissue and cells. However, it is unclear whether RNA associated with cell migration exist at the DMP-1 gene locus in SQCC cells. We identified a novel promoter-associated non-coding RNA in the antisense strand of DMP-1 gene locus, promoter-associated non-coding RNA (panRNA)-DMP-1, by the RACE method in SQCC cells and tissues, and characterized the functions of panRNA-DMP-1 in EGF-driven SQCC cell migration. The inhibition of endogenous panRNA-DMP-1 expression by specific siRNAs and exogenous over-expression of panRNA-DMP-1 resulted in increased and suppressed cellular migration toward EGF in SQCC cells, respectively, and nuclear expression of panRNA-DMP-1 was induced by EGF stimulation. Mechanistically, suppression of panRNA-DMP-1 expression increased EGFR nuclear localization upon EGF treatment and nuclear panRNA-DMP-1 physically interacted with EGFR, which was confirmed by RNA immunoprecipitation assay using a bacteriophage-delivered PP7 RNA labeling system. Furthermore, co-immunoprecipitation assay revealed that suppression of panRNA-DMP-1 stabilized EGFR interaction with STAT3, a known co-transcription factors of EGFR, to induce migratory properties in many cancer cells. Based on these findings, panRNA-DMP-1 is an EGFR-associating RNA that inhibits the EGF-induced migratory properties of SQCC possibly by regulating EGFR nuclear localization and EGFR binding to STAT3.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement , Epidermal Growth Factor/metabolism , Extracellular Matrix Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , RNA, Antisense/metabolism , RNA, Neoplasm/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epidermal Growth Factor/genetics , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/genetics , Phosphoproteins/genetics , RNA, Antisense/genetics , RNA, Neoplasm/genetics
16.
Invest New Drugs ; 39(4): 1011-1018, 2021 08.
Article in English | MEDLINE | ID: mdl-33506323

ABSTRACT

Background Alflutinib is a novel irreversible and highly selective third-generation EGFR inhibitor currently being developed for the treatment of non-small cell lung cancer patients with activating EGFR mutations and EGFR T790M drug-resistant mutation. Alflutinib is mainly metabolized via CYP3A4 to form its active metabolite AST5902. Both alflutinib and AST5902 contribute to the in vivo pharmacological activity. The aim of this study was to investigate the effects of rifampicin (a strong CYP3A4 inducer) on the pharmacokinetics of alflutinib and AST5902 in healthy volunteers, thus providing important information for drug-drug interaction evaluation and guiding clinical usage. Methods This study was designed as a single-center, open-label, and single-sequence trial over two periods. The volunteers received a single dose of 80 mg alflutinib on Day 1/22 and continuous doses of 0.6 g rifampicin on Day 15-30. Blood sampling was conducted on Day 1-10 and Day 22-31. The pharmacokinetics of alflutinib, AST5902, and the total active ingredients (alflutinib and AST5902) with or without rifampicin co-administration were respectively analyzed. Results Co-administration with rifampicin led to 86% and 60% decreases in alflutinib AUC0-∞ and Cmax, respectively, as well as 17% decrease in AST5902 AUC0-∞ and 1.09-fold increase in AST5902 Cmax. The total active ingredients (alflutinib and AST5902) exhibited 62% and 39% decreases in AUC0-∞ and Cmax, respectively. Conclusions As a strong CYP3A4 inducer, rifampicin exerted significant effects on the pharmacokinetics of alflutinib and the total active ingredients (alflutinib and AST5902). The results suggested that concomitant strong CYP3A4 inducers should be avoided during alflutinib treatment. This trial was registered at http://www.chinadrugtrials.org.cn . The registration No. is CTR20191562, and the date of registration is 2019-09-12.


Subject(s)
Cytochrome P-450 CYP3A Inducers/pharmacology , Indoles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/pharmacokinetics , Pyrimidines/pharmacokinetics , Rifampin/pharmacology , Adolescent , Adult , Area Under Curve , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , ErbB Receptors/biosynthesis , Female , Humans , Male , Middle Aged , Young Adult
17.
Head Neck Pathol ; 15(3): 743-756, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33428063

ABSTRACT

The epidermal growth factor receptor (EGFR) pathway is important in tumorigenesis of oropharyngeal carcinoma (OPC). However, the molecular mechanisms contributing to EGFR expression in OPC are not well-known. To detect relating factors and clinicopathological impact of EGFR protein expression in OPC, gene amplification/loss, point mutations including synonymous mutations, and promoter methylation of EGFR, and the viral genome load of human papillomavirus type 16 (HPV16)-E5, -E6, and -E7, after extracting HPV16-related OPCs with qPCR of HPV16-E6 and E7, were investigated in 74 OPC surgical cases, including 52 HPV-related (HPV-OPC) and 22 HPV-unrelated (nHPV-OPC). Immunohistochemical (IHC) data of EGFR expression (high, weak, and negative), validated by the qPCR of EGFR mRNA, were compared with molecular, viral, and clinicopathological data of patients. All nHPV-OPC cases were EGFR-IHC-high, whereas 21.2%, 65.4%, and 13.5% of HPV-OPC cases showed EGFR-IHC-high, -weak, -negative (p < 0.01), respectively. In HPV-OPC cases, EGFR-IHC-weak/negative status was related to promoter methylation of EGFR (p = 0.009), but not with gene amplification/loss or the point mutation of EGFR and was more often seen in HPV16-OPC cases (p = 0.049). Among HPV16-OPC cases, EGFR-IHC-weak/negative was related to high E6 expression. EGFR protein-loss was related to the tumor histology of non-keratinizing squamous cell carcinoma (SCC) (p = 0.035) but not with patient prognosis. In conclusion, decreased EGFR protein expression was more frequent in HPV-OPC than in nHPV-OPC and was related to EGFR methylation, infection of HPV16, and the viral genome load of HPV16-E6. Clinicopathologically, it was related to the tumor histology of non-keratinizing SCC.


Subject(s)
Head and Neck Neoplasms/pathology , Papillomavirus Infections/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Adult , Aged , Aged, 80 and over , DNA Methylation , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Humans , Male , Middle Aged , Oncogene Proteins, Viral , Papillomavirus Infections/virology , Repressor Proteins , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology , Viral Load
18.
Biotechnol Bioeng ; 118(4): 1466-1475, 2021 04.
Article in English | MEDLINE | ID: mdl-33331661

ABSTRACT

Epidermal growth factor receptor (EGFR) is a clinically validated target for a multitude of human cancers. The receptor is activated upon ligand binding through a critical dimerization step. Dimerization can be replicated in vitro by locally concentrating the receptor kinase domains on the surface of lipid-based vesicles. In this study we investigated the use of coiled coils to induce spontaneous receptor kinase domain dimerization in vitro to form non-membrane-bound artificial receptor mimics in solution. Two engineered forms of EGFR kinase domain fused to coiled coil complementary peptides were designed to self-associate upon mixing. Two fusion protein species (P3-EGFR and P4-EGFR) independently showed the same activity and polymerization profile known to exist with EGFR kinase domains. Upon mixing the two species, coiled coil heterodimers were formed that induced EGFR association to form dimers of the kinase domains. This was accompanied by 11.5-fold increase in the phosphorylation rate indicative of kinase domain activation equivalent to the levels achieved using vesicle localization and mimicking in vivo ligand-induced activation. This study presents a soluble tyrosine kinase receptor mimic capable of spontaneous in vitro activation that can facilitate functional and drug discovery studies for this clinically important receptor class.


Subject(s)
Dimerization , ErbB Receptors , Protein Engineering , Animals , ErbB Receptors/biosynthesis , ErbB Receptors/chemistry , ErbB Receptors/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sf9 Cells , Spodoptera
19.
J Ethnopharmacol ; 269: 113745, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33359859

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ephedrae Herba (EH, Ephedra sinica Stapf.) and Armeniacae Semen Amarum (ASA, Prunus armeniaca L. var. ansu Maxim.) have been used to treat asthma, cold, fever, and cough in China for thousands of years. AIM OF THE STUDY: In this study, we aimed to investigate the optimal ratio of EH and ASA compatibility (EAC) to reduce airway injury in asthmatic rats and its possible mechanism. METHODS: Rats were sensitized with a mixture of acetylcholine chloride and histamine bisphosphate 1 h before sensitization by intragastric administration of EAC or dexamethasone or saline for 7 days. Subsequently, the ultrastructure of rat airway epithelial tissue changes, apoptosis of the airway epithelial cells, and the expression of mRNA and protein of EGRF and Bcl-2 were detected. RESULTS: Transmission electron microscope: EAC (groups C and E) had the most prominent effect on repairing airway epithelial cells' ultrastructural changes in asthmatic rats. TUNEL: dexamethasone and EAC (groups B、C、E and F) inhibited the apoptosis of airway epithelial cells in asthmatic rats (P < 0.05). In situ hybridization: EAC (group E) inhibited the overexpression of EGFR and Bcl-2 mRNA (P < 0.05).Western Blotting: EAC (groups A、B、C、E and F) inhibited the upregulation of airway epithelial EGFR and Bcl-2 protein expression (P < 0.01). CONCLUSIONS: Our findings indicate that EAC can inhibit abnormal changes in airway epithelial structure and apoptosis of airway epithelial cells, thereby alleviating airway injury. In this study, the best combination of EH and ASA to alleviate airway epithelial injury in asthmatic rats was group E (EH: ASA = 8: 4.5).


Subject(s)
Asthma/drug therapy , Drugs, Chinese Herbal/pharmacology , Ephedra sinica/chemistry , Prunus armeniaca/chemistry , Respiratory System/drug effects , Acetylcholine/toxicity , Animals , Apoptosis/drug effects , Asthma/chemically induced , Disease Models, Animal , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/therapeutic use , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Histamine/analogs & derivatives , Histamine/toxicity , Male , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats, Sprague-Dawley , Respiratory System/injuries , Respiratory System/pathology , Respiratory System/ultrastructure , Trachea/drug effects , Trachea/injuries , Trachea/pathology , Trachea/ultrastructure
20.
Medicine (Baltimore) ; 99(49): e23321, 2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33285708

ABSTRACT

The relationship between splenic volume and the outcome of chemoradiotherapy for lung cancer has rarely been studied or addressed. The purpose of our study was to investigate whether splenic volume was associated with prognosis in patients treated with chemoradiotherapy for advanced or locally advanced non-small cell lung cancer (NSCLC).A retrospective investigation was conducted. Finally, 202 patients met the criteria and were included in the study. All patients were divided into 2 groups according to the optimum cutoff value of splenic volume for overall survival (OS). The optimum cutoff value was identified by X-tile software, and the OS and disease-free survival (DFS) were compared between the 2 groups of patients. The impact of splenic volume and other clinical characteristics on OS and DFS was analyzed using the Kaplan-Meier method and Cox proportional hazards model. Clinical characteristics were compared using chi-square or Fisher exact tests.The median (range) of splenic volume was 156.03 (28.55-828.11) cm. The optimal cutoff value of splenic volume was 288.4 cm. For univariate analyses, high splenic volume was associated with decreased OS (P = .025) and DFS (P = .044). In multivariate analyses, splenic volume remained an independent predictor of OS as a binary dependent variable (P = .003).Excessive splenic volume was associated with decreased OS and DFS in patients with NSCLC treated with chemoradiotherapy. Splenic volume should be regarded as an independent prognostic factor for patients treated with chemoradiotherapy for advanced or locally advanced NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/therapy , Chemoradiotherapy/methods , Lung Neoplasms/mortality , Lung Neoplasms/therapy , Spleen/pathology , Age Factors , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Comorbidity , ErbB Receptors/biosynthesis , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Retrospective Studies , Sex Factors , Spleen/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL