Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.489
Filter
1.
Cardiorenal Med ; 14(1): 334-349, 2024.
Article in English | MEDLINE | ID: mdl-38801815

ABSTRACT

INTRODUCTION: Large prospective trials have demonstrated that finerenone could reduce the risk of cardiovascular death and progression of renal failure among patients with chronic kidney disease associated heart failure and/or type 2 diabetes mellitus (T2DM). The aim of this study was to explore the molecular mechanism of finerenone in the treatment of cardiorenal diseases through network pharmacology. METHODS: The STITH, SwissTargetPrediction, PharmMapper, DrugBank, and ChEMBL databases were used to screen the targets of finerenone. The disease-related targets were retrieved from the DisGeNET, GeneCards, CTD, OMIM, and MalaCards databases. The protein-protein interaction (PPI) network was conducted with STRING database and Cytoscape software. The clusterProfiler R package was used to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The interactions of key targets and finerenone were analyzed by molecular docking in Autodock software. Diabetes mellitus was induced by intraperitoneal injection of streptozotocin. Histopathology of myocardial and renal tissues was observed by hematoxylin-eosin (HE) staining, and detection of protein expressions was conducted using Western blotting. RESULTS: A total of 111 potential cardiorenal targets of finerenone were identified. The main mechanisms of action may be associated with lipids and atherosclerosis, fluid shear stress and atherosclerosis, AGE-RAGE signaling pathway in diabetic complications, and diabetic cardiomyopathy. The hub targets demonstrated by the PPI network were CASP3, ALB, MMP9, EGFR, ANXA5, IGF1, SRC, TNFRSF1A, IL2, and PPARG, and the docking results suggested that finerenone could bind to these targets with high affinities. HE staining revealed the cardiorenal protection of finerenone on diabetic mice. In addition, the protein expressions of CASP3 and EGFR were increased while ALB was decreased in myocardial and renal tissues in diabetic mice compared with control mice, which were reversed by finerenone. CONCLUSION: This study suggested that finerenone exerts cardiorenal benefits through multiple targets and pathways.


Subject(s)
Diabetes Mellitus, Experimental , Molecular Docking Simulation , Naphthyridines , Network Pharmacology , Naphthyridines/pharmacology , Animals , Mice , Diabetes Mellitus, Experimental/complications , Protein Interaction Maps , Male , ErbB Receptors/metabolism , ErbB Receptors/drug effects , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Heart Failure/drug therapy
2.
Acta Cir Bras ; 39: e391624, 2024.
Article in English | MEDLINE | ID: mdl-38808816

ABSTRACT

PURPOSE: To evaluate the chemotherapeutic activity of temozolomide counter to mammary carcinoma. METHODS: In-vitro anticancer activity has been conducted on MCF7 cells, and mammary carcinoma has been induced in Wistar rats by introduction of 7, 12-Dimethylbenz(a)anthracene (DMBA), which was sustained for 24 weeks. Histopathology, immunohistochemistry, cell proliferation study and apoptosis assay via TUNEL method was conducted to evaluate an antineoplastic activity of temozolomide in rat breast tissue. RESULTS: IC50 value of temozolomide in MCF7 cell has been obtained as 103 µM, which demonstrated an initiation of apoptosis. The temozolomide treatment facilitated cell cycle arrest in G2/M and S phase dose dependently. The treatment with temozolomide suggested decrease of the hyperplastic abrasions and renovation of the typical histological features of mammary tissue. Moreover, temozolomide therapy caused the downregulation of epidermal growth factor receptor, extracellular signal-regulated kinase, and metalloproteinase-1 expression and upstream of p53 and caspase-3 proliferation to indicate an initiation of apoptotic events. CONCLUSIONS: The occurrence of mammary carcinoma has been significantly decreased by activation of apoptotic pathway and abrogation of cellular propagation that allowable for developing a suitable mechanistic pathway of temozolomide in order to facilitate chemotherapeutic approach.


Subject(s)
Antineoplastic Agents, Alkylating , Apoptosis , ErbB Receptors , Rats, Wistar , Temozolomide , Temozolomide/pharmacology , Temozolomide/therapeutic use , Animals , Apoptosis/drug effects , Female , ErbB Receptors/drug effects , ErbB Receptors/antagonists & inhibitors , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Humans , MCF-7 Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Immunohistochemistry , Reproducibility of Results , Rats , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology
3.
Bioorg Med Chem Lett ; 98: 129575, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38065292

ABSTRACT

The C797S mutation is one of the major factors behind resistance to the third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors. Herein, we describe the discovery of the 2,4-diaminonicotinamide derivative 5j, which shows potent inhibitory activity against EGFR del19/T790M/C797S and L858R/T790M/C797S. We also report the structure-activity relationship of the 2,4-diaminonicotinamide derivatives and the co-crystal structure of 5j and EGFR del19/T790M/C797S.


Subject(s)
ErbB Receptors , Lung Neoplasms , Niacinamide , Humans , Drug Resistance, Neoplasm , ErbB Receptors/drug effects , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , /pharmacology , Niacinamide/analogs & derivatives , Niacinamide/chemistry
4.
Cells ; 12(6)2023 03 16.
Article in English | MEDLINE | ID: mdl-36980255

ABSTRACT

Background: ZR2002 is a dual EGFR-DNA-targeting combi-molecule that carries a chloroethyl group at the six-position of the quinazoline ring designed to alkylate DNA. Despite its good pharmacokinetics, ZR2002 is metabolized in vivo into dechlorinated metabolites, losing the DNA-alkylating function required to damage DNA. To increase the DNA damage activity in tumor cells in vivo, we compared ZR2002 with two of its 6-N,N-disubstituted analogs: "JS61", with a nitrogen mustard function at the six-position of the quinazoline ring, and "JS84", with an N-methyl group. Methods: Tumor xenografts were performed with the human Saos-2 osteosarcoma cell line expressing EGFR. Mice were treated with ZR2002, JS84 or JS61, and the tumor burden was measured with a caliper and CT/PET imaging. Drug metabolism was analyzed with LC-MS. EGFR and ɣ-H2AX phosphorylation were quantified via Western blot analysis and immunohistochemistry. Results: In vivo analysis showed that significant tumor growth inhibition was only achieved when ZR2002 was administered in its naked form. The metabolic dealkylation of JS61 and JS84 did not release sufficient concentrations of ZR2002 for the intratumoral inhibition of P-EGFR or enhanced levels of P-H2AX. Conclusions: The results in toto suggest that intratumoral concentrations of intact ZR2002 are correlated with the highest inhibition of P-EGFR and induction of DNA damage in vivo. ZR2002 may well represent a good drug candidate for the treatment of EGFR-expressing osteosarcoma.


Subject(s)
ErbB Receptors , Osteosarcoma , Quinazolines , Animals , Humans , Mice , DNA/chemistry , ErbB Receptors/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Heterografts , Osteosarcoma/drug therapy , Prodrugs , Quinazolines/pharmacology , Quinazolines/therapeutic use
5.
Bioorg Med Chem Lett ; 79: 129066, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36410591

ABSTRACT

Treatment of advanced stage epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) is often complicated by the occurrence of acquired resistance, which emphasizes the need for improved treatment options. Based on a previously reported structure-activity relationship (SAR) study of Spautin-1, which resulted in the discovery of 10a, the search for more potent analogues was envisaged through optimization of the amine substituent. Our search led to the discovery of analogue 15b, harbouring the 2-[4-(4-fluoro-phenoxy)-phenyl]ethylamine substituent, among other potent and original analogues, with nanomolar activity towards EGFR-mutant NSCLC cells. Moreover, this compound 15b showed good selectivity for cancer cells over healthy lung epithelial cells and provides additive effects with food and drug administration (FDA) approved EGFR-tyrosine kinase inhibitors (TKIs), as proven by the co-administration of 15b with Afatinib. Altogether, we report promising lead compounds which show the potential to improve current treatment options.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/drug effects , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Quinazolines/chemistry , Quinazolines/pharmacology
6.
Acta Pharmacol Sin ; 44(5): 1083-1094, 2023 May.
Article in English | MEDLINE | ID: mdl-36411339

ABSTRACT

Although several KRasG12C inhibitors have displayed promising efficacy in clinical settings, acquired resistance developed rapidly and circumvented the activity of KRasG12C inhibitors. To explore the mechanism rendering acquired resistance to KRasG12C inhibitors, we established a series of KRASG12C-mutant cells with acquired resistance to AMG510. We found that differential activation of receptor tyrosine kinases (RTKs) especially EGFR or IGF1R rendered resistance to AMG510 in different cellular contexts by maintaining the activation of MAPK and PI3K signaling. Simultaneous inhibition of EGFR and IGF1R restored sensitivity to AMG510 in resistant cells. PI3K integrates signals from multiple RTKs and the level of phosphorylated AKT was revealed to negatively correlate with the anti-proliferative activity of AMG510 in KRASG12C-mutant cells. Concurrently treatment of a novel PI3Kα inhibitor CYH33 with AMG510 exhibited a synergistic effect against parental and resistant KRASG12C-mutant cells in vitro and in vivo, which was accompanied with concomitant inhibition of AKT and MAPK signaling. Taken together, these findings revealed the potential mechanism rendering acquired resistance to KRasG12C inhibitors and provided a mechanistic rationale to combine PI3Kα inhibitors with KRasG12C inhibitors for therapy of KRASG12C-mutant cancers in future clinical trials.


Subject(s)
Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins p21(ras) , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Mutation , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Immune Checkpoint Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics
7.
PLoS One ; 17(10): e0276497, 2022.
Article in English | MEDLINE | ID: mdl-36269747

ABSTRACT

Skin rash is a well-known predictive marker of the response to cetuximab (Cmab) in metastatic colorectal cancer (mCRC). However, the mechanism of skin rash development is not well understood. Following exposure to EGFR-targeted therapies, changes in IL-8 levels have been reported. The aim of this study was to evaluate the association between skin rash and inflammatory cytokine levels, including IL-8. Between 2014 and 2017, we prospectively enrolled 38 mCRC patients who underwent chemotherapy with either Cmab or bevacizumab (Bmab) at two hospitals. We performed multiplex cytokine ELISA with 20 inflammatory cytokines including E-selectin, GM-CSF, IFN-alpha, IFN-γ, IL-1 alpha, IL-1 beta, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, IL-17A, IP-10, MCP-1, MIP-1 alpha, MIP-1 beta, P-selectin, sICAM-1, and TNF-alpha at baseline before cycle 1, 24 h after cycle 1, before cycle 2 (= 14 d), and before cycle 3 (= 28 d). Cytokine levels were compared using ANOVA after log-transformation. IL-8 genotypes in 30 patients treated with Cmab were determined using the polymerase chain reaction restriction fragment length polymorphism technique. Depending on the RAS mutational status, 30 and eight patients were treated with Cmab and Bmab-based chemotherapy, respectively. Skin rash developed in 23 (76.6%) of the 30 patients treated with Cmab plus FOLFIRI, after cycle 1. Only the mean log-transformed serum IL-8 level in patients with skin toxicity was statistically lower (2.83 ± 0.15) than in patients who did not experience skin toxicity (3.65 ± 0.27) and received Bmab (3.10 ± 0.26) (ANOVA test, p value = 0.0341). In addition, IL-8 polymorphism did not affect IL-8 levels, skin toxicity, or tumor response in Cmab treated patients. This study suggests that the inflammatory cytokine levels might be affected by Cmab exposure and are associated with the development of skin rash in mCRC patients. Further studies are warranted to evaluate this interaction in Cmab treated patients.


Subject(s)
Cetuximab , Colorectal Neoplasms , Exanthema , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Cetuximab/adverse effects , Chemokine CXCL10 , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , E-Selectin , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Exanthema/etiology , Exanthema/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Interleukin-10 , Interleukin-13 , Interleukin-17 , Interleukin-1alpha , Interleukin-1beta , Interleukin-4 , Interleukin-6 , Interleukin-8 , P-Selectin , Tumor Necrosis Factor-alpha/therapeutic use
8.
Lung Cancer ; 172: 117-123, 2022 10.
Article in English | MEDLINE | ID: mdl-36063602

ABSTRACT

OBJECTIVES: Currently, whether patients with rare epidermal growth factor receptor (EGFR) mutations could benefit from EGFR tyrosine kinase inhibitors (TKIs) demands further studies. Limited clinical data are available regarding the molecular characteristics and clinical response in non-small-cell lung cancer (NSCLC) patients harboring EGFR E709-T710delinsX mutations, a rare mutation type in exon 18 of EGFR. In this study, we aimed to explore the molecular distribution and clinical outcome of EGFR E709-T710delinsX mutated Chinese patients. METHODS: Next-generation sequencing (NGS) tests were performed in 15,078 NSCLC patients. A multicenter retrospective cohort involving 17 advanced lung cancer patients with EGFR E709-T710delinsX was collected to evaluate clinical responses to diverse therapies. In silico protein structure modeling was conducted to predict drug response. RESULTS: 5905 EGFR mutant patients (39.2%, 5905/15078) were identified, with 26 cases (0.44%, 26/5905) harbored EGFR E709-T710delinsD. Afatinib showed a better overall objective response rate (ORR) compared with the first-generation (1G) EGFR TKIs and chemotherapy with significant difference. Superior progression free survival (PFS) was also observed in patients treated with afatinib (afatinib 10.85 m vs 1G EGFR TKIs 4.0 m vs chemotherapy 2.8 m, p = 0.0007). In silico protein structure modeling predicts better binding of afatinib with EGFR E709-T710delinsD compared with other EGFR TKIs. CONCLUSIONS: This is the largest studies for EGFR E709-T710delinsX, with 26 cases with EGFR E709-T710delinsX being identified (0.44% in EGFR mutant NSCLC, 0.17% in NSCLC patients). This study also firstly revealed that afatinib might exert superior antitumor activity to the 1G EGFR TKIs and chemotherapy in EGFR E709-T710delinsX mutant patients.


Subject(s)
Afatinib , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Afatinib/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/drug effects , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies
9.
Crit Rev Oncol Hematol ; 174: 103681, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35462030

ABSTRACT

The human epidermal growth factor receptor (HER, ErbB) family has four members, epidermal growth factor receptor (EGFR), HER2, HER3, and HER4. Although distinct in ligands and functions, all of the HER family members are receptor tyrosine kinases playing important roles in the pathogenesis of cancers. In the era of precision medicine, the HER family is one of the most important and successful cancer therapeutic targets, hallmarked by the approval of anti-EGFR therapies for the treatment of colorectal cancer and non-small cell lung cancer, and anti-HER2 therapies for the treatment of breast cancer and gastric cancer. This review briefly discusses how HER family members were discovered, their functions and roles in cancer, and most importantly, the developmental history and recent updates of therapies targeting HER family members, with colorectal cancer as a focus. We also discussed the patient selection and drug resistance to anti-EGFR therapies in the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms , ErbB Receptors , Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism
10.
Biomed Pharmacother ; 146: 112608, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35062071

ABSTRACT

BACKGROUND: Gastric ulcer (GU) belongs to gastric mucosal irritation and damage. 20(S)-ginsenoside Rg3 (Rg3) has shown anti-oxidant, antiinflammation, and tissue repair effects which are essential for GU treatment. However, the solubility of Rg3 is poor and low gastrointestinal absorption may limit its anti-ulcer effects. As a result, we aim to increase the gastric retention time and gastric absorption of Rg3 to achieve better GU treatment efficacy. METHODS: The mPEG-b-P(Glu-co-Phe) nanoparticles loaded with Rg3 (Rg3-NPs) were developed. The characteristics of Rg3-NPs, including the morphology, diameter, and stability were analyzed. The Rg3 release profiles, gastric retention of Rg3, in vitro cytotoxicity, and pharmacokinetics of Rg3 were assessed. An alcohol-induced rats GU model was performed, and the rats were randomly separated into five treatment groups. Biochemical analysis, gross evaluation, histopathology, and immunohistochemical analysis were applied to further analyze the anti-ulcer effects of Rg3-NPs. RESULTS: Rg3-NPs were successfully prepared and the Rg3 release was pH sensitive. The gastric retention time of Rg3 is longer in Rg3-NPs group than that in Rg3 group. By slightly increasing nitric oxide (NO), obviously increasing epidermal growth factor (EGF), EGF receptor (EGFR), and superoxide dismutase (SOD), and decreasing endothelin-1 (ET-1) and nitric oxide synthase (NOS2), Rg3-NPs possess better GU treatment efficacy than Rg3. CONCLUSIONS: Rg3-NPs can increase gastric retention time and gastric absorption of Rg3 and promote its GU treatment efficacy.


Subject(s)
Ginsenosides/pharmacokinetics , Glutamic Acid/analogs & derivatives , Phenylalanine/analogs & derivatives , Polyethylene Glycols/pharmacokinetics , Stomach Ulcer/pathology , Animals , ErbB Receptors/drug effects , Gastric Mucosa/metabolism , Gastrointestinal Absorption , Ginsenosides/administration & dosage , Glutamic Acid/administration & dosage , Glutamic Acid/pharmacokinetics , Nanoparticles/metabolism , Phenylalanine/administration & dosage , Phenylalanine/pharmacokinetics , Polyethylene Glycols/administration & dosage , Rats , Rats, Wistar
11.
BMC Pulm Med ; 22(1): 36, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35027025

ABSTRACT

BACKGROUND: The resistance to epidermal growth factor receptor (EGFR)- tyrosine kinase inhibitors (TKIs) therapy is currently the major clinical challenge in the treatment of lung cancer. This study aims to reveal the role of glucagon-like peptide (GLP) 2 and GLP-2 receptor (GLP2R) signaling on the EGFR-TKIs and cisplatin resistance of lung cancer cells. METHODS: The common differentially expressed genes in PC9 and HCC827 cells that were individually resistant to one of the three EGFR-TKIs (dacomitinib, osimertinib and afatinib) were screened. The data were from GSE168043 and GSE163913. The expression of GLP2R in drug-resistant cells was detected by western blot. The effect of GLP2R expression down- or up-regulation on resistance to dacomitinib, osimertinib, afatinib or cisplatin was measured by CCK-8 and flow cytometry assays. The long-acting analog of GLP-2, teduglutide, treated the parental cells. RESULTS: A total of 143 common differentially expressed genes were identified. Compared with the parent cells, the GLP2R expression in drug-resistant cell lines was significantly up-regulated. The exogenous expression of GLP2R in parental cells enhanced cell viability, while knockdown of GLP2R levels in drug-resistant cell lines inhibited cell viability. In addition, teduglutide treatment also enhanced the viability of lung cancer cells. CONCLUSION: GLP2-GLP2R signal may change the sensitivity of cells to EGFR-TKIs and cisplatin. The development of GLP-2 or GLP2R inhibitors may be beneficial to the clinical treatment of lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , ErbB Receptors/drug effects , Glucagon-Like Peptide Receptors/drug effects , Glucagon-Like Peptides/drug effects , Lung Neoplasms/drug therapy , Cell Line, Tumor , ErbB Receptors/genetics , Glucagon-Like Peptide Receptors/genetics , Humans , Lung Neoplasms/genetics , Protein Kinase Inhibitors
12.
Cancer Res Treat ; 54(1): 140-149, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33940786

ABSTRACT

PURPOSE: Epidermal growth factor receptor kinase domain duplication (EGFR-KDD) is a rare and poorly understood oncogenic mutation in non-small cell lung cancer (NSCLC). We aimed to investigate the acquired resistance mechanism of EGFR-KDD against EGFR-TKIs. MATERIALS AND METHODS: We identified EGFR-KDD in tumor tissue obtained from a patient with stage IV lung adenocarcinoma and established the patient-derived cell line SNU-4784. We also established several EGFR-KDD Ba/F3 cell lines: EGFR-KDD wild type (EGFR-KDDWT), EGFR-KDD domain 1 T790M (EGFR-KDDD1T), EGFR-KDD domain 2 T790M (EGFR-KDDD2T), and EGFR-KDD both domain T790M (EGFR-KDDBDT). We treated the cells with EGFR tyrosine kinase inhibitors (TKIs) and performed cell viability assays, immunoblot assays, and ENU (N-ethyl-N-nitrosourea) mutagenesis screening. RESULTS: In cell viability assays, SNU-4784 cells and EGFR-KDDWT Ba/F3 cells were sensitive to 2nd generation and 3rd generation EGFR TKIs. In contrast, the T790M-positive EGFR-KDD Ba/F3 cell lines (EGFR-KDDT790M) were only sensitive to 3rd generation EGFR TKIs. In ENU mutagenesis screening, we identified the C797S mutation in kinase domain 2 of EGFR-KDDBDT Ba/F3 cells. Based on this finding, we established an EGFR-KDD domain 1 T790M/domain 2 cis-T790M+C797S (EGFR-KDDT/T+C) Ba/F3 model, which was resistant to EGFR TKIs and anti-EGFR monoclonal antibody combined with EGFR TKIs. CONCLUSION: Our study reveals that the T790M mutation in EGFR-KDD confers resistance to 1st and 2nd generation EGFR TKIs, but is sensitive to 3rd generation EGFR TKIs. In addition, we identified that the C797S mutation in kinase domain 2 of EGFR-KDDT790M mediates a resistance mechanism against 3rd generation EGFR TKIs.


Subject(s)
Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/drug effects , Lung Neoplasms/genetics , Cell Line, Tumor , Erlotinib Hydrochloride/administration & dosage , Humans , Mutation/drug effects , Protein Kinase Inhibitors/administration & dosage
13.
Nat Commun ; 12(1): 7014, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853306

ABSTRACT

Inhibition of RTK pathways in cancer triggers an adaptive response that promotes therapeutic resistance. Because the adaptive response is multifaceted, the optimal approach to blunting it remains undetermined. TNF upregulation is a biologically significant response to EGFR inhibition in NSCLC. Here, we compared a specific TNF inhibitor (etanercept) to thalidomide and prednisone, two drugs that block TNF and also other inflammatory pathways. Prednisone is significantly more effective in suppressing EGFR inhibition-induced inflammatory signals. Remarkably, prednisone induces a shutdown of bypass RTK signaling and inhibits key resistance signals such as STAT3, YAP and TNF-NF-κB. Combined with EGFR inhibition, prednisone is significantly superior to etanercept or thalidomide in durably suppressing tumor growth in multiple mouse models, indicating that a broad suppression of adaptive signals is more effective than blocking a single component. We identify prednisone as a drug that can effectively inhibit adaptive resistance with acceptable toxicity in NSCLC and other cancers.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Glucocorticoids/pharmacology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung , Cytokines/metabolism , Disease Models, Animal , ErbB Receptors/drug effects , Female , Humans , Lung Neoplasms , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Prednisone , STAT3 Transcription Factor/metabolism , Thalidomide , Tumor Necrosis Factor Inhibitors , Up-Regulation
14.
Adv Sci (Weinh) ; 8(22): e2101999, 2021 11.
Article in English | MEDLINE | ID: mdl-34622577

ABSTRACT

Targeting the epidermal growth factor receptor (EGFR) with tyrosine kinase inhibitors (TKIs) is one of the major precision medicine treatment options for lung adenocarcinoma. Due to common development of drug resistance to first- and second-generation TKIs, third-generation inhibitors, including osimertinib and rociletinib, have been developed. A model of EGFR-driven lung cancer and a method to develop tumors of distinct epigenetic states through 3D organotypic cultures are described here. It is discovered that activation of the EGFR T790M/L858R mutation in lung epithelial cells can drive lung cancers with alveolar or bronchiolar features, which can originate from alveolar type 2 (AT2) cells or bronchioalveolar stem cells, but not basal cells or club cells of the trachea. It is also demonstrated that these clones are able to retain their epigenetic differences through passaging orthotopically in mice and crucially that they have distinct drug vulnerabilities. This work serves as a blueprint for exploring how epigenetics can be used to stratify patients for precision medicine decisions.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , ErbB Receptors/drug effects , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Nude , Precision Medicine/methods , Treatment Outcome
15.
BMC Complement Med Ther ; 21(1): 239, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34560864

ABSTRACT

BACKGROUND: According to the recent global cancer statistics, breast cancer is the leading cause of deaths among women with 2.3 million new cases globally. Likewise, cervical cancer is also among the leading causes of mortality among women. Polygonum hydropiper is traditionally known for its cytotoxic effects and several bioactive cytotoxic compounds were isolated from it. This study was aimed to isolate potential anticancer compounds from its most potent fractions and evaluate their anticancer potentials. METHODS: Based on our earlier studies, active fractions including chloroform and ethyl acetate were subjected to column chromatography for isolation of compounds. Chemical structures of isolated compounds were confirmed via 1H NMR, 13C NMR, mass spectrometry. Purified compounds were tested for cytotoxicity against breast cancer cells (MCF-7), cervical cancer cells (HeLA) and NIH/3T3 fibroblasts cells cultures using MTT assy. Anti-angiogenic potentials of isolated compounds were evaluated via chorioallantoic membrane assay. Anti-tumor studies were done using Agrobacterium tumefaciens induced potato tumor assay. Furthermore, to understand the binding modes of Isolated compounds, molecular docking was performed against EGFR, HER2 and VEGFR using MOE as docking software. RESULTS: Two bioactive compounds PH-1 (4-methyl-5-oxo-tetrahydrofuran-3-yl acetate) and PH-2 (methyl 4-hydroxy-3-methoxybenzoate) were purified from the active fractions. In cytotoxicity studies, PH-1 exhibited highest cytotoxicity against HeLA cells with 87.50% lethality at 1 mgmL-1 concentration and LD50 of 60 µgmL-1. Likewise, PH-2 showed 82.33% cytotoxicity against HeLA cells with LD50 of 160 µgmL-1. Similarly, PH-1 and PH-2 exhibited LD50 of 170 and 380 µgmL-1 respectively. Moreover, PH-1 and PH-2 were also very potent cytotoxic compounds against NIH/3T3 cells with 81.45 and 85.55% cytotoxicity at 1 mgL-1 concentration and LD50 of 140 and 58 µgL-1 respectively. Isolated compounds exhibited considerable anti-angiogenic potentials with IC50 of 340 and 500 µgL-1 respectively for PH-1 and PH-2. In anti-tumor assay, PH-1 and PH-2 exhibited 81.15 and 76.09% inhibitions with LD50 of 340 and 550 µgL-1 respectively. Both compounds selectively binds with EGFR and HER2 receptors with low binding energies. Both compounds exhibited stronger interactions with VEGFR through binding pocket residues Lys868, Val916 and Asp1046. CONCLUSIONS: Both compounds cause considerable cytotoxicity against cancer cells. The anti-angiogenic and anti-tumor results suggests additional tumor suppressive properties. Docking analysis suggests that these compound not only has the ability to bind to EGFR and HER2 but also equally binds to VEGFR and may act as potential anti-angiogenic agents.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Phytochemicals/pharmacology , Polygonum , Antineoplastic Agents, Phytogenic/toxicity , Cell Culture Techniques , ErbB Receptors/drug effects , Female , HeLa Cells/drug effects , Humans , MCF-7 Cells , Molecular Docking Simulation , Phytochemicals/toxicity , Receptor, ErbB-2/drug effects , Receptors, Vascular Endothelial Growth Factor/drug effects
16.
Thorac Cancer ; 12(19): 2585-2593, 2021 10.
Article in English | MEDLINE | ID: mdl-34490724

ABSTRACT

BACKGROUND: Transformation to small cell lung cancer (SCLC) is a resistance mechanism of epidermal growth factor receptor (EGFR) mutant lung adenocarcinoma (LADC) patients treated with EGFR tyrosine kinase inhibitors (TKIs). Here, we describe the clinical characteristics and prognosis of these patients and explore the treatment modes after transformation. METHODS: EGFR-mutant LADC patients with SCLC transformation were retrospectively included in the study. Demographic and clinical data were collected. Survival outcomes and corresponding influential factors were analyzed. RESULTS: Twenty-nine patients were included in the study. The median progression-free survival (PFS) of patients who received first-line EGFR-TKIs was 13.1 months. The median time to SCLC transformation was 27.5 months. After transformation, the objective response rates of patients who received first-line chemotherapy with or without EGFR-TKIs were 43.8% and 37.5%, respectively. The median PFS of patients reveiving chemotherapy with EGFR-TKIs was significantly longer than that of patients receiving chemotherapy without EGFR-TKIs (5.2 vs. 3.0 months; HR, 0.19; 95% CI: 0.05-0.72; p = 0.014). However, there was no significant difference in median overall survival (OS) between patients who received chemotherapy with or without EGFR-TKIs (14.8 vs. 13.0 months; p = 0.474). In the multivariate Cox proportional hazards regression analysis, both anti-angiogenic treatment (HR, 0.04; 95% CI: 0.01-0.29; p = 0.001) and local radiotherapy (HR, 0.28; 95% CI: 0.08-0.97; p = 0.044) were significantly associated with better patient OS after transformation. CONCLUSIONS: Compared with chemotherapy alone, the combination of chemotherapy and EGFR-TKIs as first-line treatment after SCLC transformation can benefit patients in PFS but not in OS. However, anti-angiogenic therapies and local radiotherapy can significantly prolong OS after transformation.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , ErbB Receptors/drug effects , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Small Cell Lung Carcinoma/drug therapy , Adenocarcinoma of Lung/genetics , Adult , Aged , Drug Therapy, Combination , ErbB Receptors/genetics , Female , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Mutation , Progression-Free Survival , Retrospective Studies , Small Cell Lung Carcinoma/genetics
17.
J Virol ; 95(20): e0119521, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34379506

ABSTRACT

Zika virus (ZIKV) is a flavivirus that is well known for the epidemic in the Americas in 2015 and 2016 in which microcephaly in newborns and other neurological complications were connected to ZIKV infection. Many aspects of the ZIKV viral life cycle, including binding and entry into the host cell, are still enigmatic. Based on the observation that CHO cells lack expression of the epidermal growth factor receptor (EGFR) and are not permissive for various ZIKV strains, the relevance of EGFR for the viral life cycle was analyzed. Infection of A549 cells by ZIKV leads to a rapid internalization of EGFR that colocalizes with the endosomal marker EEA1. Moreover, infection by different ZIKV strains is associated with an activation of EGFR and the subsequent activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling cascade. However, treatment of the cells with methyl-ß-cyclodextrin (MßCD), which on the one hand leads to an activation of EGFR but on the other hand prevents EGFR internalization, impairs ZIKV infection. Specific inhibition of EGFR or of the Ras-Raf-MEK-ERK signal transduction cascade hinders ZIKV infection by inhibition of ZIKV entry. In accordance with this, knockout of EGFR expression impedes ZIKV entry. In the case of an already established infection, inhibition of EGFR or of downstream signaling does not affect viral replication. Taken together, these data demonstrate the relevance of EGFR in the early stages of ZIKV infection and identify EGFR as a target for antiviral strategies. IMPORTANCE These data deepen the knowledge about the ZIKV infection process and demonstrate the relevance of EGFR for ZIKV entry. In light of the fact that a variety of specific and efficient inhibitors of EGFR and of EGFR-dependent signaling have been developed and licensed, repurposing of these substances could be a helpful tool to prevent the spreading of ZIKV infection in an epidemic outbreak.


Subject(s)
Virus Internalization/drug effects , Zika Virus/metabolism , A549 Cells , Animals , CHO Cells , Cell Line , Chlorocebus aethiops , Cricetulus , ErbB Receptors/drug effects , ErbB Receptors/metabolism , ErbB Receptors/physiology , Host Microbial Interactions/physiology , Humans , Life Cycle Stages , Signal Transduction/drug effects , Vero Cells , Virus Replication/genetics , Virus Replication/physiology , Zika Virus/pathogenicity , Zika Virus Infection/virology , beta-Cyclodextrins/pharmacology
18.
Medicine (Baltimore) ; 100(30): e26375, 2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34397683

ABSTRACT

RATIONALE: Besides the T790 M mutation, it may coexist with bypass pathway activation in real clinical cases for patients with EGFR mutations who resisted to the first- and second-generation tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC). There are limited clinical trial data describing the efficacy of osimertinib combined with MET inhibition in EGFR T790M-mutant NSCLC patients with Met amplification. PATIENT CONCERNS: A non-smoking 53-year-old male patient with lung adenocarcinoma underwent gefitinib, afatinib, and osimertinib combined with crizotinib treatment and developed different EGFR resistance mutations. DIAGNOSES: The patient was diagnosed with lung adenocarcinoma (stage cT4N2M0, IIIB). After resistance to the therapy targeting EGFR exon 21 L858R point mutation, T790 M mutation was detected in liquid biopsy and Met amplification was detected via tissue biopsy by next-generation sequencing (NGS). INTERVENTIONS: The patient received systemic treatments, including chemotherapy, gefitinib, afatinib, and osimertinib combined with crizotinib. OUTCOMES: The patient died of multisystem organ failure and had an overall survival of 24 months. LESSONS: Although osimertinib combined with crizotinib therapy showed dramatic tumor shrinkage in both the primary tumor and bone metastasis to an EGFR T790M-mutant NSCLC patient with MET amplification, the progression-free survival (PFS) was only two months.


Subject(s)
Acrylamides/adverse effects , Aniline Compounds/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Crizotinib/adverse effects , Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Crizotinib/therapeutic use , ErbB Receptors/drug effects , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Tomography, X-Ray Computed/methods
19.
Drugs ; 81(11): 1349-1353, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34292533

ABSTRACT

Amivantamab (amivantamab-vmjw; Rybrevant™), a bispecific monoclonal antibody targeting epidermal growth factor receptor (EGFR) and mesenchymal epithelial transition factor (MET), is being developed by Janssen Biotech for the treatment of non-small cell lung cancer (NSCLC). On 21 May 2021, amivantamab received its first approval in the USA for the treatment of adult patients with locally advanced or metastatic NSCLC harbouring EGFR Exon 20 insertion mutations whose disease has progressed on or after platinum-based chemotherapy. Amivantamab is in preregistration for NSCLC in the EU, Australia, Japan, Canada, Switzerland and China. This article summarizes the milestones in the development of amivantamab leading to this first approval for NSCLC.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/pathology , Drug Approval , ErbB Receptors/drug effects , ErbB Receptors/genetics , Humans , Lung Neoplasms/pathology , Multicenter Studies as Topic , Neoplasm Metastasis , Neoplasm Staging , Randomized Controlled Trials as Topic , United States , United States Food and Drug Administration
20.
Clin Lung Cancer ; 22(6): 541-548, 2021 11.
Article in English | MEDLINE | ID: mdl-34140248

ABSTRACT

BACKGROUND: Onalespib is a novel heat shock protein 90 inhibitor (HSP90i). Previous preclinical and clinical studies with HSP90i have demonstrated activity in EGFR-mutant non-small cell lung cancer (NSCLC). This study sought to determine the safety and tolerability of onalespib plus erlotinib in EGFR-mutant NSCLC and to evaluate the preliminary efficacy of the combination in epidermal growth factor receptor exon 20 insertion (EGFRex20ins) NSCLC. PATIENTS AND METHODS: Standard 3+3 dose escalation was followed by a phase II expansion in EGFRex20ins. The phase II component targeted a response rate of 25% versus a background rate of 5%. Prospective next-generation sequencing (NGS) of 70 cancer-related genes, including EGFR, via plasma circulating tumor DNA (ctDNA) was performed. Toxicity was graded by Common Terminology Criteria for Adverse Events (CTCAE), version 4, and response was determined by Response Evaluation Criteria in Solid Tumours (RECIST) 1.1. RESULTS: Eleven patients were treated (nine dose escalation, two dose expansion). Two dose-limiting toxicities (DLTs) occurred in dose level (DL) 0 and zero in DL -1 (minus). In 10 EGFRex20ins patients, no responses were observed, median progression-free survival was 5.4 months (95% confidence interval, 0.9-5.7), and the disease control rate (DCR) was 40% (median, 3.5 months). EGFRex20ins was detected in nine of 10 ctDNA samples at baseline; on-treatment ctDNA clearance was not observed. Grade 3 diarrhea was the predominant toxicity in 45% of patients. The recommended phase II dose is DL -1 (minus): erlotinib 150 mg orally every morning and onalespib 120 mg/m2 intravenously on days 1, 8, and 15 every 28 days. CONCLUSION: Overlapping toxicities of erlotinib and onalespib, mainly diarrhea, limited the tolerability of this combination, and limited clinical activity was observed, so the trial was closed early. Plasma EGFRex20ins ctDNA was detected in the majority of patients; failure to clear ctDNA was consistent with lack of tumor response (NCT02535338).


Subject(s)
Benzamides/administration & dosage , Benzamides/pharmacology , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/pharmacology , Isoindoles/administration & dosage , Isoindoles/pharmacology , Lactates/therapeutic use , Lung Neoplasms/drug therapy , Mutation/drug effects , Mutation/genetics , Aged , California , ErbB Receptors/drug effects , ErbB Receptors/genetics , Female , Humans , Male , Middle Aged , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...