General Information of the Molecule (ID: Mol04140)
Name
Oncogenic epidermal growth factor receptor (EGFR) ,Homo sapiens
Synonyms
Proto-oncogene c-ErbB-1; Receptor tyrosine-protein kinase erbB-1
    Click to Show/Hide
Molecule Type
Protein
Gene Name
EGFR
Gene ID
1956
Location
chr7:55019017-55211628[+]
Sequence
MRPSGTAGAALLALLAALCPASRALEEKKVCQGTSNKLTQLGTFEDHFLSLQRMFNNCEV
VLGNLEITYVQRNYDLSFLKTIQEVAGYVLIALNTVERIPLENLQIIRGNMYYENSYALA
VLSNYDANKTGLKELPMRNLQEILHGAVRFSNNPALCNVESIQWRDIVSSDFLSNMSMDF
QNHLGSCQKCDPSCPNGSCWGAGEENCQKLTKIICAQQCSGRCRGKSPSDCCHNQCAAGC
TGPRESDCLVCRKFRDEATCKDTCPPLMLYNPTTYQMDVNPEGKYSFGATCVKKCPRNYV
VTDHGSCVRACGADSYEMEEDGVRKCKKCEGPCRKVCNGIGIGEFKDSLSINATNIKHFK
NCTSISGDLHILPVAFRGDSFTHTPPLDPQELDILKTVKEITGFLLIQAWPENRTDLHAF
ENLEIIRGRTKQHGQFSLAVVSLNITSLGLRSLKEISDGDVIISGNKNLCYANTINWKKL
FGTSGQKTKIISNRGENSCKATGQVCHALCSPEGCWGPEPRDCVSCRNVSRGRECVDKCN
LLEGEPREFVENSECIQCHPECLPQAMNITCTGRGPDNCIQCAHYIDGPHCVKTCPAGVM
GENNTLVWKYADAGHVCHLCHPNCTYGCTGPGLEGCPTNGPKIPSIATGMVGALLLLLVV
ALGIGLFMRRRHIVRKRTLRRLLQERELVEPLTPSGEAPNQALLRILKETEFKKIKVLGS
GAFGTVYKGLWIPEGEKVKIPVAIKELREATSPKANKEILDEAYVMASVDNPHVCRLLGI
CLTSTVQLITQLMPFGCLLDYVREHKDNIGSQYLLNWCVQIAKGMNYLEDRRLVHRDLAA
RNVLVKTPQHVKITDFGLAKLLGAEEKEYHAEGGKVPIKWMALESILHRIYTHQSDVWSY
GVTVWELMTFGSKPYDGIPASEISSILEKGERLPQPPICTIDVYMIMVKCWMIDADSRPK
FRELIIEFSKMARDPQRYLVIQGDERMHLPSPTDSNFYRALMDEEDMDDVVDADEYLIPQ
QGFFSSPSTSRTPLLSSLSATSNNSTVACIDRNGLQSCPIKEDSFLQRYSSDPTGALTED
SIDDTFLPVPEYINQSVPKRPAGSVQNPVYHNQPLNPAPSRDPHYQDPHSTAVGNPEYLN
TVQPTCVNSTFDSPAHWAQKGSHQISLDNPDYQQDFFPKEAKPNGIFKGSTAENAEYLRV
APQSSEFIGA
    Click to Show/Hide
Function
Receptor tyrosine kinase binding ligands of the EGF family and activating several signaling cascades to convert extracellular cues into appropriate cellular responses (PubMed:10805725, PubMed:27153536, PubMed:2790960, PubMed:35538033). Known ligands include EGF, TGFA/TGF- alpha, AREG, epigen/EPGN, BTC/betacellulin, epiregulin/EREG and HBEGF/heparin-binding EGF (PubMed:12297049, PubMed:15611079, PubMed:17909029, PubMed:20837704, PubMed:27153536, PubMed:2790960, PubMed:7679104, PubMed:8144591, PubMed:9419975). Ligand binding triggers receptor homo- and/or heterodimerization and autophosphorylation on key cytoplasmic residues. The phosphorylated receptor recruits adapter proteins like GRB2 which in turn activates complex downstream signaling cascades. Activates at least 4 major downstream signaling cascades including the RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC and STATs modules (PubMed:27153536). May also activate the NF-kappa-B signaling cascade (PubMed:11116146). Also directly phosphorylates other proteins like RGS16, activating its GTPase activity and probably coupling the EGF receptor signaling to the G protein-coupled receptor signaling (PubMed:11602604). Also phosphorylates MUC1 and increases its interaction with SRC and CTNNB1/beta-catenin (PubMed:11483589). Positively regulates cell migration via interaction with CCDC88A/GIV which retains EGFR at the cell membrane following ligand stimulation, promoting EGFR signaling which triggers cell migration (PubMed:20462955). Plays a role in enhancing learning and memory performance (By similarity). Plays a role in mammalian pain signaling (long-lasting hypersensitivity) (By similarity). .; Isoform 2 may act as an antagonist of EGF action.; (Microbial infection) Acts as a receptor for hepatitis C virus (HCV) in hepatocytes and facilitates its cell entry. Mediates HCV entry by promoting the formation of the CD81-CLDN1 receptor complexes that are essential for HCV entry and by enhancing membrane fusion of cells expressing HCV envelope glycoproteins. .
    Click to Show/Hide
Uniprot ID
EGFR_HUMAN
Ensembl ID
ENSG00000146648
HGNC ID
HGNC:3236
        Click to Show/Hide the Complete Species Lineage
Kingdom: Metazoa
Phylum: Chordata
Class: Mammalia
Order: Primates
Family: Hominidae
Genus: Homo
Species: Homo sapiens
Type(s) of Resistant Mechanism of This Molecule
  ADTT: Aberration of the Drug's Therapeutic Target
  MRAP: Metabolic Reprogramming via Altered Pathways
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
12 drug(s) in total
Click to Show/Hide the Full List of Drugs
Afatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [5]
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Afatinib
Molecule Alteration Missense mutation
Exon 20 insertion mutations
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K-Akt signaling pathway Inhibition hsa04151
In Vitro Model Ba/F3 murine cells Bone marrow Homo sapiens (Human) N.A.
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
GeneSeq assay
Experiment for
Drug Resistance
Cell proliferation assay; Immunoblotting assay
Mechanism Description Mechanisms of acquired EGFR TKI resistance of this mutant remained underreported.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [5]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Afatinib
Molecule Alteration Missense mutation
Exon 20 insertion mutations
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K-Akt signaling pathway Inhibition hsa04151
In Vitro Model Ba/F3 murine cells Bone marrow Homo sapiens (Human) N.A.
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
GeneSeq assay
Experiment for
Drug Resistance
Cell proliferation assay; Immunoblotting assay
Mechanism Description Mechanisms of acquired EGFR TKI resistance of this mutant remained underreported.
Cetuximab
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.0] [6]
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Sensitive Drug Cetuximab
Molecule Alteration Expression
Down-regulation
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation EGF-EGFR signaling pathway Regulation N.A.
In Vivo Model Mouse model Mus musculus
Experiment for
Molecule Alteration
Immunoprecipitation assay; LC-MS/MS analysis
Experiment for
Drug Resistance
Cellular ROS and lipid peroxidation level assay; LOXL3 enzymatic assay; In vitro kinase assay
Mechanism Description To overcome chemotherapy resistance, novel strategies sensitizing cancer cells to chemotherapy are required. Here, we screen the lysyl-oxidase (LOX) family to clarify its contribution to chemotherapy resistance in liver cancer. LOXL3 depletion significantly sensitizes liver cancer cells to Oxaliplatin by inducing ferroptosis. Chemotherapy-activated EGFR signaling drives LOXL3 to interact with TOM20, causing it to be hijacked into mitochondria, where LOXL3 lysyl-oxidase activity is reinforced by phosphorylation at S704. Metabolic adenylate kinase 2 (AK2) directly phosphorylates LOXL3-S704. Phosphorylated LOXL3-S704 targets dihydroorotate dehydrogenase (DHODH) and stabilizes it by preventing its ubiquitin-mediated proteasomal degradation. K344-deubiquitinated DHODH accumulates in mitochondria, in turn inhibiting chemotherapy-induced mitochondrial ferroptosis. CRISPR-Cas9-mediated site-mutation of mouse LOXL3-S704 to D704 causes a reduction in lipid peroxidation. Using an advanced liver cancer mouse model, we further reveal that low-dose Oxaliplatin in combination with the DHODH-inhibitor Leflunomide effectively inhibit liver cancer progression by inducing ferroptosis, with increased chemotherapy sensitivity and decreased chemotherapy toxicity.
Cisplatin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.00] [7]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.00]
Resistant Drug Cisplatin
Molecule Alteration phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCA-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213A-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213B-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Cell cycle distribution assay; Colony formation assay
Mechanism Description The results demonstrated that CCA-GemR cells grow more slowly compared to their parental cell lines. Cell cycle analysis revealed an increase in KKU-213A-GemR and KKU-213B-GemR cell accumulation in the G1 phase. Moreover, cross-resistance to 5-FU and cisplatin was observed in all CCA-GemR cells. The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem.
Erlotinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [5]
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Erlotinib
Molecule Alteration Missense mutation
EGFR-K745_E746insIPVAIK; Exon 19 deletion, L858R, L861Q, G719S, A763_Y764insFQEA
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K-Akt signaling pathway Inhibition hsa04151
In Vitro Model Ba/F3 murine cells Bone marrow Homo sapiens (Human) N.A.
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
GeneSeq assay
Experiment for
Drug Resistance
Cell proliferation assay; Immunoblotting assay
Mechanism Description Mechanisms of acquired EGFR TKI resistance of this mutant remained underreported.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [5]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Erlotinib
Molecule Alteration Missense mutation
EGFR-K745_E746insIPVAIK; Exon 19 deletion, L858R, L861Q, G719S, A763_Y764insFQEA
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K-Akt signaling pathway Inhibition hsa04151
In Vitro Model Ba/F3 murine cells Bone marrow Homo sapiens (Human) N.A.
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
GeneSeq assay
Experiment for
Drug Resistance
Cell proliferation assay; Immunoblotting assay
Mechanism Description Mechanisms of acquired EGFR TKI resistance of this mutant remained underreported.
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.00] [7]
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.00]
Sensitive Drug Erlotinib
Molecule Alteration phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCA-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213A-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213B-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Cell cycle distribution assay; Colony formation assay
Mechanism Description The results demonstrated that CCA-GemR cells grow more slowly compared to their parental cell lines. Cell cycle analysis revealed an increase in KKU-213A-GemR and KKU-213B-GemR cell accumulation in the G1 phase. Moreover, cross-resistance to 5-FU and cisplatin was observed in all CCA-GemR cells. The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem.
Fluorouracil
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.00] [7]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.00]
Resistant Drug Fluorouracil
Molecule Alteration phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCA-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213A-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213B-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Cell cycle distribution assay; Colony formation assay
Mechanism Description The results demonstrated that CCA-GemR cells grow more slowly compared to their parental cell lines. Cell cycle analysis revealed an increase in KKU-213A-GemR and KKU-213B-GemR cell accumulation in the G1 phase. Moreover, cross-resistance to 5-FU and cisplatin was observed in all CCA-GemR cells. The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem.
Fulvestrant
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Triple-negative breast cancer [ICD-11: 2C60.9] [8]
Resistant Disease Triple-negative breast cancer [ICD-11: 2C60.9]
Resistant Drug Fulvestrant
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR/HER2 signaling pathway Regulation N.A.
In Vitro Model MCF7 (Ful-R) cells Breast Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Mechanism Description In this study, we investigated the molecular mechanism underlying the loss of ER, FOXO3a, and induction of HER2 in fulvestrant-resistant breast cancer. Short-term fulvestrant treatment degraded ER proteins via the ubiquitin-proteasome degradation pathway in MCF7 cells. MCF7 cells turn into highly proliferative cells (fulvestrant-resistant cells: Ful-R) after long-term fulvestrant treatment. These cells exhibit markedly suppressed estrogen and progesterone receptor levels. The phosphorylation of EGFR, HER2, and ERK was induced in Ful-R, and these phosphorylation inhibitors suppressed cell proliferation in Ful-R.
Disease Class: Triple-negative breast cancer [ICD-11: 2C60.9] [8]
Resistant Disease Triple-negative breast cancer [ICD-11: 2C60.9]
Resistant Drug Fulvestrant
Molecule Alteration Phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR/HER2 signaling pathway Regulation N.A.
In Vitro Model MCF7 (Ful-R) cells Breast Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Mechanism Description In this study, we investigated the molecular mechanism underlying the loss of ER, FOXO3a, and induction of HER2 in fulvestrant-resistant breast cancer. Short-term fulvestrant treatment degraded ER proteins via the ubiquitin-proteasome degradation pathway in MCF7 cells. MCF7 cells turn into highly proliferative cells (fulvestrant-resistant cells: Ful-R) after long-term fulvestrant treatment. These cells exhibit markedly suppressed estrogen and progesterone receptor levels. The phosphorylation of EGFR, HER2, and ERK was induced in Ful-R, and these phosphorylation inhibitors suppressed cell proliferation in Ful-R.
Gefitinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Metabolic Reprogramming via Altered Pathways (MRAP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [9]
Metabolic Type Nucleic acid metabolism
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Gefitinib
Molecule Alteration Mutation
.
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
Experiment for
Molecule Alteration
CE-TOF/MS
Experiment for
Drug Resistance
Cell proliferation assay
Mechanism Description It is confirmed that purine metabolism catalyzed by HPRT1 promotes the proliferation of EGFR-mutant LUAD in vitro and in vivo. Furthermore, the study of the mechanism shows that HIF-1alpha transcriptionally regulates HPRT1 to accelerate purine nucleotides synthesis to promote cell proliferation and tumorigenesis. Finally, inhibition of HPRT1 coupled with EGFR-TKIs significantly inhibits the tumor growth of EGFR-mutant LUAD
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [9]
Metabolic Type Nucleic acid metabolism
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Gefitinib
Molecule Alteration Mutation
.
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PC-9 cells Lung Homo sapiens (Human) CVCL_B260
Experiment for
Molecule Alteration
CE-TOF/MS
Experiment for
Drug Resistance
Cell proliferation assay
Mechanism Description It is confirmed that purine metabolism catalyzed by HPRT1 promotes the proliferation of EGFR-mutant LUAD in vitro and in vivo. Furthermore, the study of the mechanism shows that HIF-1alpha transcriptionally regulates HPRT1 to accelerate purine nucleotides synthesis to promote cell proliferation and tumorigenesis. Finally, inhibition of HPRT2 coupled with EGFR-TKIs significantly inhibits the tumor growth of EGFR-mutant LUAD
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [9]
Metabolic Type Nucleic acid metabolism
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Gefitinib
Molecule Alteration Mutation
.
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model H3255 cells Lung Homo sapiens (Human) CVCL_6831
Experiment for
Molecule Alteration
CE-TOF/MS
Experiment for
Drug Resistance
Cell proliferation assay
Mechanism Description It is confirmed that purine metabolism catalyzed by HPRT1 promotes the proliferation of EGFR-mutant LUAD in vitro and in vivo. Furthermore, the study of the mechanism shows that HIF-1alpha transcriptionally regulates HPRT1 to accelerate purine nucleotides synthesis to promote cell proliferation and tumorigenesis. Finally, inhibition of HPRT3 coupled with EGFR-TKIs significantly inhibits the tumor growth of EGFR-mutant LUAD
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [9]
Metabolic Type Nucleic acid metabolism
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Gefitinib
Molecule Alteration Mutation
.
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model H1975 cells Lung Homo sapiens (Human) CVCL_B0JT
Experiment for
Molecule Alteration
CE-TOF/MS
Experiment for
Drug Resistance
Cell proliferation assay
Mechanism Description It is confirmed that purine metabolism catalyzed by HPRT1 promotes the proliferation of EGFR-mutant LUAD in vitro and in vivo. Furthermore, the study of the mechanism shows that HIF-1alpha transcriptionally regulates HPRT1 to accelerate purine nucleotides synthesis to promote cell proliferation and tumorigenesis. Finally, inhibition of HPRT4 coupled with EGFR-TKIs significantly inhibits the tumor growth of EGFR-mutant LUAD
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [9]
Metabolic Type Nucleic acid metabolism
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Gefitinib
Molecule Alteration Mutation
.
Experimental Note Revealed Based on the Cell Line Data
In Vivo Model Male nude mice Mice
Experiment for
Molecule Alteration
CE-TOF/MS
Experiment for
Drug Resistance
Tumor volume assay
Mechanism Description It is confirmed that purine metabolism catalyzed by HPRT1 promotes the proliferation of EGFR-mutant LUAD in vitro and in vivo. Furthermore, the study of the mechanism shows that HIF-1alpha transcriptionally regulates HPRT1 to accelerate purine nucleotides synthesis to promote cell proliferation and tumorigenesis. Finally, inhibition of HPRT5 coupled with EGFR-TKIs significantly inhibits the tumor growth of EGFR-mutant LUAD
Gemcitabine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.00] [7]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.00]
Resistant Drug Gemcitabine
Molecule Alteration phosphorylation
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model KKU-213A-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Cell cycle distribution assay; Colony formation assay
Mechanism Description The results demonstrated that CCA-GemR cells grow more slowly compared to their parental cell lines. Cell cycle analysis revealed an increase in KKU-213A-GemR and KKU-213B-GemR cell accumulation in the G1 phase. Moreover, cross-resistance to 5-FU and cisplatin was observed in all CCA-GemR cells. The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem.
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.00] [7]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.00]
Resistant Drug Gemcitabine
Molecule Alteration phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCA-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213B-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Cell cycle distribution assay; Colony formation assay
Mechanism Description The results demonstrated that CCA-GemR cells grow more slowly compared to their parental cell lines. Cell cycle analysis revealed an increase in KKU-213A-GemR and KKU-213B-GemR cell accumulation in the G1 phase. Moreover, cross-resistance to 5-FU and cisplatin was observed in all CCA-GemR cells. The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem.
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.0] [7]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Resistant Drug Gemcitabine
Molecule Alteration Phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR signaling pathway Inhibition hsa01521
In Vitro Model CCA-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Protein component assay; Western blot assay
Mechanism Description The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem. EGFR is a potential target molecule for reducing Gem resistance and enhancing its anti-tumor effects in patients with CCA.
Ivermectin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [ICD-11: 2A20.0] [10]
Sensitive Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Sensitive Drug Ivermectin
Molecule Alteration Phosphorylation
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR/STAT3/ERK signalling pathway Regulation N.A.
In Vitro Model K562/FLM cells Blood Homo sapiens (Human) CVCL_E7CM
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description It was found that ivermectin effectively suppressed the expression of autophagy and transport proteins in K562/FLM cells, reduced the activity of the aforementioned phosphoproteins, and promoted apoptotic cell death. The significant effects of ivermectin might offer a novel therapeutic strategy to overcome flumatinib resistance and optimize the treatment outcomes of CML.
Lenvatinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.0] [6]
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Sensitive Drug Lenvatinib
Molecule Alteration Expression
Down-regulation
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation EGF-EGFR signaling pathway Regulation N.A.
In Vivo Model Mouse model Mus musculus
Experiment for
Molecule Alteration
Immunoprecipitation assay; LC-MS/MS analysis
Experiment for
Drug Resistance
Cellular ROS and lipid peroxidation level assay; LOXL3 enzymatic assay; In vitro kinase assay
Mechanism Description To overcome chemotherapy resistance, novel strategies sensitizing cancer cells to chemotherapy are required. Here, we screen the lysyl-oxidase (LOX) family to clarify its contribution to chemotherapy resistance in liver cancer. LOXL3 depletion significantly sensitizes liver cancer cells to Oxaliplatin by inducing ferroptosis. Chemotherapy-activated EGFR signaling drives LOXL3 to interact with TOM20, causing it to be hijacked into mitochondria, where LOXL3 lysyl-oxidase activity is reinforced by phosphorylation at S704. Metabolic adenylate kinase 2 (AK2) directly phosphorylates LOXL3-S704. Phosphorylated LOXL3-S704 targets dihydroorotate dehydrogenase (DHODH) and stabilizes it by preventing its ubiquitin-mediated proteasomal degradation. K344-deubiquitinated DHODH accumulates in mitochondria, in turn inhibiting chemotherapy-induced mitochondrial ferroptosis. CRISPR-Cas9-mediated site-mutation of mouse LOXL3-S704 to D704 causes a reduction in lipid peroxidation. Using an advanced liver cancer mouse model, we further reveal that low-dose Oxaliplatin in combination with the DHODH-inhibitor Leflunomide effectively inhibit liver cancer progression by inducing ferroptosis, with increased chemotherapy sensitivity and decreased chemotherapy toxicity.
Osimertinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [5]
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug Osimertinib
Molecule Alteration Missense mutation
Exon 20 insertion mutations
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K-Akt signaling pathway Inhibition hsa04151
In Vitro Model Ba/F3 murine cells Bone marrow Homo sapiens (Human) N.A.
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
GeneSeq assay
Experiment for
Drug Resistance
Cell proliferation assay; Immunoblotting assay
Mechanism Description Mechanisms of acquired EGFR TKI resistance of this mutant remained underreported.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Lung adenocarcinoma [ICD-11: 2C25.0] [5]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Osimertinib
Molecule Alteration Missense mutation
Exon 20 insertion mutations
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K-Akt signaling pathway Inhibition hsa04151
In Vitro Model Ba/F3 murine cells Bone marrow Homo sapiens (Human) N.A.
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
GeneSeq assay
Experiment for
Drug Resistance
Cell proliferation assay; Immunoblotting assay
Mechanism Description Mechanisms of acquired EGFR TKI resistance of this mutant remained underreported.
Regorafenib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.0] [2]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Resistant Drug Regorafenib
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation RAS/RAF/ERK signaling pathway Activation hsa04010
In Vitro Model 7721-R cells Liver Homo sapiens (Human) N.A.
97H-R cells Liver Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Annexin V-FITC/PI double-staining assay; Flow cytometry assay; Colony formation assay; CCK8 assay
Mechanism Description The expression of EGFR, a member of the receptor tyrosine kinase (RTK) family, was significantly increased in acquired regorafenib-resistant HCC cells compared with parental cells. Pharmacological inhibition of EGFR with gefitinib restored the sensitivity of regorafenib-resistant HCC cells to regorafenib. In a xenograft mouse model, gefitinib sensitized resistant tumors to regorafenib. Additionally, levels of RAS, RAF, and P-ERK1/2, components of the downstream EGFR signaling pathway, were positively associated with EGFR expression. EGFR overexpression promotes acquired resistance to regorafenib through RAS/RAF/ERK bypass activation in HCC.
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.0] [2]
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Resistant Drug Regorafenib
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation RAS/RAF/ERK signaling pathway Activation hsa04010
In Vitro Model SMMC-7721 cells Liver Homo sapiens (Human) CVCL_0534
MHCC97H cells Liver Homo sapiens (Human) CVCL_4972
In Vivo Model BALB/c nude xenograft model Mus musculus
Experiment for
Molecule Alteration
High-throughput proteomics assay
Experiment for
Drug Resistance
Direct microscopic assay; CCK8 assay; Colony formation assay; Annexin V-FITC/propidium iodide double staining assay; Cell cycle assay; Western blot assay; A xenograft model assay
Mechanism Description The expression of EGFR, a member of the receptor tyrosine kinase (RTK) family, was significantly increased in acquired regorafenib-resistant HCC cells compared with parental cells. Pharmacological inhibition of EGFR with gefitinib restored the sensitivity of regorafenib-resistant HCC cells to regorafenib. In a xenograft mouse model, gefitinib sensitized resistant tumors to regorafenib. Additionally, levels of RAS, RAF, and P-ERK1/2, components of the downstream EGFR signaling pathway, were positively associated with EGFR expression.
Clinical Trial Drug(s)
1 drug(s) in total
Click to Show/Hide the Full List of Drugs
Flumatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [ICD-11: 2A20.0] [10]
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Resistant Drug Flumatinib
Molecule Alteration Phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR/STAT3/ERK signalling pathway Regulation N.A.
In Vitro Model K562/FLM cells Blood Homo sapiens (Human) CVCL_E7CM
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Through cellular experimentation, we explored the resistance mechanisms, which indicated that K562/FLM cells evade flumatinib cytotoxicity by enhancing autophagy, increasing the expression of membrane transport proteins, particularly P-glycoprotein, ABCC1 and ABCC4, as well as enhancing phosphorylation of p-EGFR, p-ERK and p-STAT3 proteins.
Investigative Drug(s)
1 drug(s) in total
Click to Show/Hide the Full List of Drugs
Erlotinib/Gemcitabine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Cholangiocarcinoma [ICD-11: 2C12.00] [7]
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.00]
Sensitive Drug Erlotinib/Gemcitabine
Molecule Alteration phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCA-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213A-GemR cells Bile duct Homo sapiens (Human) N.A.
KKU-213B-GemR cells Bile duct Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay
Experiment for
Drug Resistance
Cell cycle distribution assay; Colony formation assay
Mechanism Description The results demonstrated that CCA-GemR cells grow more slowly compared to their parental cell lines. Cell cycle analysis revealed an increase in KKU-213A-GemR and KKU-213B-GemR cell accumulation in the G1 phase. Moreover, cross-resistance to 5-FU and cisplatin was observed in all CCA-GemR cells. The Proteome Profiler Human Phospho-Kinase Array showed increased phosphorylation of EGFR in CCA-GemR cells. Erlotinib, a specific inhibitor of EGFR, significantly enhanced the anti-tumor activity of Gem with a synergistic effect (Combination index <1). Western blot analysis confirmed that phosphorylation of EGFR increased in cells treated with Gem, whereas the expression was significantly decreased in cells treated with either erlotinib alone or in combination with Gem.
References
Ref 1 Coccinic acid exhibits anti-tumor efficacy against NSCLC harboring EGFR L858R/T790M mutation via the EGFR/STAT3 pathway. Bioorg Chem. 2025 Jan;154:108038.
Ref 2 EGFR bypass activation mediates acquired resistance to regorafenib in hepatocellular carcinoma. Front Med (Lausanne). 2024 Nov 13;11:1464610.
Ref 3 Guggulsterone from Commiphora mukul potentiates anti-glioblastoma efficacy of temozolomide in vitro and in vivo via down-regulating EGFR/PI3K/Akt signaling and NF-kappaB activation. J Ethnopharmacol. 2023 Jan 30;301:115855.
Ref 4 An allosteric inhibitor against the therapy-resistant mutant forms of EGFR in non-small cell lung cancer. Nat Cancer. 2022 Apr;3(4):402-417.
Ref 5 EGFR exon 19 insertion EGFR-K745_E746insIPVAIK and others with rare XPVAIK amino-acid insertions: Preclinical and clinical characterization of the favorable therapeutic window to all classes of approved EGFR kinase inhibitors. Lung Cancer. 2023 Jul;181:107250.
Ref 6 Lysyl oxidase-like 3 restrains mitochondrial ferroptosis to promote liver cancer chemoresistance by stabilizing dihydroorotate dehydrogenase. Nat Commun. 2023 May 30;14(1):3123.
Ref 7 Targeting EGFR Activation to Overcome Gemcitabine Resistance in Cholangiocarcinoma. Anticancer Res. 2024 Dec;44(12):5393-5404.
Ref 8 Loss of ERalpha involved-HER2 induction mediated by the FOXO3a signaling pathway in fulvestrant-resistant breast cancer. Biochem Biophys Res Commun. 2025 Jan;742:151056.
Ref 9 HIF-1alpha-HPRT1 axis promotes tumorigenesis and gefitinib resistance by enhancing purine metabolism in EGFR-mutant lung adenocarcinoma. J Exp Clin Cancer Res. 2024 Sep 30;43(1):269.
Ref 10 Overcoming flumatinib resistance in chronic myeloid leukaemia: Insights into cellular mechanisms and ivermectin's therapeutic potential. J Cell Mol Med. 2024 Jul;28(14):e18539.

If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Yu.