Molecule Information
General Information of the Molecule (ID: Mol00061)
Name |
Epidermal growth factor receptor (EGFR)
,Homo sapiens
|
||||
---|---|---|---|---|---|
Synonyms |
Proto-oncogene c-ErbB-1; Receptor tyrosine-protein kinase erbB-1; ERBB; ERBB1; HER1
Click to Show/Hide
|
||||
Molecule Type |
Protein
|
||||
Gene Name |
EGFR
|
||||
Gene ID | |||||
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. Known ligands include EGF, TGFA/TGF-alpha, AREG, epigen/EPGN, BTC/betacellulin, epiregulin/EREG and HBEGF/heparin-binding EGF. 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. May also activate the NF-kappa-B signaling cascade. 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. Also phosphorylates MUC1 and increases its interaction with SRC and CTNNB1/beta-catenin. 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. Plays a role in enhancing learning and memory performance.
Click to Show/Hide
|
||||
Uniprot ID | |||||
Ensembl ID | |||||
HGNC ID | |||||
Click to Show/Hide the Complete Species Lineage | |||||
Type(s) of Resistant Mechanism of This Molecule
ADTT: Aberration of the Drug's Therapeutic Target
EADR: Epigenetic Alteration of DNA, RNA or Protein
UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
25 drug(s) in total
Afatinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [1], [2], [3] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Afatinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Exon sequencing assay | |||
Experiment for Drug Resistance |
Progression-free and post-progression survival asaay | |||
Mechanism Description | T790M was detected in half of the lung adenocarcinoma after acquiring resistance to afatinib. T790M is still the major acquired resistance mechanism. First-generation EGFR TkI exposure did not influence the prevalence of T790M in lung cancer acquired resistance to afatinib. | |||
Disease Class: Non-small cell lung cancer | [4], [5], [6] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Afatinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Directional sequencing assay; Direct sequencing assay; Sanger sequencing assay | |||
Experiment for Drug Resistance |
Progression-free and post-progression survival asaay; Analysis of progression-free survival (PFS) assay; Overall survival assay | |||
Mechanism Description | The identification of T790M as acquired resistance mechanism was clinically feasible. Although T790M had no prognostic or predictive role in the present study, further research is necessary to identify patients with T790M-mutant tumors who might benefit from newly developed T790M-specific TkIs.T790M is likely a common resistance mechanism in patients treated with first-line afatinib. Although repeat biopsies at progression are crucial in elucidating resistance mechanisms, this study suggests that clinical and technical issues often limit their feasibility, highlighting the importance of developing. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Esophagogastric cancer | [7] | |||
Sensitive Disease | Esophagogastric cancer [ICD-11: 2B71.1] | |||
Sensitive Drug | Afatinib | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | In summary, we find that concurrent amplification of EGFR and ERBB2 is associated with response to the HER kinase inhibitor afatinib in patients with trastuzumab-refractory EG cancer. Heterogeneous uptake of 89Zr-trastuzumab measured noninvasively by PET was associated with disease progression. Analyses of multiple disease sites sampled at the time of disease progression indicated several potential mediators of afatinib resistance, including loss of EGFR amplification and gain of MET amplification. |
Brigatinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Disease Class: Solid tumour/cancer | [8] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Brigatinib | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HepG2 cells | Liver | Homo sapiens (Human) | CVCL_0027 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
SUP-M2 cells | Colon | Homo sapiens (Human) | CVCL_2209 | |
KARPAS-299 cells | Peripheral blood | Homo sapiens (Human) | CVCL_1324 | |
SU-DHL-1 cells | Pleural effusion | Homo sapiens (Human) | CVCL_0538 | |
L-82 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2098 | |
HCC78 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2061 | |
H838 cells | Lymph node | Homo sapiens (Human) | CVCL_1594 | |
H-4-II-E cells | Liver | Rattus norvegicus (Rat) | CVCL_0284 | |
H358 cells | Lung | Homo sapiens (Human) | CVCL_1559 | |
H2228 cells | Lung | Homo sapiens (Human) | CVCL_1543 | |
DEL cells | Pleural effusion | Homo sapiens (Human) | CVCL_1170 | |
In Vivo Model | SCID beige mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | Non-small cell lung cancers (NSCLCs) harboring ALK gene rearrangements (ALK+) typically become resistant to the first-generation anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor (TKI) crizotinib through development of secondary resistance mutations in ALK or disease progression in the brain. Mutations that confer resistance to second-generation ALK TKIs ceritinib and alectinib have also been identified. Brigatinib is the only TKI to maintain substantial activity against the most recalcitrant ALK resistance mutation, G1202R. The unique, potent, and pan-ALK mutant activity of brigatinib could be rationalized by structural analyses. | |||
Disease Class: Solid tumour/cancer | [8] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Brigatinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HepG2 cells | Liver | Homo sapiens (Human) | CVCL_0027 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
SUP-M2 cells | Colon | Homo sapiens (Human) | CVCL_2209 | |
KARPAS-299 cells | Peripheral blood | Homo sapiens (Human) | CVCL_1324 | |
SU-DHL-1 cells | Pleural effusion | Homo sapiens (Human) | CVCL_0538 | |
L-82 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2098 | |
HCC78 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2061 | |
H838 cells | Lymph node | Homo sapiens (Human) | CVCL_1594 | |
H-4-II-E cells | Liver | Rattus norvegicus (Rat) | CVCL_0284 | |
H358 cells | Lung | Homo sapiens (Human) | CVCL_1559 | |
H2228 cells | Lung | Homo sapiens (Human) | CVCL_1543 | |
DEL cells | Pleural effusion | Homo sapiens (Human) | CVCL_1170 | |
In Vivo Model | SCID beige mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | Non-small cell lung cancers (NSCLCs) harboring ALK gene rearrangements (ALK+) typically become resistant to the first-generation anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor (TKI) crizotinib through development of secondary resistance mutations in ALK or disease progression in the brain. Mutations that confer resistance to second-generation ALK TKIs ceritinib and alectinib have also been identified. Brigatinib is the only TKI to maintain substantial activity against the most recalcitrant ALK resistance mutation, G1202R. The unique, potent, and pan-ALK mutant activity of brigatinib could be rationalized by structural analyses. |
Cetuximab
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Colorectal cancer | [9] | |||
Resistant Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Resistant Drug | Cetuximab | |||
Molecule Alteration | Missense mutation | p.G465E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Colon cells | Colon | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy assay | |||
Mechanism Description | Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations. | |||
Disease Class: Metastatic colorectal cancer | [10] | |||
Resistant Disease | Metastatic colorectal cancer [ICD-11: 2D85.0] | |||
Resistant Drug | Cetuximab | |||
Molecule Alteration | Missense mutation | p.S492R |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Standard-of-care sequencing assay | |||
Mechanism Description | K-RAS and EGFR ectodomain-acquired mutations in patients with metastatic colorectal cancer (mCRC) have been correlated with acquired resistance to anti-EGFR monoclonal antibodies (mAbs). |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Colorectal cancer | [11] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | Cetuximab | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | SW480 cells | Colon | Homo sapiens (Human) | CVCL_0546 |
HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | microRNA-7 expression in colorectal cancer is associated with poor prognosis and regulates cetuximab sensitivity via EGFR regulation. |
Cisplatin
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Non-small cell lung cancer | [12] | |||
Sensitive Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Sensitive Drug | Cisplatin | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-21 mainly achieves drug resistance by inhibiting cisplatin-induced apoptosis, and its specific mechanisms include the following: (1) improving the expression level of EGFR and protecting the toxic effect of tumor cells during chemotherapy; (2) Increase the expression of LRP and decrease the effective concentration of the target drug through the barrier of drug transport between nucleus and cell; (3) Enhance the expression of multidrug resistance associated protein (MRP1) and assist in pumping chemotherapeutic drugs from the inside to the outside of the cell. |
Dacomitinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [13] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | Dacomitinib | |||
Molecule Alteration | IF-insertion | p.Y764_V765insHH (c.2292_2293insCATCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; SDS-PAGE assay | |||
Experiment for Drug Resistance |
MTS assay; Crystal violet staining assay | |||
Mechanism Description | Mutation in the covalent binding site of either EGFR or HER2 is sufficient to lead to drug resistance. | |||
Disease Class: Solid tumour/cancer | [13] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | Dacomitinib | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; SDS-PAGE assay | |||
Experiment for Drug Resistance |
MTS assay; Crystal violet staining assay | |||
Mechanism Description | Mutation in the covalent binding site of either EGFR or HER2 is sufficient to lead to drug resistance. | |||
Disease Class: Solid tumour/cancer | [13] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | Dacomitinib | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; SDS-PAGE assay | |||
Experiment for Drug Resistance |
MTS assay; Crystal violet staining assay | |||
Mechanism Description | Mutation in the covalent binding site of either EGFR or HER2 is sufficient to lead to drug resistance. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [13] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Complex-indel | p.D770_770delinsGY (c.2308_2310delinsGGTTAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; SDS-PAGE assay | |||
Experiment for Drug Resistance |
MTS assay; Crystal violet staining assay | |||
Mechanism Description | Mutation in the covalent binding site of either EGFR or HER2 is sufficient to lead to drug resistance. | |||
Disease Class: Lung adenocarcinoma | [13] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; SDS-PAGE assay | |||
Experiment for Drug Resistance |
MTS assay; Crystal violet staining assay | |||
Mechanism Description | Mutation in the covalent binding site of either EGFR or HER2 is sufficient to lead to drug resistance. | |||
Disease Class: Lung adenocarcinoma | [13] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | IF-insertion | p.P772_H773insPNP (c.2317_2318insCTAACCCTC) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; SDS-PAGE assay | |||
Experiment for Drug Resistance |
MTS assay; Crystal violet staining assay | |||
Mechanism Description | Mutation in the covalent binding site of either EGFR or HER2 is sufficient to lead to drug resistance. | |||
Disease Class: Lung adenocarcinoma | [14] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Solid tumour/cancer | [15] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Missense mutation | p.E709K (c.2125G>A) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
myelomonocyti cells | Bone marrow | Homo sapiens (Human) | N.A. | |
macrophage-like cells | N.A. | . | N.A. | |
IL3-dependent murine pro-B cells | Blood | Homo sapiens (Human) | N.A. | |
Balb/C mouse leukemia cells | Blood | Mus musculus (Mouse) | CVCL_9099 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay | |||
Disease Class: Solid tumour/cancer | [15] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Complex-indel | p.E709_T710delinsD (c.2127_2129delAAC) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
myelomonocyti cells | Bone marrow | Homo sapiens (Human) | N.A. | |
macrophage-like cells | N.A. | . | N.A. | |
IL3-dependent murine pro-B cells | Blood | Homo sapiens (Human) | N.A. | |
Balb/C mouse leukemia cells | Blood | Mus musculus (Mouse) | CVCL_9099 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay | |||
Disease Class: Lung adenocarcinoma | [16] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Missense mutation | p.G719S (c.2155G>A) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [16] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Missense mutation | p.G719C (c.2155G>T) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [16] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Missense mutation | p.G719A (c.2156G>C) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Solid tumour/cancer | [15] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | Missense mutation | p.G719A (c.2156G>C) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
myelomonocyti cells | Bone marrow | Homo sapiens (Human) | N.A. | |
macrophage-like cells | N.A. | . | N.A. | |
IL3-dependent murine pro-B cells | Blood | Homo sapiens (Human) | N.A. | |
Balb/C mouse leukemia cells | Blood | Mus musculus (Mouse) | CVCL_9099 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay | |||
Disease Class: Lung adenocarcinoma | [17] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Dacomitinib | |||
Molecule Alteration | IF-deletion | p.G729_D761 (c.2185_2283) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lung | . |
Dasatinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Dasatinib | |||
Molecule Alteration | Missense mutation | p.E711K |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Docetaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Non-small cell lung cancer | [19] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Docetaxel | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | The transfection of miR 27b mimics led to downregulation of the expression levels of EGFR, whilst miR 27b inhibitors upregulated the expression levels of EGFR. Furthermore, it was demonstrated that the transfection of miR 27b mimics significantly suppressed the apoptosis and promote the viability of A549 human lung carcinoma cells. In line with this, the introduction of miR 27b inhibitors significantly induced apoptosis and inhibited the proliferation of A549 cells. These results indicate that miR 27b may promote NSCLC cell viability and enhance resistance to docetaxel treatment through direct inhibition of EGFR expression. |
Doxorubicin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Prostate cancer | [20] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Resistant Drug | Doxorubicin | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | LncRNA LOXL1-AS1/miR-let-7a-5p/EGFR-related pathway regulates the doxorubicin resistance of prostate cancer DU-145 cells. |
Erlotinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [21], [22], [23] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall and disease-free assay | |||
Mechanism Description | Among patients with acquired resistance to EGFR TkIs, the presence of T790M defines a clinical subset with a relatively favorable prognosis and more indolent progression. | |||
Disease Class: Non-small cell lung cancer | [24], [25] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Computed tomography assay | |||
Mechanism Description | In addition, three (8%) patients acquired EGFR amplifications in their resistant specimens, all of which also acquired the classic T790M EGFR mutation. Moreover, in two cases with high-level EGFR amplification (>10-fold), it was clear by comparison of the peak heights on the SNaPshot chromatogram that the T790M allele was the amplified allele. They have identified several resistance mechanisms, two of which-EGFR mutation T790M and MET amplification have been validated in the clinic. | |||
Disease Class: Non-small cell lung cancer | [26] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.C797S+p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
EGFR/TKLS mediated apoptosis signaling pathway | Inhibition | hsa01521 | ||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy; ATP-binding pocket affinity comparison assay | |||
Mechanism Description | Several mechanisms of resistance have been described to EGFR-TkIs, such as the occurrence of secondary mutation (T790M, C797S), the activation of alternative signalling (Met, HGF, AXL, Hh, IGF-1R), the aberrance of the downstream pathways (AkT mutations, loss of PTEN), the impairment of the EGFR-TkIs-mediated apoptosis pathway (BCL2-like 11/BIM deletion polymorphism) and histological transformation. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [27] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.D761Y |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired resistance can occur through failure of drug delivery to the target, as in isolated central nervous system (CNS) progression, or by selection of biological variants during TkI exposure. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [27] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.L747S |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired resistance can occur through failure of drug delivery to the target, as in isolated central nervous system (CNS) progression, or by selection of biological variants during TkI exposure. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [27] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired resistance can occur through failure of drug delivery to the target, as in isolated central nervous system (CNS) progression, or by selection of biological variants during TkI exposure. At the point of acquired resistance, the T790M substitution may be accompanied by amplification of the EGFR gene as well. | |||
Disease Class: Non-small cell lung cancer | [28] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Next generation sequencing assay | |||
Experiment for Drug Resistance |
Multivariate analysis of overall or disease-free survival assay | |||
Mechanism Description | One example is the acquisition of the T790M substitution in the membrane receptor EGFR conferring resistance to gefitinb and erlotinib in lung cancer in approximately 50% of patients. | |||
Disease Class: Non-small cell lung cancer | [29] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Erlotinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Wnt signaling pathway | Activation | hsa04310 | |
mTOR signaling pathway | Activation | hsa04150 | ||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H2170 cells | Lung | Homo sapiens (Human) | CVCL_1535 | |
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
MTT cell viability assay | |||
Mechanism Description | H1975 cells are positive for the T790M EGFR mutation, which confers resistance to current EGFR TkI therapies. |
Gefitinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Non-small cell lung cancer | [26] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.C797S |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK/MAPKsignaling pathway | Activation | hsa04210 | |
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy; ATP-binding pocket affinity comparison assay | |||
Mechanism Description | Known mechanisms are secondary resistance mutations occurring in the ATP-binding domain (such as T790M and C797S), mutation or amplification of bypass signallings (such as AXL, Hh, ERBb2, CRIPTO, etc), activating mutations in the downstream pathways (PI3k, AkT, MEk, RAF), low levels of mRNA or polymorphisms of the pro-apoptotic protein BIM, induction of a transcription programme for EMT and phenotypical changes, or induction of elevated tumour PD-L1 levels. | |||
Disease Class: Non-small cell lung cancer | [30], [31], [32] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK/MAPKsignaling pathway | Activation | hsa04210 | |
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy; ATP-binding pocket affinity comparison assay | |||
Mechanism Description | Known mechanisms are secondary resistance mutations occurring in the ATP-binding domain (such as T790M and C797S), mutation or amplification of bypass signallings (such as AXL, Hh, ERBb2, CRIPTO, etc), activating mutations in the downstream pathways (PI3k, AkT, MEk, RAF), low levels of mRNA or polymorphisms of the pro-apoptotic protein BIM, induction of a transcription programme for EMT and phenotypical changes, or induction of elevated tumour PD-L1 levels. | |||
Disease Class: Lung adenocarcinoma | [33], [34], [35] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall and disease-free assay | |||
Mechanism Description | A secondary T790M mutation of EGFR accounted for half the tumors with acquired resistance to gefitinib in Japanese patients. Other drug-resistant secondary mutations are uncommon in the EGFR gene. | |||
Disease Class: Lung squamous cell carcinoma | [36], [37], [38] | |||
Resistant Disease | Lung squamous cell carcinoma [ICD-11: 2C25.3] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Computed tomography (CT) scanning assay; Bone scintigraphy assay; Magnetic resonance imaging assay | |||
Mechanism Description | C-Met amplification, epithelial-mesenchymal transition, and kRAS and BRAF mutations were ruled out as alternative resistance mechanisms in the T790M-negative lung rebiopsy, suggesting that alternative oncogene aberrations such as HER2/Neu amplification, hepatocyte growth factor release by the tumor microenvironment, or other unidentified pathways contributed to the TkI resistance that was observed in the primary lesion. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [39] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
MGB SNP detection kit assay; Mutation Detection assay | |||
Experiment for Drug Resistance |
Digital PCR assay | |||
Mechanism Description | Resistance mechanisms to EGFR-TkI therapy in EGFR-mutated NSCLC include secondary EGFR T790M mutation, c-Met amplification, PIk3CA mutation, and transformation to small-cell lung cancer. | |||
Disease Class: Non-small cell lung cancer | [26] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.C797S+p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
EGFR/TKLS mediated apoptosis signaling pathway | Inhibition | hsa01521 | ||
Epithelial mesenchymal transition signaling pathway | Activation | hsa01521 | ||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy; ATP-binding pocket affinity comparison assay | |||
Mechanism Description | Several mechanisms of resistance have been described to EGFR-TkIs, such as the occurrence of secondary mutation (T790M, C797S), the activation of alternative signalling (Met, HGF, AXL, Hh, IGF-1R), the aberrance of the downstream pathways (AkT mutations, loss of PTEN), the impairment of the EGFR-TkIs-mediated apoptosis pathway (BCL2-like 11/BIM deletion polymorphism) and histological transformation. | |||
Disease Class: Lung adenocarcinoma | [40] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.D761Y |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct Sanger sequencing analysis | |||
Experiment for Drug Resistance |
CellTiter-Blue cell viability assay | |||
Mechanism Description | The T790M mutation is common in patients with acquired resistance. The limited spectrum of TkI-resistant mutations in EGFR, which binds to erlotinib in the active conformation, contrasts with a wider range of second-site mutations seen with acquired resistance to imatinib, which binds to ABL and kIT, respectively, in closed conformations. Collectively, our data suggest that the type and nature of kinase inhibitor resistance mutations may be influenced by both anatomic site and mode of binding to the kinase target. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [27] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.L747S |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired resistance can occur through failure of drug delivery to the target, as in isolated central nervous system (CNS) progression, or by selection of biological variants during TkI exposure. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [27] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.D761Y |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired resistance can occur through failure of drug delivery to the target, as in isolated central nervous system (CNS) progression, or by selection of biological variants during TkI exposure. | |||
Disease Class: EGFR-mutant non-small cell lung cancer | [27] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired resistance can occur through failure of drug delivery to the target, as in isolated central nervous system (CNS) progression, or by selection of biological variants during TkI exposure. At the point of acquired resistance, the T790M substitution may be accompanied by amplification of the EGFR gene as well. | |||
Disease Class: Non-small cell lung cancer | [28] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Next generation sequencing assay | |||
Experiment for Drug Resistance |
Multivariate analysis of overall or disease-free survival assay | |||
Mechanism Description | One example is the acquisition of the T790M substitution in the membrane receptor EGFR conferring resistance to gefitinb and erlotinib in lung cancer in approximately 50% of patients. | |||
Disease Class: Non-small cell lung cancer | [33] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Cos-7 cells | Lung | Homo sapiens (Human) | CVCL_0224 |
NIH-3T3 cells | Embryo | Mus musculus (Mouse) | CVCL_0594 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Mechanism Description | The DNA sequence of the EGFR gene in his tumor biopsy specimen at relapse revealed the presence of a second point mutation, resulting in threonine-to-methionine amino acid change at position 790 of EGFR. Structural modeling and biochemical studies showed that this second mutation led to gefitinib resistance. | |||
Disease Class: Non-small cell lung cancer | [33] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Cos-7 cells | Lung | Homo sapiens (Human) | CVCL_0224 |
NIH-3T3 cells | Embryo | Mus musculus (Mouse) | CVCL_0594 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Mechanism Description | The DNA sequence of the EGFR gene in his tumor biopsy specimen at relapse revealed the presence of a second point mutation, resulting in threonine-to-methionine amino acid change at position 790 of EGFR. Structural modeling and biochemical studies showed that this second mutation led to gefitinib resistance. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Non-small cell lung cancer | [41] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
H1299 cells | Lung | Homo sapiens (Human) | CVCL_0060 | |
Sk-MES-1 cells | Lung | Homo sapiens (Human) | CVCL_0630 | |
In Vivo Model | Tumor xenograft in vivo model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Long Noncoding RNA LINC00460 promotes the gefitinib resistance of nonsmall cell lung cancer through EGFR by sponging miR-769-5p. | |||
Disease Class: Non-small cell lung cancer | [42] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell colony | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-138 inhibit the protein level of EGFR and reverses gefitinib resistance in lung cancer cells. | |||
Disease Class: Lung cancer | [43] | |||
Resistant Disease | Lung cancer [ICD-11: 2C25.5] | |||
Resistant Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Non-small cell lung cancer | [26] | |||
Sensitive Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Sensitive Drug | Gefitinib | |||
Molecule Alteration | Missense mutation | p.L858R |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK/MAPKsignaling pathway | Activation | hsa04210 | |
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy; ATP-binding pocket affinity comparison assay | |||
Mechanism Description | The two most common EGFR-activating mutations are small in-frame deletions in exon 19 (particularly E746-A750del) and amino acid substitution in exon 21 (leucine to arginine at codon 858 (L858R)), which collectively account for >90% of known activating EGFR mutations.2 3 These two alterations are the best-characterised mutations conferring sensitivity to EGFR-tyrosine kinase inhibitor (EGFR-TkI) therapy, resulting in higher response rates (RR) (up to 70%) and longer median survival (up to 24-30 months) than those observed in patients with wild-type (WT) EGFR. The higher sensitivity of these mutations relays in an increased affinity of the ATP-binding pocket for EGFR-TkIs as compared with WT EGFR. | |||
Disease Class: Non-small cell lung cancer | [26] | |||
Sensitive Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Sensitive Drug | Gefitinib | |||
Molecule Alteration | Frameshift mutation | p.E746-A750del |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK/MAPKsignaling pathway | Activation | hsa04210 | |
In Vitro Model | NSCLC cells | Lung | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy; ATP-binding pocket affinity comparison assay | |||
Mechanism Description | The two most common EGFR-activating mutations are small in-frame deletions in exon 19 (particularly E746-A750del) and amino acid substitution in exon 21 (leucine to arginine at codon 858 (L858R)), which collectively account for >90% of known activating EGFR mutations.2 3 These two alterations are the best-characterised mutations conferring sensitivity to EGFR-tyrosine kinase inhibitor (EGFR-TkI) therapy, resulting in higher response rates (RR) (up to 70%) and longer median survival (up to 24-30 months) than those observed in patients with wild-type (WT) EGFR. The higher sensitivity of these mutations relays in an increased affinity of the ATP-binding pocket for EGFR-TkIs as compared with WT EGFR. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Non-small cell lung cancer | [44] | |||
Sensitive Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Sensitive Drug | Gefitinib | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
CCD-19Lu cells | Lung | Homo sapiens (Human) | CVCL_2382 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
MRC-5 cells | Lung | Homo sapiens (Human) | CVCL_0440 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | microRNA-200a directly targets and downregulates egfr and c-met to inhibit migration, invasion, and gefitinib resistance in non-small cell lung cancer. | |||
Disease Class: Lung adenocarcinoma | [45] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Gefitinib | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
EGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 | |
H1299 cells | Lung | Homo sapiens (Human) | CVCL_0060 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
16HBE cells | Lung | Homo sapiens (Human) | CVCL_0112 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; EdU assay | |||
Mechanism Description | GAS5 was significantly downregulated in lung adenocarcinoma tissues compared with the paired adjacent non-tumorous tissue samples. Furthermore, lower GAS5 expression levels were associated with larger tumor sizes, poor tumor differentiation, and advanced pathological stages. However, GAS5 was almost equally expressed between benign tumors compared with the adjacent normal tissues. GAS5 was also overexpressed in EGFR-TkI sensitive cell lines compared with the resistant cell line. Using MTT, EdU incorporation, and colony formation assays, we showed that GAS5-expressing A549 cells displayed an elevated level of cell death. In addition to its pro-apoptotic effect in the A549 cell line, GAS5 overexpression also suppressed the growth of A549-derived tumors in nude mice treated with gefitinib. GAS5 overexpression was inversely correlated with the expression of the EGFR pathway and IGF-1R proteins. | |||
Disease Class: Lung adenocarcinoma | [46] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Gefitinib | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
RGFR signaling pathway | Inhibition | hsa05200 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | EGFR was negatively regulated by miR-7 mimic transfection, and downregulation of EGFR expression at the protein level largely correlated with elevated levels of miR-7 in the gefitinib-resistant cells. The results of the present study suggest that miR-7 may have central roles in the development of resistance to endocrine therapy in resistant cells through regulating the expression of EGFR in cancer cells. | |||
Disease Class: Non-small cell lung cancer | [47] | |||
Sensitive Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Sensitive Drug | Gefitinib | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
EGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
NCI-H1650 cells | Lung | Homo sapiens (Human) | CVCL_1483 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-133b suppresses the expression of EGFR, miR-133b transfection may modulate apoptosis, invasion and sensitivity to EGFR-TkI through the EGFR signaling pathways. |
Icotinib hydrochloride
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Non-small cell lung cancer | [4], [5], [6] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Icotinib hydrochloride | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Directional sequencing assay; Direct sequencing assay | |||
Experiment for Drug Resistance |
Progression-free and post-progression survival asaay | |||
Mechanism Description | The identification of T790M as acquired resistance mechanism was clinically feasible. Although T790M had no prognostic or predictive role in the present study, further research is necessary to identify patients with T790M-mutant tumors who might benefit from newly developed T790M-specific TkIs. |
Imatinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Imatinib | |||
Molecule Alteration | Missense mutation | p.E711K |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Lapatinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.V292E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.R705G |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.L760F |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.K284E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.I706T |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.G696E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.A822V |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.V292M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.P741S |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.G288D |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Lapatinib | |||
Molecule Alteration | Missense mutation | p.E711K |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Nilotinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Nilotinib | |||
Molecule Alteration | Missense mutation | p.E711K |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Olmutinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [48] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Olmutinib | |||
Molecule Alteration | Missense mutation | p.C797S |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Computed tomography assay; Progression-free survival assay | |||
Mechanism Description | A recent preclinical study reported that EGFR C797S, L718Q, and L844V mutations cause resistance to both WZ4002 and CO-1686 but only EGFR C797S leads to AZD9291 resistance, suggesting that C797S can serve as acquired resistance to irreversible pyrimidine-based EGFR inhibitors given the similar chemical structure of third-generation EGFR TkIs. |
Osimertinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Non-small cell lung cancer | [49] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Osimertinib | |||
Molecule Alteration | Missense mutation | p.C797S |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Cell-free plasma DNA assay; Next generation assay; Droplet digital PCR assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Acquired EGFR C797S mediates resistance to AZD9291 in advanced non-small cell lung cancer harboring EGFR T790M. | |||
Disease Class: Non-small cell lung cancer | [50] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Osimertinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Sanger sequencing assay; Fluorescence in situ hybridization assay; Real-time polymerase chain reaction assay; Targeted exome sequencing assay | |||
Experiment for Drug Resistance |
Computed tomography assay | |||
Mechanism Description | Acquired resistance mechanisms of AZD9291 in patients with EGFRT790M-mutant NSCLC who failed treatment with first-generation EGFR TkIs include the loss of EGFRT790M-mutant clones plus alternative pathway activation or histologic transformation and EGFR ligand-dependent activation. |
Paclitaxel
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Non-small cell lung cancer | [51] | |||
Sensitive Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Sensitive Drug | Paclitaxel | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
95D cells | Lung | Homo sapiens (Human) | CVCL_7110 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | Overexpression of miR-7 sensitizes NSCLC cells to PTX and inhibites EGFR expression in A549 cells. |
Panitumumab
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Colorectal cancer | [9] | |||
Resistant Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Resistant Drug | Panitumumab | |||
Molecule Alteration | Missense mutation | p.G465E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Colon cells | Colon | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay | |||
Experiment for Drug Resistance |
Liquid biopsy assay | |||
Mechanism Description | Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations. | |||
Disease Class: Metastatic colorectal cancer | [10] | |||
Resistant Disease | Metastatic colorectal cancer [ICD-11: 2D85.0] | |||
Resistant Drug | Panitumumab | |||
Molecule Alteration | Missense mutation | p.S492R |
||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Standard-of-care sequencing assay | |||
Mechanism Description | K-RAS and EGFR ectodomain-acquired mutations in patients with metastatic colorectal cancer (mCRC) have been correlated with acquired resistance to anti-EGFR monoclonal antibodies (mAbs). | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Metastatic colorectal cancer | [52] | |||
Resistant Disease | Metastatic colorectal cancer [ICD-11: 2D85.0] | |||
Resistant Drug | Panitumumab | |||
Molecule Alteration | Expression | Up-regulation |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
EGFR signaling pathway | Activation | hsa01521 | ||
ERBB2/MET/IGF-1R signalling pathway | Activation | hsa04520 | ||
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
PI3K/AKT/mTOR signaling pathway | Activation | hsa04151 | ||
Experiment for Drug Resistance |
Progression-free survival (PFS) analysis; Overall survival (OS) analysis | |||
Mechanism Description | Cetuximab and panitumumab are monoclonal antibodies (mAbs) against epidermal growth factor receptor (EGFR) that are effective agents for metastatic colorectal cancer (mCRC). The mechanisms of resistance refer to intrinsic and extrinsic alterations of tumours. The intrinsic mechanisms include EGFR ligand overexpression, EGFR alteration, RAS/RAF/PI3K gene mutations, ERBB2/MET/IGF-1R activation, metabolic remodelling, microsatellite instability and autophagy. Extrinsic alterations mainly disrupt the tumour microenvironment, specifically immune cells, cancer-associated fibroblasts (CAFs) and angiogenesis. |
Pemetrexed
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Non-small cell lung cancer | [39] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Pemetrexed | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
MGB SNP detection kit assay; Mutation Detection assay | |||
Experiment for Drug Resistance |
Digital PCR assay | |||
Mechanism Description | Resistance mechanisms to EGFR-TkI therapy in EGFR-mutated NSCLC include secondary EGFR T790M mutation, c-Met amplification, PIk3CA mutation, and transformation to small-cell lung cancer. |
Pertuzumab
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.V292E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.R705G |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.L760F |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.K284E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.I706T |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.G696E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.A822V |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.V292M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.P741S |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.G288D |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Pertuzumab | |||
Molecule Alteration | Missense mutation | p.E711K |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Temozolomide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Glioblastoma multiforme | [53] | |||
Sensitive Disease | Glioblastoma multiforme [ICD-11: 2A00.03] | |||
Sensitive Drug | Temozolomide | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | U251 cells | Brain | Homo sapiens (Human) | CVCL_0021 |
U87 cells | Brain | Homo sapiens (Human) | CVCL_0022 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; Luciferase reporter assay | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | miR181b modulates chemosensitivity of glioblastoma multiforme cells to temozolomide by targeting the epidermal growth factor receptor. |
Trastuzumab
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.V292E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.I706T |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.R705G |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.L760F |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.K284E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.G696E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.A822V |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.V292M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.P741S |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.G288D |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab | |||
Molecule Alteration | Missense mutation | p.E711K |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Breast cancer | [54] | |||
Sensitive Disease | Breast cancer [ICD-11: 2C60.3] | |||
Sensitive Drug | Trastuzumab | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | MCF-7 cells | Breast | Homo sapiens (Human) | CVCL_0031 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Colcount colony counter assay | |||
Mechanism Description | miR-7 suppression of HER2deta16 oncogenic activity is mediated through inactivation of Src kinase and suppression of EGFR expression implies that targeting these pathways would also suppress HER2deta16 tumorigenesis. HER2deta16 suppresses expression of the miR-7 tumor suppressor and reestablished miR-7 expression significantly inhibits HER2deta16 mediated tumor cell proliferation and migration and miR-7 sensitizes HER2deta16 expressing cells to trastuzumab treatment. |
Trastuzumab emtansine
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.V292E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.R705G |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.L760F |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.K284E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.I706T |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.G696E |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.A822V |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.V292M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.P741S |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. | |||
Disease Class: HER2 positive breast cancer | [18] | |||
Resistant Disease | HER2 positive breast cancer [ICD-11: 2C60.8] | |||
Resistant Drug | Trastuzumab emtansine | |||
Molecule Alteration | Missense mutation | p.G288D |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Plasma | Blood | Homo sapiens (Human) | N.A. |
Experiment for Molecule Alteration |
Next-generation sequencing assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Positron emission tomography/Computed tomography assay | |||
Mechanism Description | Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred mutations in the resistant group were associated with the resistance of targeted therapy. |
Vandetanib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [55] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Vandetanib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
H1650 cells | Pleural effusion | Homo sapiens (Human) | CVCL_4V01 | |
Calu-6 cells | Lung | Homo sapiens (Human) | CVCL_0236 | |
In Vivo Model | Mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The missense mutation p.L858R (c.2573T>G) in gene EGFR cause the sensitivity of Vandetanib by aberration of the drug's therapeutic target | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [56] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Vandetanib | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | LOVO cells | Colon | Homo sapiens (Human) | CVCL_0399 |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
PC-14 cells | Lung | Homo sapiens (Human) | CVCL_1640 | |
WiDR cells | Colon | Homo sapiens (Human) | CVCL_2760 | |
SkBR3 cells | Breast | Homo sapiens (Human) | CVCL_0033 | |
SkOV3 cells | Ovary | Homo sapiens (Human) | CVCL_0532 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The if-deletion p.E746_A750delELREA (c.2236_2250del15) in gene EGFR cause the sensitivity of Vandetanib by unusual activation of pro-survival pathway. |
Vemurafenib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Melanoma | [57] | |||
Sensitive Disease | Melanoma [ICD-11: 2C30.0] | |||
Sensitive Drug | Vemurafenib | |||
Molecule Alteration | Expression | Down-regulation |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
MAPK/PI3K/AKT signaling pathway | Inhibition | hsa05235 | ||
In Vitro Model | A375 cells | Skin | Homo sapiens (Human) | CVCL_0132 |
Mel-CV cells | Skin | Homo sapiens (Human) | CVCL_S996 | |
Experiment for Molecule Alteration |
Immunohistochemical staining assay; Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-7 expression was decreased in both VemR A375 and Mel-CVR melanoma cells and its low expression contributed to BRAFi resistance. Furthermore, by decreasing the expression levels of EGFR, IGF-1R and CRAF, miR-7 could inhibit the activation of RAS/RAF/MEk/ERk (MAPk) and PI3k/AkT pathway and partially reverse the resistance to BRAFi in VemR A375 melanoma cells. |
Patented Agent(s)
1 drug(s) in total
Abivertinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [58] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Abivertinib | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Hela cells | Cervix uteri | Homo sapiens (Human) | CVCL_0030 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
BEAS-2B cells | Bronchus | Homo sapiens (Human) | CVCL_0168 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
HT1080 cells | Acetabulum | Homo sapiens (Human) | CVCL_0317 | |
In Vivo Model | Nu/Nu nude mouse xenograft model | Mus musculus | ||
Experiment for Drug Resistance |
WST-1 assay | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [58] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Abivertinib | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Hela cells | Cervix uteri | Homo sapiens (Human) | CVCL_0030 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
BEAS-2B cells | Bronchus | Homo sapiens (Human) | CVCL_0168 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
HT1080 cells | Acetabulum | Homo sapiens (Human) | CVCL_0317 | |
In Vivo Model | Nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Immunoblotting analysis; ELISA assay | |||
Experiment for Drug Resistance |
WST-1 assay |
Clinical Trial Drug(s)
10 drug(s) in total
Nazartinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Disease Class: Lung adenocarcinoma | [59] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Nazartinib | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
HaCaT cells | Tongue | Homo sapiens (Human) | CVCL_0038 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
In Vivo Model | NSG mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGF816 Exerts Anticancer Effects in Non-Small Cell Lung Cancer by Irreversibly and Selectively Targeting Primary and Acquired Activating Mutations in the EGF Receptor | |||
Disease Class: Solid tumour/cancer | [59] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Nazartinib | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
HaCaT cells | Tongue | Homo sapiens (Human) | CVCL_0038 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
In Vivo Model | NSG mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGF816 Exerts Anticancer Effects in Non-Small Cell Lung Cancer by Irreversibly and Selectively Targeting Primary and Acquired Activating Mutations in the EGF Receptor | |||
Disease Class: Solid tumour/cancer | [59] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Nazartinib | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
HaCaT cells | Tongue | Homo sapiens (Human) | CVCL_0038 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
In Vivo Model | NSG mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGF816 Exerts Anticancer Effects in Non-Small Cell Lung Cancer by Irreversibly and Selectively Targeting Primary and Acquired Activating Mutations in the EGF Receptor |
Selumetinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [60] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Selumetinib | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
In Vivo Model | BALB/C nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; Immunohistochemistry assay | |||
Experiment for Drug Resistance |
MTT assay |
AUY922
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Disease Class: Solid tumour/cancer | [61] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGFR exon 20 insertion mutants associate with the heat shock protein 90 (Hsp90) chaperone system. The non-geldanamycin Hsp90 inhibitor luminespib (formerly AUY922) degrades EGFR exon 20 mutations, downstream targets and induces apoptosis. | |||
Disease Class: Solid tumour/cancer | [61] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.Y764_V765insHH (c.2292_2293insCATCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGFR exon 20 insertion mutants associate with the heat shock protein 90 (Hsp90) chaperone system. The non-geldanamycin Hsp90 inhibitor luminespib (formerly AUY922) degrades EGFR exon 20 mutations, downstream targets and induces apoptosis. | |||
Disease Class: Solid tumour/cancer | [61] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGFR exon 20 insertion mutants associate with the heat shock protein 90 (Hsp90) chaperone system. The non-geldanamycin Hsp90 inhibitor luminespib (formerly AUY922) degrades EGFR exon 20 mutations, downstream targets and induces apoptosis. | |||
Disease Class: Solid tumour/cancer | [61] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.P772_H773insGNP (c.2316_2317insGGTAACCCT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGFR exon 20 insertion mutants associate with the heat shock protein 90 (Hsp90) chaperone system. The non-geldanamycin Hsp90 inhibitor luminespib (formerly AUY922) degrades EGFR exon 20 mutations, downstream targets and induces apoptosis. | |||
Disease Class: Solid tumour/cancer | [61] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.H773 (c.2317_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | EGFR exon 20 insertion mutants associate with the heat shock protein 90 (Hsp90) chaperone system. The non-geldanamycin Hsp90 inhibitor luminespib (formerly AUY922) degrades EGFR exon 20 mutations, downstream targets and induces apoptosis. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [61] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
NCI-H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter Glo assay; IC50 assay | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.A767_V769dupASV (c.2308_2309insCAAGCGTAG) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.D770_P772 (c.2308_2316) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.D770_N771insGV (c.2310_2311insGGTGTA) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.D770_N771insGF (c.2310_2311insGGTTTT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.S768_D770dupSVD (c.2311_2312insGCGTAGACA) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.P772_H773 (c.2314_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.P772_H773insYNP (c.2315_2316insTTATAACCC) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | Duplication | p.H773 (c.2317_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.H773_V774insAH (c.2320_2321insCACATG) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.A767_V774ins |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.V769_D770insASV |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.A770_N771insNPY |
||
Experimental Note | Identified from the Human Clinical Data | |||
Disease Class: Lung adenocarcinoma | [62] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AUY922 | |||
Molecule Alteration | IF-insertion | p.D770_N771insSVD |
||
Experimental Note | Identified from the Human Clinical Data |
Naquotinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Disease Class: Solid tumour/cancer | [63] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | Naquotinib | |||
Molecule Alteration | IF-insertion | p.Y764_V765insHH (c.2292_2293insCATCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Disease Class: Solid tumour/cancer | [63] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | Complex-indel | p.L747_P753delinsS (c.2240_2257del18) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Solid tumour/cancer | [63] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Solid tumour/cancer | [63] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Solid tumour/cancer | [63] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | IF-insertion | p.P772_H773insGNP (c.2316_2317insGGTAACCCT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Solid tumour/cancer | [63] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Lung adenocarcinoma | [63] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Lung adenocarcinoma | [63] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. | |||
Disease Class: Lung adenocarcinoma | [63] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Naquotinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9ER cells | N.A. | . | N.A. | |
BID007 cells | Pleural effusion | Homo sapiens (Human) | CVCL_W890 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
MTS assay; FACS assay | |||
Mechanism Description | There is a mechanism for resistance associated with EGFR Y764_V765insHH mutation, one of the most resistant mutations. We demonstrated that in Y764_V765insHH, histidine residues inserted in the Val765 and Met766 positions upregulated the EGFR kinase activity and caused steric insensitivity to the particular EGFR-TKIs. |
Poziotinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [64] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The if-insertion p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Solid tumour/cancer | [64] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.A767_V769 (c.2299_2307) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Solid tumour/cancer | [64] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.S768_D770 (c.2302_2310) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Solid tumour/cancer | [64] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.N771_H773 (c.2311_2319) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Solid tumour/cancer | [64] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | IF-insertion | p.P772_H773insGNP (c.2316_2317insGGTAACCCT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The if-insertion p.P772_H773insGNP (c.2316_2317insGGTAACCCT) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.A767_V769 (c.2299_2307) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Complex-indel | p.N771_771delinsFH (c.2311_2313delinsTTTCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The complex-indel p.N771_771delinsFH (c.2311_2313delinsTTTCAT) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | IF-insertion | p.P772_H773insGNP (c.2316_2317insGGTAACCCT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The if-insertion p.P772_H773insGNP (c.2316_2317insGGTAACCCT) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.A767_V769 (c.2299_2307) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.S768_D770 (c.2302_2310) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Complex-indel | p.D770_770delinsGY (c.2308_2310delinsGGTTAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The complex-indel p.D770_770delinsGY (c.2308_2310delinsGGTTAT) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.D770_P772 (c.2308_2316) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.D770_P772 (c.2308_2316) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | IF-insertion | p.D770_N771insG (c.2310_2311insGGT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The if-insertion p.D770_N771insG (c.2310_2311insGGT) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The duplication p.N771_H773 (c.2311_2319) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target. | |||
Disease Class: Lung adenocarcinoma | [64] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Poziotinib | |||
Molecule Alteration | Missense mutation | p.S768I (c.2303G>T) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
CUTO14 cells | N.A. | . | N.A. | |
In Vivo Model | Female nu/nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay | |||
Mechanism Description | The missense mutation p.S768I (c.2303G>T) in gene EGFR cause the sensitivity of Poziotinib by aberration of the drug's therapeutic target |
Tesevatinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [65] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Tesevatinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Computed tomography assay | |||
Mechanism Description | The most common mechanism of acquired resistance is caused by the development of the T790M mutation in exon 20 of EGFR in nearly 50% of patients, whereas 10% of the patients have amplification of the oncogene MET as a means of resistance. |
AEE-788
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [66] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | AEE-788 | |||
Molecule Alteration | Missense mutation | p.N826S (c.2477A>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter96 Proliferation assay | |||
Mechanism Description | The missense mutation p.N826S (c.2477A>G) in gene EGFR cause the resistance of AEE-788 by aberration of the drug's therapeutic target | |||
Disease Class: Lung adenocarcinoma | [67] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | AEE-788 | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Experiment for Molecule Alteration |
Enzyme kinetic assays | |||
Mechanism Description | The missense mutation p.T790M (c.2369C>T) in gene EGFR cause the resistance of AEE-788 by aberration of the drug's therapeutic target |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [66] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AEE-788 | |||
Molecule Alteration | Complex-indel | p.L747_P753delinsS (c.2240_2257del18) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter96 Proliferation assay | |||
Mechanism Description | The complex-indel p.L747_P753delinsS (c.2240_2257del18) in gene EGFR cause the sensitivity of AEE-788 by aberration of the drug's therapeutic target. | |||
Disease Class: Solid tumour/cancer | [66] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AEE-788 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter96 Proliferation assay | |||
Mechanism Description | The missense mutation p.L858R (c.2573T>G) in gene EGFR cause the sensitivity of AEE-788 by aberration of the drug's therapeutic target |
Lifirafenib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [68] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Lifirafenib | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HT29 Cells | Colon | Homo sapiens (Human) | CVCL_A8EZ |
SW620 cells | Colon | Homo sapiens (Human) | CVCL_0547 | |
HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 | |
A375 cells | Skin | Homo sapiens (Human) | CVCL_0132 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
COLO205 cells | Colon | Homo sapiens (Human) | CVCL_F402 | |
WiDR cells | Colon | Homo sapiens (Human) | CVCL_2760 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
SkMEL28 cells | Skin | Homo sapiens (Human) | CVCL_0526 | |
In Vivo Model | Female NOD/SCID and BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter-Glo assay; Tumor volume measurement assay | |||
Disease Class: Solid tumour/cancer | [68] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Lifirafenib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HT29 Cells | Colon | Homo sapiens (Human) | CVCL_A8EZ |
SW620 cells | Colon | Homo sapiens (Human) | CVCL_0547 | |
HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 | |
A375 cells | Skin | Homo sapiens (Human) | CVCL_0132 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 | |
COLO205 cells | Colon | Homo sapiens (Human) | CVCL_F402 | |
WiDR cells | Colon | Homo sapiens (Human) | CVCL_2760 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
SkMEL28 cells | Skin | Homo sapiens (Human) | CVCL_0526 | |
In Vivo Model | Female NOD/SCID and BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter-Glo assay; Tumor volume measurement assay |
CUDC-101
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [69] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | CUDC-101 | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vivo Model | Female athymic mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Perkin-Elmer ATPlite assay | |||
Mechanism Description | The missense mutation p.T790M (c.2369C>T) in gene EGFR cause the sensitivity of CUDC-101 by aberration of the drug's therapeutic target |
Lazertinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [70] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Lazertinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9GR cells | Lung | Homo sapiens (Human) | CVCL_V337 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
H2291 cells | Lung | Homo sapiens (Human) | CVCL_1546 | |
In Vivo Model | Female BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Experiment for Drug Resistance |
Cell-free kinase assay; IC50 assay | |||
Disease Class: Lung adenocarcinoma | [70] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Lazertinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9GR cells | Lung | Homo sapiens (Human) | CVCL_V337 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
H2291 cells | Lung | Homo sapiens (Human) | CVCL_1546 | |
In Vivo Model | Female BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Experiment for Drug Resistance |
Cell-free kinase assay; IC50 assay | |||
Disease Class: Lung adenocarcinoma | [70] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Lazertinib | |||
Molecule Alteration | Missense mutation | p.L861Q (c.2582T>A) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9GR cells | Lung | Homo sapiens (Human) | CVCL_V337 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
H2291 cells | Lung | Homo sapiens (Human) | CVCL_1546 | |
In Vivo Model | Female BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Experiment for Drug Resistance |
Cell-free kinase assay; IC50 assay |
Discontinued Drug(s)
2 drug(s) in total
Rociletinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Non-small cell lung cancer | [39], [71], [72] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Rociletinib | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/mTOR signaling pathway | Activation | hsa04151 | |
In Vitro Model | PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 |
Experiment for Molecule Alteration |
Blood-based tumor genotyping assay; Liquid Biopsies assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Overall and disease-free assay | |||
Mechanism Description | Similarly,resistance to the third-generation inhibitor rociletinib may not only be mediated by EGFR (L798I, C797S) mutations, but also by alterations of MET, PIk3CA, ERRB2, and kRAS, and by the negative selection of T790M-mutant subclones. | |||
Disease Class: Non-small cell lung cancer | [71], [72] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Rociletinib | |||
Molecule Alteration | Missense mutation | p.L798I |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/mTOR signaling pathway | Activation | hsa04151 | |
In Vitro Model | PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 |
Experiment for Molecule Alteration |
Blood-based tumor genotyping assay; Liquid Biopsies assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Overall and disease-free assay | |||
Mechanism Description | Similarly,resistance to the third-generation inhibitor rociletinib may not only be mediated by EGFR (L798I, C797S) mutations, but also by alterations of MET, PIk3CA, ERRB2, and kRAS, and by the negative selection of T790M-mutant subclones. | |||
Disease Class: Non-small cell lung cancer | [71], [72] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Rociletinib | |||
Molecule Alteration | Missense mutation | p.C797S |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/mTOR signaling pathway | Activation | hsa04151 | |
In Vitro Model | PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 |
Experiment for Molecule Alteration |
Blood-based tumor genotyping assay; Liquid Biopsies assay; Circulating-free DNA assay | |||
Experiment for Drug Resistance |
Overall and disease-free assay | |||
Mechanism Description | Similarly,resistance to the third-generation inhibitor rociletinib may not only be mediated by EGFR (L798I, C797S) mutations, but also by alterations of MET, PIk3CA, ERRB2, and kRAS, and by the negative selection of T790M-mutant subclones. |
S-1
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: EGFR-mutant non-small cell lung cancer | [39] | |||
Resistant Disease | EGFR-mutant non-small cell lung cancer [ICD-11: 2C25.7] | |||
Resistant Drug | S-1 | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
MGB SNP detection kit assay; Mutation Detection assay | |||
Experiment for Drug Resistance |
Digital PCR assay | |||
Mechanism Description | Resistance mechanisms to EGFR-TkI therapy in EGFR-mutated NSCLC include secondary EGFR T790M mutation, c-Met amplification, PIk3CA mutation, and transformation to small-cell lung cancer. |
Preclinical Drug(s)
16 drug(s) in total
AV-412
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [73] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AV-412 | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 |
TE8 cells | Esophageal | Homo sapiens (Human) | CVCL_1766 | |
A4 cells | N.A. | Mus musculus (Mouse) | CVCL_F962 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
3H-thymidine incorporation assay | |||
Mechanism Description | The missense mutation p.T790M (c.2369C>T) in gene EGFR cause the sensitivity of AV-412 by aberration of the drug's therapeutic target | |||
Disease Class: Solid tumour/cancer | [73] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | AV-412 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | A431 cells | Skin | Homo sapiens (Human) | CVCL_0037 |
TE8 cells | Esophageal | Homo sapiens (Human) | CVCL_1766 | |
A4 cells | N.A. | Mus musculus (Mouse) | CVCL_F962 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
3H-thymidine incorporation assay | |||
Mechanism Description | The missense mutation p.L858R (c.2573T>G) in gene EGFR cause the sensitivity of AV-412 by aberration of the drug's therapeutic target |
AZD-3759
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [74] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | AZD-3759 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
NCI-H838 cells | Lung | Homo sapiens (Human) | CVCL_1594 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NCI-H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
In Vivo Model | Nude mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Enzyme-linked immunosorbent assay | |||
Mechanism Description | AZD3759, a selective EGFR inhibitor that can fully penetrate the blood-brain barrier (BBB), with equal free concentrations in the blood, cerebrospinal fluid, and brain tissue. Treatment with AZD3759 causes tumor regression in subcutaneous xenograft, leptomeningeal metastasis (LM), and brain metastasis (BM) lung cancer models and prevents the development of BM in nude mice. |
Buparlisib/Osimertinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [75] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Buparlisib/Osimertinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Axon guidance signaling pathway | Inhibition | hsa04360 | |
MAPK signaling pathway | Inhibition | hsa04010 | ||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
HCC4006 cells | Lung | Homo sapiens (Human) | CVCL_1269 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NCI-H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
11-18 cells | Lung | Homo sapiens (Human) | CVCL_6659 | |
In Vivo Model | Athymic female mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | Src family kinases (SFK) and focal adhesion kinase (FAK) sustain AKT and mitogen-activated protein kinase (MAPK) pathway signaling under continuous EGFR inhibition in osimertinib sensitive cells. Inhibiting either the MAPK pathway or the AKT pathway enhanced the effects of osimertinib. Combined SFK/FAK inhibition exhibited the most potent effects on growth inhibition, induction of apoptosis, and delay of acquired resistance. SFK family member YES1 was amplified in osimertinib- resistant EGFR-mutant tumor cells, the effects of which were overcome by combined treatment with osimertinib and SFK inhibitors. |
EAI045
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [76] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | EAI045 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HaCaT cells | Tongue | Homo sapiens (Human) | CVCL_0038 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
NIH-3T3 cells | Embryo | Mus musculus (Mouse) | CVCL_0594 | |
In Vivo Model | Nude mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
CellTitre-Glo assay; MTS assay | |||
Mechanism Description | EAI045 is a kind of EGFR tyrosine kinase inhibitor. |
EKI-285
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [77] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | EKI-285 | |||
Molecule Alteration | Missense mutation | p.H773L (c.2318A>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NIH-H1975 | N.A. | . | N.A. |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | The missense mutation p.H773L (c.2318A>T) in gene EGFR cause the resistance of EKI-285 by aberration of the drug's therapeutic target |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [40] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | EKI-285 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lung adenocarcinomas tissue | . | ||
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Mechanism Description | The missense mutation p.L858R (c.2573T>G) in gene EGFR cause the sensitivity of EKI-285 by aberration of the drug's therapeutic target | |||
Disease Class: Solid tumour/cancer | [78] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | EKI-285 | |||
Molecule Alteration | Missense mutation | p.L844V (c.2530C>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
HCC827EP cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
H3255DR cells | Lung | Homo sapiens (Human) | CVCL_DI56 | |
H197 cells | N.A. | . | N.A. | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay; CytoSelect 96-well cell transformation assay | |||
Disease Class: Solid tumour/cancer | [78] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | EKI-285 | |||
Molecule Alteration | Missense mutation | p.L718Q (c.2153T>A) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
HCC827EP cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
H3255DR cells | Lung | Homo sapiens (Human) | CVCL_DI56 | |
H197 cells | N.A. | . | N.A. | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay; CytoSelect 96-well cell transformation assay |
ER2
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [79] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | ER2 | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 | |
H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
H1299 cells | Lung | Homo sapiens (Human) | CVCL_0060 | |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
PC9GR cells | Lung | Homo sapiens (Human) | CVCL_V337 | |
H358 cells | Lung | Homo sapiens (Human) | CVCL_1559 | |
H322 cells | Lung | Homo sapiens (Human) | CVCL_1556 | |
In Vivo Model | Female athymic BALB-c/nu mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
FACS assay; Propidium iodide staining assay | |||
Mechanism Description | ER2 suppressed EGFR pathway and induced G1 arrest and apoptosis in NSCLC cells. ER2 inhibited EGF-stimulated VEGF production. ER2 induced anti-tumor activity in combination with cisplatin in xenograft models. |
Erlotinib/Ganetespib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [80] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Erlotinib/Ganetespib | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NCI-H292 cells | Lung | Homo sapiens (Human) | CVCL_0455 |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
NCI-HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
NCI-H1395 cells | Lung | Homo sapiens (Human) | CVCL_1467 | |
NCI-H322 cells | Lung | Homo sapiens (Human) | CVCL_1556 | |
NCI-H1666 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1485 | |
In Vivo Model | NSCLC xenograft tumor model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Cell Titer Glo assay |
hEGFR vIII-CD3 bi-scFv
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Brain glioma | [81] | |||
Sensitive Disease | Brain glioma [ICD-11: 2A00.0] | |||
Sensitive Drug | hEGFR vIII-CD3 bi-scFv | |||
Molecule Alteration | Complex-indel | (c.89_889del11867) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | U87-MG cells | Brain | Homo sapiens (Human) | CVCL_0022 |
Experiment for Drug Resistance |
Chromium release assay; FACS assay |
JBJ-04-125-02
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [82] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | JBJ-04-125-02 | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [82] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | JBJ-04-125-02 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
In Vivo Model | Genetically engineered mouse xenograft model | Mus musculus | ||
Experiment for Drug Resistance |
MTS assay |
MM-151
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.S492R (c.1476C>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. | |||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.R451C (c.1351C>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. | |||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.S464L (c.1391C>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. | |||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.G465R (c.1393G>A) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. | |||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.G465E (c.1394G>A) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. | |||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.K467T (c.1400A>C) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. | |||
Disease Class: Colorectal cancer | [83] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | MM-151 | |||
Molecule Alteration | Missense mutation | p.I491M (c.1473A>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Inhibition | hsa04010 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | MM-151 inhibits EGFR signaling and cell growth in preclinical models, including patient-derived cells carrying mutant EGFR. Upon MM-151 treatment, EGFR ECD mutations decline in circulating cell-free tumor DNA (ctDNA) of CRC patients who previously developed resistance to EGFR blockade. |
Necitumumab
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [84] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Necitumumab | |||
Molecule Alteration | Missense mutation | p.S492R (c.1476C>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Hela cells | Cervix uteri | Homo sapiens (Human) | CVCL_0030 |
LK2 cells | Ascites | Homo sapiens (Human) | CVCL_W132 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
FACS assay |
OBX1-012
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Disease Class: Solid tumour/cancer | [85] | |||
Resistant Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Resistant Drug | OBX1-012 | |||
Molecule Alteration | IF-insertion | p.P772_H773insGNP (c.2316_2317insGGTAACCCT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | OBX1-012 treatment was highly effective against human EGFR-mutant lung cancer models with or without EGFR T790M, not only in vitro but also in vivo. However, OBX1-012 like other EGFR-TKIs failed to exhibit efficacy for the exon 20 insertion mutation or C797S mutation, which was generated by site-directed mutagenesis and stable transfection of Ba/F3 cells. |
Osimertinib/Selumetinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [75] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Osimertinib/Selumetinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Axon guidance signaling pathway | Inhibition | hsa04360 | |
MAPK signaling pathway | Inhibition | hsa04010 | ||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
HCC4006 cells | Lung | Homo sapiens (Human) | CVCL_1269 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NCI-H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
11-18 cells | Lung | Homo sapiens (Human) | CVCL_6659 | |
In Vivo Model | Athymic female mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | Src family kinases (SFK) and focal adhesion kinase (FAK) sustain AKT and mitogen-activated protein kinase (MAPK) pathway signaling under continuous EGFR inhibition in osimertinib sensitive cells. Inhibiting either the MAPK pathway or the AKT pathway enhanced the effects of osimertinib. Combined SFK/FAK inhibition exhibited the most potent effects on growth inhibition, induction of apoptosis, and delay of acquired resistance. SFK family member YES1 was amplified in osimertinib- resistant EGFR-mutant tumor cells, the effects of which were overcome by combined treatment with osimertinib and SFK inhibitors. | |||
Disease Class: Lung adenocarcinoma | [86] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Osimertinib/Selumetinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
In Vivo Model | mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
SYTOX green cucleic acid stain assay |
TAE226
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Solid tumour/cancer | [87] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAE226 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NCI-H2228 cells | Lung | Homo sapiens (Human) | CVCL_1543 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
NCI-H838 cells | Lung | Homo sapiens (Human) | CVCL_1594 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
HCC4006 cells | Lung | Homo sapiens (Human) | CVCL_1269 | |
NCI-H1395 cells | Lung | Homo sapiens (Human) | CVCL_1467 | |
SkBR3 cells | Breast | Homo sapiens (Human) | CVCL_0033 | |
HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 | |
NCI-H820 cells | Lymph node | Homo sapiens (Human) | CVCL_1592 | |
NCI-H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
NCI-H1993 cells | Lymph node | Homo sapiens (Human) | CVCL_1512 | |
NCI-H1819 cells | Lymph node | Homo sapiens (Human) | CVCL_1497 | |
NCI-H1666 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1485 | |
NCI-H1648 cells | Lymph node | Homo sapiens (Human) | CVCL_1482 | |
NCI-H1299 cells | Lymph node | Homo sapiens (Human) | CVCL_0060 | |
MKN45 cells | Liver | Homo sapiens (Human) | CVCL_0434 | |
In Vivo Model | BALB/c-nu/nu female nude mouse PC9 xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter-96 AQueous One assay | |||
Disease Class: Lung adenocarcinoma | [87] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAE226 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NCI-H2228 cells | Lung | Homo sapiens (Human) | CVCL_1543 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
NCI-H838 cells | Lung | Homo sapiens (Human) | CVCL_1594 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
HCC4006 cells | Lung | Homo sapiens (Human) | CVCL_1269 | |
NCI-H1395 cells | Lung | Homo sapiens (Human) | CVCL_1467 | |
SkBR3 cells | Breast | Homo sapiens (Human) | CVCL_0033 | |
HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 | |
NCI-H820 cells | Lymph node | Homo sapiens (Human) | CVCL_1592 | |
NCI-H3255 cells | Lung | Homo sapiens (Human) | CVCL_6831 | |
NCI-H1993 cells | Lymph node | Homo sapiens (Human) | CVCL_1512 | |
NCI-H1819 cells | Lymph node | Homo sapiens (Human) | CVCL_1497 | |
NCI-H1666 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1485 | |
NCI-H1648 cells | Lymph node | Homo sapiens (Human) | CVCL_1482 | |
NCI-H1299 cells | Lymph node | Homo sapiens (Human) | CVCL_0060 | |
MKN45 cells | Liver | Homo sapiens (Human) | CVCL_0434 | |
In Vivo Model | BALB/c-nu/nu female nude mouse PC9 xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter-96 AQueous One assay |
TAS6417
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Solid tumour/cancer | [88] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | IF-insertion | p.A763_Y764insFQEA (c.2290_2291insTTCAAGAGGCAT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Solid tumour/cancer | [88] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Solid tumour/cancer | [88] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Solid tumour/cancer | [88] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | IF-insertion | p.D770_N771insG (c.2310_2311insGGT) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Solid tumour/cancer | [88] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Solid tumour/cancer | [88] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.P772_H773 (c.2314_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Lung adenocarcinoma | [88] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Lung adenocarcinoma | [88] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.A767_V769 (c.2299_2307) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Lung adenocarcinoma | [88] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.S768_D770 (c.2302_2310) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. | |||
Disease Class: Lung adenocarcinoma | [88] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAS6417 | |||
Molecule Alteration | Duplication | p.N771_H773 (c.2311_2319) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
NCI-H23 cells | Lung | Homo sapiens (Human) | CVCL_1547 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
NCI-H1875 cells | N.A. | Homo sapiens (Human) | N.A. | |
LXF 2378L cells | N.A. | . | N.A. | |
In Vivo Model | BALB/c nude mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | TAS6417 is a novel EGFR inhibitor that targets EGFR exon 20 insertion mutations while sparing wild-type (WT) EGFR. In cell viability assays using Ba/F3 cells engineered to express human EGFR, TAS6417 inhibited EGFR with various exon 20 insertion mutations more potently than it inhibited the WT. Western blot analysis revealed that TAS6417 inhibited EGFR phosphorylation and downstream molecules in NSCLC cell lines expressing EGFR exon 20 insertions, resulting in caspase activation. These characteristics led to marked tumor regression in vivo in both a genetically engineered model and in a patient-derived xenograft model. Furthermore, TAS6417 provided a survival benefit with good tolerability in a lung orthotopic implantation mouse model. These findings support the clinical evaluation of TAS6417 as an efficacious drug candidate for patients with NSCLC harboring EGFR exon 20 insertion mutations. |
Trametinib/WZ4002
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [89] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Trametinib/WZ4002 | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
HCC4006 cells | Lung | Homo sapiens (Human) | CVCL_1269 | |
H1650 cells | Pleural effusion | Homo sapiens (Human) | CVCL_4V01 | |
HCC2935 cells | Lung | Homo sapiens (Human) | CVCL_1265 | |
DFCI81 cells | N.A. | . | N.A. | |
H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
In Vivo Model | Nu/Nu doxycycline-inducible transgenic mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Disease Class: Lung adenocarcinoma | [89] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Trametinib/WZ4002 | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 |
HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 | |
PC9 cells | Lung | Homo sapiens (Human) | CVCL_B260 | |
HCC4006 cells | Lung | Homo sapiens (Human) | CVCL_1269 | |
H1650 cells | Pleural effusion | Homo sapiens (Human) | CVCL_4V01 | |
HCC2935 cells | Lung | Homo sapiens (Human) | CVCL_1265 | |
DFCI81 cells | N.A. | . | N.A. | |
H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
In Vivo Model | Nu/Nu doxycycline-inducible transgenic mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay |
Investigative Drug(s)
11 drug(s) in total
Anti-EGFR treatment
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Non-small cell lung cancer | [90] | |||
Resistant Disease | Non-small cell lung cancer [ICD-11: 2C25.Y] | |||
Resistant Drug | Anti-EGFR treatment | |||
Molecule Alteration | Missense mutation | p.T790M |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Longitudinal quantitative cfDNA analysis | |||
Experiment for Drug Resistance |
Plasma-based tumor genotyping assay | |||
Mechanism Description | In patients with non-small cell lung cancer, the use of first-line anti-EGFR treatment in patients with activating EGFR mutations often leads to secondary resistance through an additional T790M mutation in EGFR in ~50% of initial responders. |
Bevacizumab/Erlotinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [91] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Bevacizumab/Erlotinib | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data |
Bevacizumab/Gefitinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [92] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Bevacizumab/Gefitinib | |||
Molecule Alteration | Complex-indel | p.H773_V774delinsLM (c.2317_2322delinsCTTATG) |
||
Experimental Note | Identified from the Human Clinical Data |
Cabozantinib/Erlotinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [93] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Cabozantinib/Erlotinib | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Identified from the Human Clinical Data |
EGFR TKIs
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Brain glioma | [94] | |||
Resistant Disease | Brain glioma [ICD-11: 2A00.0] | |||
Resistant Drug | EGFR TKIs | |||
Molecule Alteration | IF-deletion | p.V30_V336 (c.88_1008) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Brain | . | ||
Mechanism Description | The if-deletion p.V30_V336 (c.88_1008) in gene EGFR cause the resistance of EGFR TKIs by aberration of the drug's therapeutic target. | |||
Disease Class: Brain glioma | [95] | |||
Resistant Disease | Brain glioma [ICD-11: 2A00.0] | |||
Resistant Drug | EGFR TKIs | |||
Molecule Alteration | Copy number gain | . |
||
Experimental Note | Identified from the Human Clinical Data |
Futuximab
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Solid tumour/cancer | [96] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.R451C (c.1351C>T) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Regulation | hsa01521 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
NIH3T3 cells | Embryo | Homo sapiens (Human) | N.A. | |
EGFR cells | N.A. | . | N.A. | |
In Vivo Model | Male BALB/c nude mouse | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay; FACS assay; Crystal violet staining assay | |||
Mechanism Description | Contrary to cetuximab and panitumumab, Sym004 effectively binds to and prevents activation of all the EGFR mutants. Sym004 effectively inhibits proliferation and EGFR downstream signaling in cetuximab-resistant derivatives harboring the S492R and G465R EGFR mutations. Sym004 causes profound and sustained regression in S492R-mutant EGFR and delays tumor growth in G465R-mutant EGFR in vivo. | |||
Disease Class: Solid tumour/cancer | [96] | |||
Sensitive Disease | Solid tumour/cancer [ICD-11: 2A00-2F9Z] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.K467T (c.1400A>C) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Regulation | hsa01521 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
NIH3T3 cells | Embryo | Homo sapiens (Human) | N.A. | |
EGFR cells | N.A. | . | N.A. | |
In Vivo Model | Male BALB/c nude mouse | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay; FACS assay; Crystal violet staining assay | |||
Mechanism Description | Contrary to cetuximab and panitumumab, Sym004 effectively binds to and prevents activation of all the EGFR mutants. Sym004 effectively inhibits proliferation and EGFR downstream signaling in cetuximab-resistant derivatives harboring the S492R and G465R EGFR mutations. Sym004 causes profound and sustained regression in S492R-mutant EGFR and delays tumor growth in G465R-mutant EGFR in vivo. | |||
Disease Class: Colorectal cancer | [96] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.R451C (c.1351C>T) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Regulation | hsa01521 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
NIH3T3 cells | Embryo | Homo sapiens (Human) | N.A. | |
EGFR cells | N.A. | . | N.A. | |
In Vivo Model | Male BALB/c nude mouse | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay; FACS assay; Crystal violet staining assay | |||
Mechanism Description | Contrary to cetuximab and panitumumab, Sym004 effectively binds to and prevents activation of all the EGFR mutants. Sym004 effectively inhibits proliferation and EGFR downstream signaling in cetuximab-resistant derivatives harboring the S492R and G465R EGFR mutations. Sym004 causes profound and sustained regression in S492R-mutant EGFR and delays tumor growth in G465R-mutant EGFR in vivo. | |||
Disease Class: Colorectal cancer | [96] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.G465R (c.1393G>A) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Regulation | hsa01521 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
NIH3T3 cells | Embryo | Homo sapiens (Human) | N.A. | |
EGFR cells | N.A. | . | N.A. | |
In Vivo Model | Male BALB/c nude mouse | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay; FACS assay; Crystal violet staining assay | |||
Mechanism Description | Contrary to cetuximab and panitumumab, Sym004 effectively binds to and prevents activation of all the EGFR mutants. Sym004 effectively inhibits proliferation and EGFR downstream signaling in cetuximab-resistant derivatives harboring the S492R and G465R EGFR mutations. Sym004 causes profound and sustained regression in S492R-mutant EGFR and delays tumor growth in G465R-mutant EGFR in vivo. | |||
Disease Class: Colorectal cancer | [96] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.K467T (c.1400A>C) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Regulation | hsa01521 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
NIH3T3 cells | Embryo | Homo sapiens (Human) | N.A. | |
EGFR cells | N.A. | . | N.A. | |
In Vivo Model | Male BALB/c nude mouse | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay; FACS assay; Crystal violet staining assay | |||
Mechanism Description | Contrary to cetuximab and panitumumab, Sym004 effectively binds to and prevents activation of all the EGFR mutants. Sym004 effectively inhibits proliferation and EGFR downstream signaling in cetuximab-resistant derivatives harboring the S492R and G465R EGFR mutations. Sym004 causes profound and sustained regression in S492R-mutant EGFR and delays tumor growth in G465R-mutant EGFR in vivo. | |||
Disease Class: Colorectal cancer | [96] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.S492R (c.1476C>A) |
||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | EGFR signaling pathway | Regulation | hsa01521 | |
In Vitro Model | LIM1215 cells | Colon | Homo sapiens (Human) | CVCL_2574 |
NIH3T3 cells | Embryo | Homo sapiens (Human) | N.A. | |
EGFR cells | N.A. | . | N.A. | |
In Vivo Model | Male BALB/c nude mouse | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Promega assay; FACS assay; Crystal violet staining assay | |||
Mechanism Description | Contrary to cetuximab and panitumumab, Sym004 effectively binds to and prevents activation of all the EGFR mutants. Sym004 effectively inhibits proliferation and EGFR downstream signaling in cetuximab-resistant derivatives harboring the S492R and G465R EGFR mutations. Sym004 causes profound and sustained regression in S492R-mutant EGFR and delays tumor growth in G465R-mutant EGFR in vivo. | |||
Disease Class: Colorectal cancer | [97] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | Futuximab | |||
Molecule Alteration | Missense mutation | p.S492R (c.1476C>G) |
||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | HT29 Cells | Colon | Homo sapiens (Human) | CVCL_A8EZ |
DLD1 cells | Colon | Homo sapiens (Human) | CVCL_0248 | |
SW620 cells | Colon | Homo sapiens (Human) | CVCL_0547 | |
LS1034 cells | Colon | Homo sapiens (Human) | CVCL_1382 | |
COLO205 cells | Colon | Homo sapiens (Human) | CVCL_F402 | |
GEO cells | Colon | Homo sapiens (Human) | CVCL_0271 | |
HCT15 cells | Colon | Homo sapiens (Human) | CVCL_0292 | |
SW480 cells | Colon | Homo sapiens (Human) | CVCL_0546 | |
CaCo2 cells | Colon | Homo sapiens (Human) | CVCL_0025 | |
HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 | |
LOVO cells | Colon | Homo sapiens (Human) | CVCL_0399 | |
LS174T cells | Colon | Homo sapiens (Human) | CVCL_1384 | |
SW948 cells | Colon | Homo sapiens (Human) | CVCL_0632 | |
SW403 cells | Colon | Homo sapiens (Human) | CVCL_0545 | |
SW837 cells | Colon | Homo sapiens (Human) | CVCL_1729 | |
T84 cells | Colon | Homo sapiens (Human) | CVCL_0555 | |
SW1463 cells | Rectum | Homo sapiens (Human) | CVCL_1718 | |
H716 cells | Ascites | Homo sapiens (Human) | CVCL_1581 | |
H508 cells | Abdominal wall | Homo sapiens (Human) | CVCL_1564 | |
SNUC2A cells | Cecum | Homo sapiens (Human) | CVCL_1709 | |
COLO678 cells | Colon | Homo sapiens (Human) | CVCL_1129 | |
GP5D cells | Colon | Homo sapiens (Human) | CVCL_1235 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
WST-1 assay |
PKI-587
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [98] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | PKI-587 | |||
Molecule Alteration | IF-deletion | p.E746_A750delELREA (c.2236_2250del15) |
||
Experimental Note | Identified from the Human Clinical Data |
Staurosporine
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [99] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | Staurosporine | |||
Molecule Alteration | Missense mutation | p.T790M (c.2369C>T) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Lung | . |
TAS-121
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Colorectal cancer | [100] | |||
Sensitive Disease | Colorectal cancer [ICD-11: 2B91.1] | |||
Sensitive Drug | TAS-121 | |||
Molecule Alteration | Missense mutation | p.G719S (c.2155G>A) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
SW48 cells | Colon | Homo sapiens (Human) | CVCL_1724 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
In Vivo Model | Male BALB/cA nude mouse xenograft model | Mus musculus | ||
Experiment for Drug Resistance |
CellTiter-Glo assay; IC50 assay | |||
Disease Class: Lung adenocarcinoma | [100] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAS-121 | |||
Molecule Alteration | Missense mutation | p.G719S (c.2155G>A) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
SW48 cells | Colon | Homo sapiens (Human) | CVCL_1724 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
In Vivo Model | Male BALB/cA nude mouse xenograft model | Mus musculus | ||
Experiment for Drug Resistance |
CellTiter-Glo assay; IC50 assay | |||
Disease Class: Lung adenocarcinoma | [100] | |||
Sensitive Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Sensitive Drug | TAS-121 | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCC827 cells | Lung | Homo sapiens (Human) | CVCL_2063 |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
SW48 cells | Colon | Homo sapiens (Human) | CVCL_1724 | |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
In Vivo Model | Male BALB/cA nude mouse xenograft model | Mus musculus | ||
Experiment for Drug Resistance |
CellTiter-Glo assay; IC50 assay |
Unspecified CTLA4 antibody/Unspecified PD-1 antibody
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Disease Class: Lung adenocarcinoma | [101] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Unspecified CTLA4 antibody/Unspecified PD-1 antibody | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data |
Unspecified PD-L1 antibody
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Disease Class: Lung adenocarcinoma | [101] | |||
Resistant Disease | Lung adenocarcinoma [ICD-11: 2C25.0] | |||
Resistant Drug | Unspecified PD-L1 antibody | |||
Molecule Alteration | Missense mutation | p.L858R (c.2573T>G) |
||
Experimental Note | Identified from the Human Clinical Data |
Disease- and Tissue-specific Abundances of This Molecule
ICD Disease Classification 02
Brain cancer [ICD-11: 2A00]
Differential expression of molecule in resistant diseases | ||
The Studied Tissue | Nervous tissue | |
The Specified Disease | Brain cancer | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 8.30E-26; Fold-change: 6.21E-02; Z-score: 1.32E-01 | |
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
The Studied Tissue | Brainstem tissue | |
The Specified Disease | Glioma | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 2.09E-05; Fold-change: 3.96E-01; Z-score: 1.90E+02 | |
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
The Studied Tissue | White matter | |
The Specified Disease | Glioma | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 6.32E-04; Fold-change: 3.88E-01; Z-score: 1.69E+00 | |
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
The Studied Tissue | Brainstem tissue | |
The Specified Disease | Neuroectodermal tumor | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 1.84E-08; Fold-change: -1.12E+00; Z-score: -4.45E+00 | |
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
Lung cancer [ICD-11: 2C25]
Differential expression of molecule in resistant diseases | ||
The Studied Tissue | Lung | |
The Specified Disease | Lung cancer | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 1.65E-01; Fold-change: -4.23E-02; Z-score: -8.52E-02 | |
The Expression Level of Disease Section Compare with the Adjacent Tissue | p-value: 1.58E-03; Fold-change: -2.14E-01; Z-score: -3.48E-01 | |
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
Melanoma [ICD-11: 2C30]
Differential expression of molecule in resistant diseases | ||
The Studied Tissue | Skin | |
The Specified Disease | Melanoma | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 1.60E-01; Fold-change: -3.39E-01; Z-score: -2.30E-01 | |
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
Breast cancer [ICD-11: 2C60]
Differential expression of molecule in resistant diseases | ||
The Studied Tissue | Breast tissue | |
The Specified Disease | Breast cancer | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 1.04E-65; Fold-change: -8.87E-01; Z-score: -1.53E+00 | |
The Expression Level of Disease Section Compare with the Adjacent Tissue | p-value: 2.77E-16; Fold-change: -9.49E-01; Z-score: -1.79E+00 | |
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
Prostate cancer [ICD-11: 2C82]
Differential expression of molecule in resistant diseases | ||
The Studied Tissue | Prostate | |
The Specified Disease | Prostate cancer | |
The Expression Level of Disease Section Compare with the Healthy Individual Tissue | p-value: 7.43E-04; Fold-change: -5.92E-01; Z-score: -8.22E-01 | |
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
|
||
Disease-specific Molecule Abundances | Click to View the Clearer Original Diagram | |
Tissue-specific Molecule Abundances in Healthy Individuals
References
If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.