General Information of the Molecule (ID: Mol00593)
Name
GTPase Nras (NRAS) ,Homo sapiens
Synonyms
Transforming protein N-Ras; HRAS1
    Click to Show/Hide
Molecule Type
Protein
Gene Name
NRAS
Gene ID
4893
Location
chr1:114704469-114716771[-]
Sequence
MTEYKLVVVGAGGVGKSALTIQLIQNHFVDEYDPTIEDSYRKQVVIDGETCLLDILDTAG
QEEYSAMRDQYMRTGEGFLCVFAINNSKSFADINLYREQIKRVKDSDDVPMVLVGNKCDL
PTRTVDTKQAHELAKSYGIPFIETSAKTRQGVEDAFYTLVREIRQYRMKKLNSSDDGTQG
CMGLPCVVM
    Click to Show/Hide
Function
Ras proteins bind GDP/GTP and possess intrinsic GTPase activity.
    Click to Show/Hide
Uniprot ID
RASN_HUMAN
Ensembl ID
ENSG00000213281
HGNC ID
HGNC:7989
        Click to Show/Hide the Complete Species Lineage
Kingdom: Metazoa
Phylum: Chordata
Class: Mammalia
Order: Primates
Family: Hominidae
Genus: Homo
Species: Homo sapiens
Type(s) of Resistant Mechanism of This Molecule
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
19 drug(s) in total
Click to Show/Hide the Full List of Drugs
Binimetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Binimetinib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Binimetinib
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.Q61R (c.182A>G) in gene NRAS cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Binimetinib
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.Q61L (c.182A>T) in gene NRAS cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Binimetinib
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.Q61R (c.182A>G) in gene NRAS cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Binimetinib
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.Q61L (c.182A>T) in gene NRAS cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Bosutinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [2], [3]
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Resistant Drug Bosutinib
Molecule Alteration Missense mutation
p.G12V
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT signaling pathway Activation hsa04030
RAF/KRAS/MEK signaling pathway Activation hsa04010
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
U937 cells Blood Homo sapiens (Human) CVCL_0007
K562 cells Blood Homo sapiens (Human) CVCL_0004
KCL-22 cells Bone marrow Homo sapiens (Human) CVCL_2091
Sup-B15 cells Bone marrow Homo sapiens (Human) CVCL_0103
HEL cells Blood Homo sapiens (Human) CVCL_0001
HMC-1.2 cells Blood Homo sapiens (Human) CVCL_H205
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay; Sanger Sequencing assay
Mechanism Description This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient.
Cetuximab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Metastatic colorectal cancer [4]
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Resistant Drug Cetuximab
Molecule Alteration Missense mutation
p.G12C
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation EGFR/RAS signaling pathway Inhibition hsa01521
In Vitro Model LIM1215 cells Colon Homo sapiens (Human) CVCL_2574
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 biopsies assay; Functional analyses of cell populations assay
Mechanism Description Acquired resistance to EGFR blockade is driven by the emergence of kRAS/NRAS mutations or the development of EGFR extracellular domain (ECD) variants, which impair antibody binding.
Cisplatin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung cancer [5]
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
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 viability Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay; Caspase-3 Activity Assay
Mechanism Description miR-29a renders lung cancer cells more sensitive to cisplatin treatment and miR-29a and cisplatin combination promoted apoptotic effect through targeting NRAS in lung cancer cells.
Disease Class: Gastric cancer [6]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
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 SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description NRAS and E2F2 as the direct targets of miR-26a were further confirmed in luciferase activity assays and miR-26a-mediated these two genes expression analysis. Our results also found that knockdown of NRAS or E2F2 sensitize GC cells to cisplatin. miR-26a overexpression has been demonstrated to improve the sensitivity of GC cells to cisplatin and this effect was considered to be mediated via its targets NRAS and E2F2.
Dabrafenib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [7], [8], [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Dabrafenib
Molecule Alteration Missense mutation
p.Q61K
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Whole-exome sequencing assay; Sanger sequencing assay; Next generation assay; Single PCR-based analysis
Experiment for
Drug Resistance
Progression-free and post-progression survival asaay; Computed tomography assay; Positron emission tomography assay
Mechanism Description Another post-relapse tumor harbored an acquired NRASQ61k missense mutation together with focal BRAF amplification. The resistant tumor from a third patient harbored both a MEk2 mutation and BRAF amplification. Resistance mechanisms are identified in 9/11 progressing tumours and MAPk reactivation occurred in 9/10 tumours, commonly via BRAF amplification and mutations activating NRAS and MEk2. Our data confirming that MEk2C125S, but not the synonymous MEk1C121S protein, confers resistance to combination therapy highlight the functional differences between these kinases and the preponderance of MEk2 mutations in combination therapy-resistant melanomas.
Disease Class: Melanoma [9], [10]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Dabrafenib
Molecule Alteration Missense mutation
p.Q61R
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next generation assay; Single PCR-based analysis
Experiment for
Drug Resistance
Computed tomography assay; Positron emission tomography assay; Progression-free and overall survival assay
Mechanism Description NRAS mutations (Q61R and Q61k in codon 61) were detected in two of ten patients (20%). Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Dabrafenib
Molecule Alteration Missense mutation
p.Q61L
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Dabrafenib
Molecule Alteration Missense mutation
p.G13R
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Dabrafenib
Molecule Alteration Missense mutation
p.G12R
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9], [11]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Dabrafenib
Molecule Alteration Missense mutation
p.G12D
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy. The Prog that did not show evidence of MAPk reactivation by GSEA had two identified resistance mechanisms (MEk2E207k and NRASG12D), but both variants occurred at low frequency (13 and 15% allelic frequency, respectively, by whole-exome sequencing), suggesting heterogeneity within the Prog metastasis.
Dasatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [2], [3]
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Resistant Drug Dasatinib
Molecule Alteration Missense mutation
p.G12V
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT signaling pathway Activation hsa04030
RAF/KRAS/MEK signaling pathway Activation hsa04010
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
U937 cells Blood Homo sapiens (Human) CVCL_0007
K562 cells Blood Homo sapiens (Human) CVCL_0004
KCL-22 cells Bone marrow Homo sapiens (Human) CVCL_2091
Sup-B15 cells Bone marrow Homo sapiens (Human) CVCL_0103
HEL cells Blood Homo sapiens (Human) CVCL_0001
HMC-1.2 cells Blood Homo sapiens (Human) CVCL_H205
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay; Sanger Sequencing assay
Mechanism Description This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient.
Imatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [2], [3]
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Resistant Drug Imatinib
Molecule Alteration Missense mutation
p.G12V
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT signaling pathway Activation hsa04030
RAF/KRAS/MEK signaling pathway Activation hsa04010
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
U937 cells Blood Homo sapiens (Human) CVCL_0007
K562 cells Blood Homo sapiens (Human) CVCL_0004
KCL-22 cells Bone marrow Homo sapiens (Human) CVCL_2091
Sup-B15 cells Bone marrow Homo sapiens (Human) CVCL_0103
HEL cells Blood Homo sapiens (Human) CVCL_0001
HMC-1.2 cells Blood Homo sapiens (Human) CVCL_H205
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay; Sanger Sequencing assay
Mechanism Description This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient.
Lapatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: HER2 positive breast cancer [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Lapatinib
Molecule Alteration Missense mutation
p.V14A
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Lapatinib
Molecule Alteration Missense mutation
p.F78L
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Lapatinib
Molecule Alteration Missense mutation
p.F28S
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Lapatinib
Molecule Alteration Missense mutation
p.A66T
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
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [2], [3]
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Resistant Drug Nilotinib
Molecule Alteration Missense mutation
p.G12V
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT signaling pathway Activation hsa04030
RAF/KRAS/MEK signaling pathway Activation hsa04010
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
U937 cells Blood Homo sapiens (Human) CVCL_0007
K562 cells Blood Homo sapiens (Human) CVCL_0004
KCL-22 cells Bone marrow Homo sapiens (Human) CVCL_2091
Sup-B15 cells Bone marrow Homo sapiens (Human) CVCL_0103
HEL cells Blood Homo sapiens (Human) CVCL_0001
HMC-1.2 cells Blood Homo sapiens (Human) CVCL_H205
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay; Sanger Sequencing assay
Mechanism Description This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient.
Paclitaxel
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Breast cancer [13]
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
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 migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDM231 cells Breast Homo sapiens (Human) CVCL_5T76
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description microRNA-22 sensitized breast cancer cells to paclitaxel by downregulation of NRAS.
Pamidronate
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Breast cancer [14]
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Resistant Drug Pamidronate
Molecule Alteration Expression
Down-regulation
Experimental Note Identified from the Human Clinical Data
In Vitro Model U266 cells Bone marrow Homo sapiens (Human) CVCL_0566
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
Pseudomonas aeruginosa strain B-730P/17 287
Experiment for
Drug Resistance
CellTiter 96 one solution proliferation assay
Mechanism Description In response to pamidronate, there was significant inhibition of cell proliferation in MDA-231 and SKBR-3 cells, compared to MDA-175 cells. This correlated with their respective basal levels of N-ras and H-ras. N-ras and H-ras protein levels were both reduced in MDA-231 cells, and to lesser extent in SKBR-3 cells, following exposure to pamidronate, whereas these markers were not altered in MDA-175 cells, resistance to pamidronate may result from low levels of GTPase-activating proteins, such as N-ras and H-ras, in tumor cells.
Panitumumab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Metastatic colorectal cancer [4]
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Resistant Drug Panitumumab
Molecule Alteration Missense mutation
p.G12C
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation EGFR/RAS signaling pathway Inhibition hsa01521
In Vitro Model LIM1215 cells Colon Homo sapiens (Human) CVCL_2574
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 biopsies assay; Functional analyses of cell populations assay
Mechanism Description Acquired resistance to EGFR blockade is driven by the emergence of kRAS/NRAS mutations or the development of EGFR extracellular domain (ECD) variants, which impair antibody binding.
Disease Class: Colorectal cancer [15]
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Resistant Drug Panitumumab
Molecule Alteration Mutation
.
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
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: Colorectal cancer [16]
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Resistant Drug Panitumumab
Molecule Alteration Mutation
.
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Sanger sequencing assay; Next-generation sequencing assay
Mechanism Description Mutations in kRAS, NRAS, and BRAF and amplification of ERBB2 and MET drive primary (de novo) resistance to anti-EGFR treatment.
Pembrolizumab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [17]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Pembrolizumab
Molecule Alteration Missense mutation
p.Q61R
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Circulating tumour DNA (ctDNA) analysis; Whole genome plasma DNA sequencing assay
Experiment for
Drug Resistance
Computer tomography (CT) assay; Positron emission tomography assay
Mechanism Description Mutations in NRAS have been found in 8-26% of patients with acquired resistance to BRAF inhibitors. We analysed the presence of NRASQ61k and NRASQ61R in the ctDNA extracted from 7 melanoma patients with progressive disease who had previously responded to treatment with vemurafenib (n = 2) or dabrafenib/trametinib (n = 5). Two samples were positive for NRASQ61k and one sample had an NRASQ61R mutation, all three were derived from patients treated with dabrafenib/trametinib.
Pertuzumab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: HER2 positive breast cancer [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Pertuzumab
Molecule Alteration Missense mutation
p.V14A
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Pertuzumab
Molecule Alteration Missense mutation
p.F78L
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Pertuzumab
Molecule Alteration Missense mutation
p.F28S
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Pertuzumab
Molecule Alteration Missense mutation
p.A66T
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.
Ponatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Chronic myeloid leukemia [2], [3]
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Resistant Drug Ponatinib
Molecule Alteration Missense mutation
p.G12V
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT signaling pathway Activation hsa04030
RAF/KRAS/MEK signaling pathway Activation hsa04010
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
U937 cells Blood Homo sapiens (Human) CVCL_0007
K562 cells Blood Homo sapiens (Human) CVCL_0004
KCL-22 cells Bone marrow Homo sapiens (Human) CVCL_2091
Sup-B15 cells Bone marrow Homo sapiens (Human) CVCL_0103
HEL cells Blood Homo sapiens (Human) CVCL_0001
HMC-1.2 cells Blood Homo sapiens (Human) CVCL_H205
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay; Sanger Sequencing assay
Mechanism Description This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient.
Trametinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [18]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Trametinib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
SW1271 cells Lung Homo sapiens (Human) CVCL_1716
H2347 cells Lung Homo sapiens (Human) CVCL_1550
H2087 cells Lymph node Homo sapiens (Human) CVCL_1524
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
Cell Titer blue reagent assay
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Trametinib by unusual activation of pro-survival pathway
Trastuzumab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: HER2 positive breast cancer [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab
Molecule Alteration Missense mutation
p.V14A
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab
Molecule Alteration Missense mutation
p.F78L
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab
Molecule Alteration Missense mutation
p.F28S
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab
Molecule Alteration Missense mutation
p.A66T
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.
Trastuzumab emtansine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: HER2 positive breast cancer [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab emtansine
Molecule Alteration Missense mutation
p.V14A
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab emtansine
Molecule Alteration Missense mutation
p.F78L
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab emtansine
Molecule Alteration Missense mutation
p.F28S
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 [12]
Resistant Disease HER2 positive breast cancer [ICD-11: 2C60.8]
Resistant Drug Trastuzumab emtansine
Molecule Alteration Missense mutation
p.A66T
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.
Vemurafenib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Multiple myeloma [19]
Resistant Disease Multiple myeloma [ICD-11: 2A83.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61H
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Ion Torrent semiconductor-based targeted resequencing assay
Experiment for
Drug Resistance
Whole-body magnetic resonance imaging (MRI) assay
Mechanism Description Although all 5 reference lesions biopsied in month 10 still harbored a BRAFV600E mutation in all MM cells, an additio.l monoallelic NRAS mutation was detectable in each of the 3 lesions resistant to the full dose of vemurafenib. Of note, each lesion harbored a unique, independent, yet clo.l NRAS mutation (NRAS G13R, NRAS G12A, and NRAS Q61H, respectively).
Disease Class: Multiple myeloma [19]
Resistant Disease Multiple myeloma [ICD-11: 2A83.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.G13R
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Ion Torrent semiconductor-based targeted resequencing assay
Experiment for
Drug Resistance
Whole-body magnetic resonance imaging (MRI) assay
Mechanism Description Although all 5 reference lesions biopsied in month 10 still harbored a BRAFV600E mutation in all MM cells, an additio.l monoallelic NRAS mutation was detectable in each of the 3 lesions resistant to the full dose of vemurafenib. Of note, each lesion harbored a unique, independent, yet clo.l NRAS mutation (NRAS G13R, NRAS G12A, and NRAS Q61H, respectively).
Disease Class: Multiple myeloma [19]
Resistant Disease Multiple myeloma [ICD-11: 2A83.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.G12A
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Ion Torrent semiconductor-based targeted resequencing assay
Experiment for
Drug Resistance
Whole-body magnetic resonance imaging (MRI) assay
Mechanism Description Although all 5 reference lesions biopsied in month 10 still harbored a BRAFV600E mutation in all MM cells, an additio.l monoallelic NRAS mutation was detectable in each of the 3 lesions resistant to the full dose of vemurafenib. Of note, each lesion harbored a unique, independent, yet clo.l NRAS mutation (NRAS G13R, NRAS G12A, and NRAS Q61H, respectively).
Disease Class: Melanoma [7]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61H
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Whole Exome Sequencing assay
Experiment for
Drug Resistance
Progression-free survival assay; Overall survival assay
Mechanism Description In contrast, NRAS mutations and BRAF amplifications may still prove responsive to subsequent MEk inhibitor-based regimens, although the existing clinical data suggests that patients who progress following single-agent RAF inhibition are less likely to benefit from MEk inhibitors.
Disease Class: Melanoma [7], [9], [20]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61R
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
In Vitro Model M229 cells Skin Homo sapiens (Human) CVCL_D748
M238 cells Skin Homo sapiens (Human) CVCL_D751
M249 cells Skin Homo sapiens (Human) CVCL_D755
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61L
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [7], [9], [20]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61K
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
In Vitro Model M229 cells Skin Homo sapiens (Human) CVCL_D748
M238 cells Skin Homo sapiens (Human) CVCL_D751
M249 cells Skin Homo sapiens (Human) CVCL_D755
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.G13R
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.G12R
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.G12D
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PI3K/AKT/PTEN signaling pathway Inhibition hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description Somatic mutations in NRAS (Q61k/R/L, G12D/R and G13R) were detected till date by whole exome sequencing in 8-18% of BRAF inhibitor-resistant patients; in most cases, as a late event beyond 12 weeks of therapy.
Disease Class: Melanoma [21], [22]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61K
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Liquid biopsy assay; Next-generation sequencing assay; Circulating-free DNA assay; Digital PCR assay
Experiment for
Drug Resistance
Overall and disease-free assay
Mechanism Description Overexpression of PDGFRbeta or N-RAS(Q61k) conferred PLX4032 resistance to PLX4032-sensitive parental cell lines.
Disease Class: Melanoma [23]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61K
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
Mechanism Description BRAFV600E inhibition via vemurafenib induces paradoxical activation of MAPK through increased CRAF activity and acquired NRAS mutation. Moreover, mutations in genes upstream of RAF, such as the activating N-RASQ61K mutation, allow for BRAFV600 melanomas to escape molecular targeting.
Disease Class: Melanoma [23]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Vemurafenib
Molecule Alteration Missense mutation
p.Q61K
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
Mechanism Description BRAFV600E inhibition via vemurafenib induces paradoxical activation of MAPK through increased CRAF activity and acquired NRAS mutation. Moreover, mutations in genes upstream of RAF, such as the activating N-RASQ61K mutation, allow for BRAFV600 melanomas to escape molecular targeting.
Clinical Trial Drug(s)
4 drug(s) in total
Click to Show/Hide the Full List of Drugs
Selumetinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [24]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Selumetinib
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL 30 cells Skin Homo sapiens (Human) CVCL_0039
SkMEL 2 cells Skin Homo sapiens (Human) CVCL_0069
SH4 cells Skin Mus musculus (Mouse) CVCL_7702
MEXF-535 cells Skin Homo sapiens (Human) N.A.
MEXF-1792 cells Skin Homo sapiens (Human) N.A.
MEXF-1341 cells Skin Homo sapiens (Human) N.A.
M14 cells Hypodermis Homo sapiens (Human) CVCL_1395
GAK cells Lnguinal lymph node Homo sapiens (Human) CVCL_1225
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Female NIH nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Crystallization assay; X-ray data collection and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Enzymatic kinase assay
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [18]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Selumetinib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
SW1271 cells Lung Homo sapiens (Human) CVCL_1716
H2347 cells Lung Homo sapiens (Human) CVCL_1550
H2087 cells Lymph node Homo sapiens (Human) CVCL_1524
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
Cell Titer blue reagent assay
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Selumetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [25]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Selumetinib
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
Experiment for
Drug Resistance
MTD assay
Mechanism Description The missense mutation p.Q61L (c.182A>T) in gene NRAS cause the sensitivity of Selumetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Selumetinib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL 30 cells Skin Homo sapiens (Human) CVCL_0039
SkMEL 2 cells Skin Homo sapiens (Human) CVCL_0069
SH4 cells Skin Mus musculus (Mouse) CVCL_7702
MEXF-535 cells Skin Homo sapiens (Human) N.A.
MEXF-1792 cells Skin Homo sapiens (Human) N.A.
MEXF-1341 cells Skin Homo sapiens (Human) N.A.
M14 cells Hypodermis Homo sapiens (Human) CVCL_1395
GAK cells Lnguinal lymph node Homo sapiens (Human) CVCL_1225
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Female NIH nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Crystallization assay; X-ray data collection and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Enzymatic kinase assay
Disease Class: Melanoma [25]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Selumetinib
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Experimental Note Identified from the Human Clinical Data
Experiment for
Drug Resistance
MTD assay
Mechanism Description The missense mutation p.Q61R (c.182A>G) in gene NRAS cause the sensitivity of Selumetinib by unusual activation of pro-survival pathway
Disease Class: Melanoma [26]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Selumetinib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin .
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Selumetinib by aberration of the drug's therapeutic target
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Selumetinib
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL 30 cells Skin Homo sapiens (Human) CVCL_0039
SkMEL 2 cells Skin Homo sapiens (Human) CVCL_0069
SH4 cells Skin Mus musculus (Mouse) CVCL_7702
MEXF-535 cells Skin Homo sapiens (Human) N.A.
MEXF-1792 cells Skin Homo sapiens (Human) N.A.
MEXF-1341 cells Skin Homo sapiens (Human) N.A.
M14 cells Hypodermis Homo sapiens (Human) CVCL_1395
GAK cells Lnguinal lymph node Homo sapiens (Human) CVCL_1225
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Female NIH nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Crystallization assay; X-ray data collection and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Enzymatic kinase assay
Tanespimycin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [27]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Tanespimycin
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Human melanoma tissue .
Mechanism Description The missense mutation p.G13D (c.38G>A) in gene NRAS cause the sensitivity of Tanespimycin by unusual activation of pro-survival pathway
LY3009120
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [24]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug LY3009120
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL 30 cells Skin Homo sapiens (Human) CVCL_0039
SkMEL 2 cells Skin Homo sapiens (Human) CVCL_0069
SH4 cells Skin Mus musculus (Mouse) CVCL_7702
MEXF-535 cells Skin Homo sapiens (Human) N.A.
MEXF-1792 cells Skin Homo sapiens (Human) N.A.
MEXF-1341 cells Skin Homo sapiens (Human) N.A.
M14 cells Hypodermis Homo sapiens (Human) CVCL_1395
GAK cells Lnguinal lymph node Homo sapiens (Human) CVCL_1225
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Female NIH nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Crystallization assay; X-ray data collection and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Enzymatic kinase assay
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug LY3009120
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL 30 cells Skin Homo sapiens (Human) CVCL_0039
SkMEL 2 cells Skin Homo sapiens (Human) CVCL_0069
SH4 cells Skin Mus musculus (Mouse) CVCL_7702
MEXF-535 cells Skin Homo sapiens (Human) N.A.
MEXF-1792 cells Skin Homo sapiens (Human) N.A.
MEXF-1341 cells Skin Homo sapiens (Human) N.A.
M14 cells Hypodermis Homo sapiens (Human) CVCL_1395
GAK cells Lnguinal lymph node Homo sapiens (Human) CVCL_1225
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Female NIH nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Crystallization assay; X-ray data collection and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Enzymatic kinase assay
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug LY3009120
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
A375 cells Skin Homo sapiens (Human) CVCL_0132
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL 30 cells Skin Homo sapiens (Human) CVCL_0039
SkMEL 2 cells Skin Homo sapiens (Human) CVCL_0069
SH4 cells Skin Mus musculus (Mouse) CVCL_7702
MEXF-535 cells Skin Homo sapiens (Human) N.A.
MEXF-1792 cells Skin Homo sapiens (Human) N.A.
MEXF-1341 cells Skin Homo sapiens (Human) N.A.
M14 cells Hypodermis Homo sapiens (Human) CVCL_1395
GAK cells Lnguinal lymph node Homo sapiens (Human) CVCL_1225
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Female NIH nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Crystallization assay; X-ray data collection and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Enzymatic kinase assay
PLX4720
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [28]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug PLX4720
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
Preclinical Drug(s)
10 drug(s) in total
Click to Show/Hide the Full List of Drugs
Alpelisib/Binimetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Alveolar rhabdomyosarcoma [29]
Sensitive Disease Alveolar rhabdomyosarcoma [ICD-11: 2B55.0]
Sensitive Drug Alpelisib/Binimetinib
Molecule Alteration Missense mutation
p.Q61H (c.183A>T)
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation RAF/MEK/ERK signaling pathway Inhibition hsa04010
PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model RMS cells Soft tissue Homo sapiens (Human) CVCL_W527
In Vivo Model Chorioallantoic membrane Gallus gallus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay; FACS; crystal violet staining
Mechanism Description Coinhibition of NRAS or MEK plus PI3Kalpha triggers widespread apoptosis in NRAS-mutated RMS cells. Subtoxic concentrations of the MEK inhibitor MEK162 and the PI3Kalpha-specific inhibitor BYL719 synergized to trigger apoptosis in NRAS-mutated RMS cells in vitro and in vivo. NRAS- or HRAS-mutated cell lines were more vulnerable to MEK162/BYL719 cotreatment than RAS wild-type cell lines, and MEK162/BYL719 cotreatment was more effective to trigger apoptosis in NRAS-mutated than RAS wild-type RMS tumors in vivo.
Disease Class: Acute myeloid leukemia [30]
Sensitive Disease Acute myeloid leukemia [ICD-11: 2A60.0]
Sensitive Drug Alpelisib/Binimetinib
Molecule Alteration Missense mutation
p.G12D (c.35G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model NOMO1 cells Bone marrow Homo sapiens (Human) CVCL_1609
BaF3 cells Bone Mus musculus (Mouse) CVCL_0161
THP1 cell Peripheral blood Homo sapiens (Human) CVCL_0006
In Vivo Model mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
CCT196969
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [28]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug CCT196969
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
CCT241161
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [28]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug CCT241161
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
Cetuximab/Trametinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Colorectal cancer [31]
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Sensitive Drug Cetuximab/Trametinib
Molecule Alteration Missense mutation
p.G12V (c.35G>T)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
CCK-81 cells N.A. Homo sapiens (Human) CVCL_2873
Compound 3144
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: T-cell lymphoma [32]
Sensitive Disease T-cell lymphoma [ICD-11: 2A60.3]
Sensitive Drug Compound 3144
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation RAS signaling pathway Inhibition hsa04014
In Vitro Model MEF cells Bone marrow Homo sapiens (Human) CVCL_M515
In Vivo Model Athymic nude Nu/Nu mouse PDX model Mus musculus
Everolimus/Binimetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Neuroblastoma [33]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Sensitive Drug Everolimus/Binimetinib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/mTOR signaling pathway Inhibition hsa04151
In Vitro Model Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
NGP cells Lung Homo sapiens (Human) CVCL_2141
CHP-212 cells Brain Homo sapiens (Human) CVCL_1125
CHP-134 cells Adrenal gland Homo sapiens (Human) CVCL_1124
Experiment for
Molecule Alteration
Western blotting analysis; PCR
Experiment for
Drug Resistance
Promega assay; FACS assay
Mechanism Description Combination of mTOR and MEK inhibitors synergistically inhibit downstream signaling and cell growth of NRAS mutant cell lines.
MK2206
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Thyroid gland cancer [34]
Sensitive Disease Thyroid gland cancer [ICD-11: 2D10.0]
Sensitive Drug MK2206
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
C643 cells Thyroid gland Homo sapiens (Human) CVCL_5969
HTH7 cells Thyroid gland Homo sapiens (Human) CVCL_6289
Hth74 cells Thyroid gland Homo sapiens (Human) CVCL_6288
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The missense mutation p.Q61R (c.182A>G) in gene NRAS cause the sensitivity of MK2206 by unusual activation of pro-survival pathway
Pimasertib/Regorafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [35]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Pimasertib/Regorafenib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
H1975 cells Lung Homo sapiens (Human) CVCL_1511
A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
LOVO cells Colon Homo sapiens (Human) CVCL_0399
H1299 cells Lung Homo sapiens (Human) CVCL_0060
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
In Vivo Model Female balb/c athymic (nu+/nu+) mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Pimasertib + Regorafenib by unusual activation of pro-survival pathway
Disease Class: Lung adenocarcinoma [35]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Pimasertib/Regorafenib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
H1975 cells Lung Homo sapiens (Human) CVCL_1511
A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
LOVO cells Colon Homo sapiens (Human) CVCL_0399
H1299 cells Lung Homo sapiens (Human) CVCL_0060
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
In Vivo Model Female balb/c athymic (nu+/nu+) mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Pimasertib + Regorafenib by unusual activation of pro-survival pathway
RAF709
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [36]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug RAF709
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vivo Model Nude mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description Targeting CRAF could sensitize response to MEK inhibitors in KRAS-mutant tumors. In addition to targeting multiple signaling nodes within the MAPK pathway to maximize both level and duration of signaling inhibition, combining RAF709 with inhibitors targeting the upstream RTK activation such as EGFR, bypass mechanisms such as YAP1 or targeting prosurvival BCL2 family members, may further enhance efficacy and reduce the development of drug resistance.
Disease Class: Melanoma [36]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug RAF709
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vivo Model Nude mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description Targeting CRAF could sensitize response to MEK inhibitors in KRAS-mutant tumors. In addition to targeting multiple signaling nodes within the MAPK pathway to maximize both level and duration of signaling inhibition, combining RAF709 with inhibitors targeting the upstream RTK activation such as EGFR, bypass mechanisms such as YAP1 or targeting prosurvival BCL2 family members, may further enhance efficacy and reduce the development of drug resistance.
Disease Class: Melanoma [36]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug RAF709
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vivo Model Nude mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description Targeting CRAF could sensitize response to MEK inhibitors in KRAS-mutant tumors. In addition to targeting multiple signaling nodes within the MAPK pathway to maximize both level and duration of signaling inhibition, combining RAF709 with inhibitors targeting the upstream RTK activation such as EGFR, bypass mechanisms such as YAP1 or targeting prosurvival BCL2 family members, may further enhance efficacy and reduce the development of drug resistance.
Selumetinib/Dactolisib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Colorectal cancer [37]
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Sensitive Drug Selumetinib/Dactolisib
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Liver .
In Vivo Model Female nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunohistochemistry assay
Mechanism Description The missense mutation p.Q61K (c.181C>A) in gene NRAS cause the sensitivity of Selumetinib + Dactolisib by unusual activation of pro-survival pathway
Investigative Drug(s)
4 drug(s) in total
Click to Show/Hide the Full List of Drugs
Braf inhibitor
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [38]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Resistant Drug Braf inhibitor
Molecule Alteration Mutation
.
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Experiment for
Molecule Alteration
Next-generation sequencing assay
Mechanism Description Data suggest that the presence of mutated NRAS in the melanoma cell population in parallel with mutated BRAF cells results in resistance to BRAF inhibitors, most probably selecting NRAS-mutated cells in the advancing tumor.
ERK inhibitors
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Solid tumour/cancer [39]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug ERK inhibitors
Molecule Alteration Missense mutation
p.G12C (c.34G>T)
Experimental Note Revealed Based on the Cell Line Data
Experiment for
Molecule Alteration
DNA sequencing assay
Experiment for
Drug Resistance
Fluorescence-activated cell sorting assay
Mechanism Description The missense mutation p.G12C (c.34G>T) in gene NRAS cause the sensitivity of ERK inhibitors by unusual activation of pro-survival pathway
N-arachidonoyl dopamine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Solid tumour/cancer [40]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug N-arachidonoyl dopamine
Molecule Alteration Missense mutation
p.G12D (c.35G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
NIH3T3 cells Embryo Homo sapiens (Human) N.A.
WEHI-3 cells Peripheral blood Mus musculus (Mouse) CVCL_3622
Bosc23 cells Fetal kidney Homo sapiens (Human) CVCL_4401
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description N-Arachidonoyl Dopamine Inhibits NRAS Neoplastic Transformation by Suppressing Its Plasma Membrane Translocation.
PKI-587
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [41]
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Resistant Drug PKI-587
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Experimental Note Identified from the Human Clinical Data
Disease- and Tissue-specific Abundances of This Molecule
ICD Disease Classification 02
Click to Show/Hide the Resistance Disease of This Class
Brain cancer [ICD-11: 2A00]
Click to Show/Hide
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: 1.15E-106; Fold-change: 8.77E-01; Z-score: 1.64E+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 Glioma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 2.97E-01; Fold-change: 1.73E-01; Z-score: 1.11E+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 White matter
The Specified Disease Glioma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 6.93E-01; Fold-change: 1.25E-02; Z-score: 2.72E-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 Brainstem tissue
The Specified Disease Neuroectodermal tumor
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 8.85E-06; Fold-change: 1.63E+00; Z-score: 2.78E+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
Chronic myeloid leukemia [ICD-11: 2A20]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Whole blood
The Specified Disease Myelofibrosis
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 1.61E-01; Fold-change: 1.78E-01; Z-score: 5.30E-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
The Studied Tissue Whole blood
The Specified Disease Polycythemia vera
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 9.11E-01; Fold-change: 7.31E-02; Z-score: 2.22E-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
Acute myeloid leukemia [ICD-11: 2A60]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Bone marrow
The Specified Disease Acute myeloid leukemia
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 7.75E-30; Fold-change: 6.11E-01; Z-score: 1.46E+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
Multiple myeloma [ICD-11: 2A83]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Bone marrow
The Specified Disease Multiple myeloma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 3.29E-04; Fold-change: 5.38E-01; Z-score: 2.03E+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 Peripheral blood
The Specified Disease Multiple myeloma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 2.65E-01; Fold-change: -1.61E-01; Z-score: -4.70E-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
Gastric cancer [ICD-11: 2B72]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Gastric tissue
The Specified Disease Gastric cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 1.88E-01; Fold-change: 7.15E-01; Z-score: 6.55E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 1.81E-04; Fold-change: 4.36E-01; Z-score: 1.21E+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
Lung cancer [ICD-11: 2C25]
Click to Show/Hide
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: 4.97E-31; Fold-change: 4.05E-01; Z-score: 1.15E+00
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 1.00E-14; Fold-change: 3.26E-01; Z-score: 7.34E-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]
Click to Show/Hide
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: 7.02E-03; Fold-change: 3.94E-01; Z-score: 6.83E-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]
Click to Show/Hide
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: 2.25E-26; Fold-change: 3.50E-01; Z-score: 6.95E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 4.60E-04; Fold-change: 2.12E-01; Z-score: 2.99E-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
Thyroid cancer [ICD-11: 2D10]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Thyroid
The Specified Disease Thyroid cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 5.65E-01; Fold-change: 3.79E-03; Z-score: 1.19E-02
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 1.83E-03; Fold-change: 1.77E-01; Z-score: 4.55E-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
Tissue-specific Molecule Abundances in Healthy Individuals
Click to Show/Hide the Molecule Abundances
References
Ref 1 MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 studyLancet Oncol. 2013 Mar;14(3):249-56. doi: 10.1016/S1470-2045(13)70024-X. Epub 2013 Feb 13.
Ref 2 European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013 Aug 8;122(6):872-84. doi: 10.1182/blood-2013-05-501569. Epub 2013 Jun 26.
Ref 3 Dissecting Genomic Aberrations in Myeloproliferative Neoplasms by Multiplex-PCR and Next Generation Sequencing. PLoS One. 2015 Apr 20;10(4):e0123476. doi: 10.1371/journal.pone.0123476. eCollection 2015.
Ref 4 Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer. Nat Commun. 2016 Dec 8;7:13665. doi: 10.1038/ncomms13665.
Ref 5 MicroRNA-29a Functions as a Tumor Suppressor and Increases Cisplatin Sensitivity by Targeting NRAS in Lung Cancer. Technol Cancer Res Treat. 2018 Jan 1;17:1533033818758905. doi: 10.1177/1533033818758905.
Ref 6 MiR-26a enhances the sensitivity of gastric cancer cells to cisplatin by targeting NRAS and E2F2. Saudi J Gastroenterol. 2015 Sep-Oct;21(5):313-9. doi: 10.4103/1319-3767.166206.
Ref 7 The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma. Cancer Discov. 2014 Jan;4(1):94-109. doi: 10.1158/2159-8290.CD-13-0617. Epub 2013 Nov 21.
Ref 8 BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin Cancer Res. 2014 Apr 1;20(7):1965-77. doi: 10.1158/1078-0432.CCR-13-3122. Epub 2014 Jan 24.
Ref 9 Tumor heterogeneity and plasticity as elusive drivers for resistance to MAPK pathway inhibition in melanoma. Oncogene. 2015 Jun 4;34(23):2951-7. doi: 10.1038/onc.2014.249. Epub 2014 Aug 11.
Ref 10 Next generation sequencing of exceptional responders with BRAF-mutant melanoma: implications for sensitivity and resistance. BMC Cancer. 2015 Feb 18;15:61. doi: 10.1186/s12885-015-1029-z.
Ref 11 Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma. Nat Commun. 2014 Dec 2;5:5694. doi: 10.1038/ncomms6694.
Ref 12 Circulating-free DNA Mutation Associated with Response of Targeted Therapy in Human Epidermal Growth Factor Receptor 2-positive Metastatic Breast Cancer. Chin Med J (Engl). 2017 Mar 5;130(5):522-529. doi: 10.4103/0366-6999.200542.
Ref 13 MicroRNA-22 Suppresses Breast Cancer Cell Growth and Increases Paclitaxel Sensitivity by Targeting NRAS. Technol Cancer Res Treat. 2018 Jan 1;17:1533033818809997. doi: 10.1177/1533033818809997.
Ref 14 Pamidronate resistance and associated low ras levels in breast cancer cells: a role for combinatorial therapy .Ann Clin Lab Sci. 2004 Summer;34(3):263-70.
Ref 15 Heterogeneity of Acquired Resistance to Anti-EGFR Monoclonal Antibodies in Patients with Metastatic Colorectal Cancer. Clin Cancer Res. 2017 May 15;23(10):2414-2422. doi: 10.1158/1078-0432.CCR-16-1863. Epub 2016 Oct 25.
Ref 16 Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov. 2014 Nov;4(11):1269-80. doi: 10.1158/2159-8290.CD-14-0462. Epub 2014 Oct 7.
Ref 17 Circulating tumor DNA to monitor treatment response and detect acquired resistance in patients with metastatic melanoma. Oncotarget. 2015 Dec 8;6(39):42008-18. doi: 10.18632/oncotarget.5788.
Ref 18 Characteristics of lung cancers harboring NRAS mutationsClin Cancer Res. 2013 May 1;19(9):2584-91. doi: 10.1158/1078-0432.CCR-12-3173. Epub 2013 Mar 20.
Ref 19 Spatially divergent clonal evolution in multiple myeloma: overcoming resistance to BRAF inhibition. Blood. 2016 Apr 28;127(17):2155-7. doi: 10.1182/blood-2015-12-686782. Epub 2016 Feb 16.
Ref 20 Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010 Dec 16;468(7326):973-7. doi: 10.1038/nature09626. Epub 2010 Nov 24.
Ref 21 COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature. 2010 Dec 16;468(7326):968-72. doi: 10.1038/nature09627. Epub 2010 Nov 24.
Ref 22 Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013 Aug;10(8):472-84. doi: 10.1038/nrclinonc.2013.110. Epub 2013 Jul 9.
Ref 23 The discovery of vemurafenib for the treatment of BRAF-mutated metastatic melanoma .Expert Opin Drug Discov. 2016 Sep;11(9):907-16. doi: 10.1080/17460441.2016.1201057. Epub 2016 Jun 23. 10.1080/17460441.2016.1201057
Ref 24 Inhibition of RAF Isoforms and Active Dimers by LY3009120 Leads to Anti-tumor Activities in RAS or BRAF Mutant CancersCancer Cell. 2015 Sep 14;28(3):384-98. doi: 10.1016/j.ccell.2015.08.002. Epub 2015 Sep 3.
Ref 25 Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancersJ Clin Oncol. 2008 May 1;26(13):2139-46. doi: 10.1200/JCO.2007.14.4956. Epub 2008 Apr 7.
Ref 26 Reversing melanoma cross-resistance to BRAF and MEK inhibitors by co-targeting the AKT/mTOR pathwayPLoS One. 2011;6(12):e28973. doi: 10.1371/journal.pone.0028973. Epub 2011 Dec 14.
Ref 27 BRAF and NRAS mutations in melanoma: potential relationships to clinical response to HSP90 inhibitorsMol Cancer Ther. 2008 Apr;7(4):737-9. doi: 10.1158/1535-7163.MCT-08-0145. Epub 2008 Mar 28.
Ref 28 Paradox-breaking RAF inhibitors that also target SRC are effective in drug-resistant BRAF mutant melanomaCancer Cell. 2015 Jan 12;27(1):85-96. doi: 10.1016/j.ccell.2014.11.006. Epub 2014 Dec 11.
Ref 29 NRAS-Mutated Rhabdomyosarcoma Cells Are Vulnerable to Mitochondrial Apoptosis Induced by Coinhibition of MEK and PI3KAlphaCancer Res. 2018 Apr 15;78(8):2000-2013. doi: 10.1158/0008-5472.CAN-17-1737. Epub 2018 Feb 6.
Ref 30 Hematopoiesis and RAS-driven myeloid leukemia differentially require PI3K isoform p110AlphaJ Clin Invest. 2014 Apr;124(4):1794-809. doi: 10.1172/JCI69927. Epub 2014 Feb 24.
Ref 31 Genome-wide chemical mutagenesis screens allow unbiased saturation of the cancer genome and identification of drug resistance mutationsGenome Res. 2017 Apr;27(4):613-625. doi: 10.1101/gr.213546.116. Epub 2017 Feb 8.
Ref 32 Multivalent Small-Molecule Pan-RAS InhibitorsCell. 2017 Feb 23;168(5):878-889.e29. doi: 10.1016/j.cell.2017.02.006.
Ref 33 Targeting the mTOR Complex by Everolimus in NRAS Mutant NeuroblastomaPLoS One. 2016 Jan 28;11(1):e0147682. doi: 10.1371/journal.pone.0147682. eCollection 2016.
Ref 34 The Akt-specific inhibitor MK2206 selectively inhibits thyroid cancer cells harboring mutations that can activate the PI3K/Akt pathwayJ Clin Endocrinol Metab. 2011 Apr;96(4):E577-85. doi: 10.1210/jc.2010-2644. Epub 2011 Feb 2.
Ref 35 Antitumor activity of pimasertib, a selective MEK 1/2 inhibitor, in combination with PI3K/mTOR inhibitors or with multi-targeted kinase inhibitors in pimasertib-resistant human lung and colorectal cancer cellsInt J Cancer. 2013 Nov;133(9):2089-101. doi: 10.1002/ijc.28236. Epub 2013 May 29.
Ref 36 Antitumor Properties of RAF709, a Highly Selective and Potent Inhibitor of RAF Kinase Dimers, in Tumors Driven by Mutant RAS or BRAFCancer Res. 2018 Mar 15;78(6):1537-1548. doi: 10.1158/0008-5472.CAN-17-2033. Epub 2018 Jan 17.
Ref 37 Inhibition of MEK and PI3K/mTOR suppresses tumor growth but does not cause tumor regression in patient-derived xenografts of RAS-mutant colorectal carcinomasClin Cancer Res. 2012 May 1;18(9):2515-25. doi: 10.1158/1078-0432.CCR-11-2683. Epub 2012 Mar 5.
Ref 38 Genetic progression of malignant melanoma. Cancer Metastasis Rev. 2016 Mar;35(1):93-107. doi: 10.1007/s10555-016-9613-5.
Ref 39 Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitorsCancer Discov. 2013 Jul;3(7):742-50. doi: 10.1158/2159-8290.CD-13-0070. Epub 2013 Apr 24.
Ref 40 N-Arachidonoyl Dopamine Inhibits NRAS Neoplastic Transformation by Suppressing Its Plasma Membrane TranslocationMol Cancer Ther. 2017 Jan;16(1):57-67. doi: 10.1158/1535-7163.MCT-16-0419. Epub 2016 Oct 19.
Ref 41 Mutations of the BRAF gene in human cancerNature. 2002 Jun 27;417(6892):949-54. doi: 10.1038/nature00766. Epub 2002 Jun 9.

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